You are on page 1of 952

DEMENTIA

F I F T H E D I T I O N
DEMENTIA
F I F T H E D I T I O N

Edited by
David Ames BA, MD, BS, FRCPsych, FRANZCP
Emeritus Professor
University of Melbourne Department of Psychiatry and
National Ageing Research Institute,
Parkville, Victoria, Australia

John O’Brien MA DM, FRCPsych, FMedSci


Foundation Professor of Old Age Psychiatry
Department of Psychiatry
University of Cambridge, Cambridge, UK

Alistair Burns CBE, MD, FRCP, FRCPsych


Professor of Old Age Psychiatry
University of Manchester, Manchester, UK
CRC Press
Taylor & Francis Group
6000 Broken Sound Parkway NW, Suite 300
Boca Raton, FL 33487-2742
© 2017 by Taylor & Francis Group, LLC
CRC Press is an imprint of Taylor & Francis Group, an Informa business

No claim to original U.S. Government works

Printed on acid-free paper


Version Date: 20161017

International Standard Book Number-13: 978-1-4987-0310-9 (Pack - Book and Ebook)

This book contains information obtained from authentic and highly regarded sources. Reasonable efforts have been made
to publish reliable data and information, but the author and publisher cannot assume responsibility for the validity of all
materials or the consequences of their use. The authors and publishers have attempted to trace the copyright holders of
all material reproduced in this publication and apologize to copyright holders if permission to publish in this form has
not been obtained. If any copyright material has not been acknowledged please write and let us know so we may rectify
in any future reprint.

Except as permitted under U.S. Copyright Law, no part of this book may be reprinted, reproduced, transmitted, or ­utilized
in any form by any electronic, mechanical, or other means, now known or hereafter invented, including photocopying,
microfilming, and recording, or in any information storage or retrieval system, without written permission from the
publishers.

For permission to photocopy or use material electronically from this work, please access www.copyright.com (http://
www.copyright.com/) or contact the Copyright Clearance Center, Inc. (CCC), 222 Rosewood Drive, Danvers, MA 01923,
978-750-8400. CCC is a not-for-profit organization that provides licenses and registration for a variety of users. For
­organizations that have been granted a photocopy license by the CCC, a separate system of payment has been arranged.

Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are used only for
identification and explanation without intent to infringe.
Visit the Taylor & Francis Web site at
http://www.taylorandfrancis.com
and the CRC Press Web site at
http://www.crcpress.com
Contents

Foreword ix
Preface x
Acknowledgements xii
Contributors xiii

PaRT 1 DEMENTIA: General aspects 1

1 Dementia: Historical overview 3


German Berrios
2 The lived experience of dementia 19
Shirley Nurock
3 Prevalence and incidence of dementia 24
Daniel W. O’Connor
4 What is dementia, and how do you assess it? Definitions, diagnostic criteria and assessment 33
Joseph P.M. Kane and Alan Thomas
5 Screening and assessment instruments for the detection and measurement of cognitive impairment 44
Leon Flicker
6 Neuropsychological assessment of dementia 52
Greg Savage
7 Neuropsychiatric aspects of dementia 59
Nilika Perera, Mehran Javeed, Constantine G. Lyketsos and Iracema Leroi
8 Measurement of behaviour disturbance, non-cognitive symptoms and quality of life 67
Ajit Shah
9 Cross-cultural issues in the assessment of cognitive impairment 79
Ajit Shah
10 Structural brain imaging 92
Robert Barber and John T. O’Brien
11 Functional brain imaging and connectivity in dementia 107
Klaus P. Ebmeier, Nicola Filippini, Clare E. Mackay, Sana Suri and Vyara Valkanova
12 Molecular brain imaging in dementia 119
Victor L. Villemagne and Christopher C. Rowe
13 Services for people with dementia 135
Sube Banerjee
14 Family carers of people with dementia 142
Katrin Seeher and Henry Brodaty
15 Dementia care in the community: Challenges for primary health and social care 161
Steve Iliffe, Jill Manthorpe and Vari Drennan
16 Managing people with dementia in the general hospital 172
Andrew Teodorczuk, Rowan Harwood and Elizabeth Sampson
17 The role of nursing in the management of dementia 183
Maree Mastwyk and Beverley Williams
18 Social work and care/case management in dementia 192
David Challis, Jane Hughes and Caroline Sutcliffe

v
vi Contents

19 Occupational therapy in dementia care 199


Alissa Westphal
20 Speech and language therapy in dementia assessment and management 208
Bronwyn Moorhouse and Caroline A. Fisher
21 Role of the physiotherapist in the management of dementia 220
Sue Lord and Lynn Rochester
22 Therapeutic effects of music in persons with dementia 229
Linda A. Gerdner and Ruth Remington
23 Psychological, behavioural and psychosocial interventions for neuropsychiatric symptoms in dementia:
What works, what does not and what needs more evidence? 235
Gill Livingston and Claudia Cooper
24 Treating problem behaviours in dementia by understanding their biological, social and psychological causes 244
Ian James and Louisa Jackman
25 Treatments for behavioural and psychological symptoms in Alzheimer’s disease and other dementias 257
Anna Burke, William J. Burke and Pierre N. Tariot
26 Psychological approaches for the practical management of cognitive impairment in dementia 279
Anne Unkenstein
27 Sexuality and dementia 285
Joe Stratford
28 Residential care for people with dementia 292
David Conn, John Snowdon and Nitin Purandare
29 Design and dementia 304
June Andrews
30 Legal issues and dementia 308
Hugh Series and Robin Jacoby
31 Driving and dementia 314
Aoife Fallon and Desmond O’Neill
32 Quality of life in dementia: Conceptual and practical issues 325
Betty S. Black and Peter V. Rabins
33 Ethical issues 339
Julian C. Hughes and Daniel Strech
34 End-of-life and palliative care in dementia 344
Julian C. Hughes and Jenny T. van der Steen
35 Advanced dementia and care at the end of life 349
Kirsten Moore and Elizabeth Sampson
36 Alzheimer’s associations and societies 359
Henry Brodaty, Nicole Pesa and Glenn Rees
37 Health economic aspects of dementia 371
Anders A. Wimo, Linus Jönsson and Bengt Winblad
38 Global challenge of dementia: What can be done? 388
Cleusa P. Ferri, Maëlenn Guerchet and Martin Prince
39 The international challenge of dementia 402
Karim Saad
40 The pharmaceutical industry and dementia: How can clinicians, researchers and industry work together for the
betterment of people with dementia and their families? Continuous vigilance and transparency is the answer 411
Martin M. Bednar and Leon Flicker

PaRT 2 MILD COGNITIVE IMPAIRMENT 418

41 Mild cognitive impairment (MCI): A historical perspective 419


Karen Ritchie and Sylvaine Artero
42 Clinical characteristics of mild cognitive impairment 426
Yonas E. Geda, Janina Krell-Roesch, Anna Pink, Kathleen A. Spangehl and Ronald C. Petersen
43 Managing the patient with mild cognitive impairment 439
Nicola T. Lautenschlager and Alexander F. Kurz
Contents vii

PaRT 3 ALZHEIMER’S DISEASE: Clinical Aspects 446

44 Risk factors for Alzheimer’s disease 447


Patricia A. Boyle and Robert S. Wilson
45 The natural history of Alzheimer’s disease 453
Jody Corey-Bloom and Michael S. Rafii
46 The neuropathology of Alzheimer’s disease 470
Colin L. Masters
47 Neurochemistry of Alzheimer’s disease 486
Paul T. Francis
48 The central role of Aβ amyloid and tau in the pathogenesis of Alzheimer’s disease 492
Craig W. Ritchie and Colin L. Masters
49 Inflammation in Alzheimer’s disease 508
Clive Holmes
50 Genetics of Alzheimer’s disease 519
Margie Smith
51 Blood and cerebrospinal fluid biomarkers for Alzheimer’s disease 528
Simone Lista, Henrik Zetterberg, Sid E. O’Bryant, Kaj Blennow and Harald Hampel
52 Established treatments for Alzheimer’s disease 539
Alexander Kurz and Nicola T. Lautenschlager
53 Drug treatments in development for Alzheimer’s disease 554
Paul Yates and Michael Woodward
54 Vitamins and complementary therapies used to treat Alzheimer’s disease 587
Dennis Chang, Genevieve Z. Steiner, Dilip Ghosh and Alan Bensoussan
55 Prevention of Alzheimer’s disease and Alzheimer’s dementia 598
Tom C. Russ, Karen Ritchie and Craig W. Ritchie
56 Trial designs 613
Serge Gauthier

PaRT 4 THE OVERLAP AND INTERACTION BETWEEN ALZHEIMER’S DISEASE AND CEREBROVASCULAR
DISEASE 618

57 Vascular factors and Alzheimer’s disease 619


Robert Stewart

PaRT 5 CEREBROVASCULAR DISEASE AND COGNITIVE IMPAIRMENT 629

58 What is vascular cognitive impairment? 630


Timo Erkinjuntti, Leonardo Pantoni and Susanna Melkas
59 The neuropathology of vascular dementia 643
Raj N. Kalaria
60 Therapeutic strategies for vascular cognitive disorders 660
Gustavo C. Román

PaRT 6 DEMENTIA WITH LEWY BODIES AND PARKINSON’S DISEASE 702

61 Dementia with Lewy bodies: A clinical overview 703


Ian G. McKeith
62 Pathology of dementia with Lewy bodies 714
Glenda Halliday, Simon Lewis and Paul Ince
63 The treatment of dementia with Lewy bodies 730
John-Paul Taylor
64 Cognitive impairment and dementia in Parkinson’s disease 740
Dag Aarsland, Kolbjørn Brønnick, Milica G. Kramberger and Joana B. Pereira
viii Contents

PaRT 7 FOCAL DEMENTIAS AND OTHER NEUROPSYCHIATRIC SYNDROMES INVOLVING COGNITIVE DECLINE 748

65 Frontotemporal dementia 749


Peter J. Nestor
66 Pick’s disease: Its relationship to progressive aphasia, semantic dementia and frontotemporal dementia 759
John Hodges
67 The genetics and molecular pathology of frontotemporal lobar degeneration 771
David M.A. Mann
68 Semantic dementia 783
Julie S. Snowden and Matthew Jones
69 Primary progressive aphasia and posterior cortical atrophy 795
Jonathan D. Rohrer, Sebastian J. Crutch and Jason D. Warren
70 The cerebellum and cognitive impairment 812
Elsdon Storey and Evelyn A. Lindsay
71 Delirium: A clinical overview 823
Andrew Teodorczuk and Daniel Davis
72 Depression with cognitive impairment 832
Sarah Shizuko Morimoto, Genevieve S. Yuen, Stephen Beres and George S. Alexopoulos
73 Schizophrenia, cognitive impairment and dementia 846
Flavie Waters, Andrew Ford and Osvaldo P. Almeida
74 Dementia in intellectual disabilities 852
Jennifer Torr
75 Alcohol-related dementia and Wernicke–Korsakoff syndrome 857
Stephen C. Bowden and Simon J. Scalzo
76 Huntington’s disease 868
Phyllis Chua, Samantha Loi and Edmond Chiu
77 Uncommon dementias (including the prion diseases) 885
Matthew Jones and David Neary
Index 898
Foreword

The modern study of dementia began 50 years ago with the many techniques and approaches that were not dreamed of
pioneering work of Martin Roth and colleagues. In the mid- 50 years ago. Computerized imaging has been available for
1960s, their ground-breaking epidemiologic study demon- 40 years, but the recent development of positron emission
strated both the high prevalence of dementia in the elderly tomography (PET) imaging ligands specific for the molecu-
and its association with ageing. Several years later they dem- lar abnormalities characteristic of several dementias is likely
onstrated that the neuropathology first described by Alois to dramatically change the evaluation of people presenting
Alzheimer in 1907 in a woman who died from dementia at the earliest symptoms of cognitive disorder, to lead to the
the age of 56 is the commonest neuropathological abnor- identification of people at high risk of developing a dementia
mality seen in dementia. syndrome in 10–20 years before they become symptomatic
This 5th edition of Dementia demonstrates how much and, possibly, to reshape the assessment of therapeutics. The
knowledge has been gained in the past half-century since ability to identify genetic variants inexpensively, accurately
these studies were conducted. Epidemiological studies and quickly is already helping to identify multiple causes
throughout the world have identified many risk factors for of what had appeared to be single dementia syndromes and
dementia and Alzheimer’s disease and these are beginning might lead to revolutionary treatments that target the cause
to translate into prospective intervention studies. Advances of dementia in specific individuals.
in molecular biology and genetic medicine have identified Finally, this edition of Dementia highlights the recogni-
specific protein abnormalities that underlie many of the tion that extensive overlap exists between disorders that tra-
neurodegenerative causes of dementia, identified genes act- ditionally have been considered conceptually distinct. The
ing in classic Mendelian patterns in some dementias and concept that ‘functional’ disorders are categorically distinct
via non-Mendelian mechanisms in most, identified prions from the disorders of cognition with identifiable neuropa-
as the causative ‘agent’ of Creutzfeldt–Jacob disease (CJD) thology, which is now passé as cognitive impairment and
and variant CJD (vCJD); they also raised the possibility that has been identified as a central feature of schizophrenia
prion-like mechanisms explain the ‘spread’ of neurodegen- and impaired cognitive performance, has been identified as
eration in many diseases. present in individuals of all ages with major depression and
As extraordinary as these biological discoveries are, this bipolar disorder. Even the dichotomous distinction between
edition of Dementia also highlights the equally impres- Alzheimer’s dementia and vascular dementia (VaD), so cen-
sive advances that have been made in treatment. Except for tral to Martin Roth’s pioneering work, is coming under
HIV-AIDS dementia, no dramatic cures have been discov- question as the co-occurrence of these two cases of demen-
ered, but an extensive research literature demonstrates the tia is now being shown to result from shared neurobiology.
effectiveness and benefits of environmental and behavioural In summary, this 5th edition of Dementia stands as a
interventions that target the symptoms of dementia, the marker of the tremendous progress that has been made in
efficacy of interventions that target the emotional distress understanding the pathophysiology of the various demen-
experienced by many family members and care providers, tias and of the advances in symptomatic treatment. Just as
the importance of organizations that provide education importantly, it provides hope that these gains in knowledge
and support and the limited but necessary role of pharma- will lead to preventative strategies that abolish or greatly
cotherapeutic treatment of Behavioural and Psychological lessen the morbidity associated with these devastating
Symptoms of Dementia (BPSD). disorders.
In addition to highlighting these extraordinary advances,
this 5th edition of Dementia reviews the development of Peter V. Rabins MD, MPH

ix
Preface

Dementia is one of the major challenges to face society psychiatric disorders such as depression and schizophre-
in the twenty-first century, numerically dwarfing many nia, in which both subtle brain changes and cognitive
other disorders which have caught the public’s imagina- impairments occur, making the traditional organic versus
tion. There are, and will continue to be, many texts on functional distinction look blurred.
dementia. Our continued goal with this fifth edition is to Overlap between chapters is a difficult issue, and
encompass, in a single comprehensive volume, all aspects although we have exercised the editorial Tippex to the best
of all types of dementia. This is a highly ambitious task, of our ability, some duplication remains. However, much
and however hard we have tried, we could obviously not of this is intentional, and some chapters would have been
succeed completely, so someone, somewhere will rightly denuded unnecessarily and could not have stood alone.
complain that we have omitted something important. In Areas where we have unashamedly fostered such overlap
addition, and somewhat reassuringly in terms of progress, include Chapters 41 through 43 on mild cognitive impair-
this is a field that continues to expand rapidly. What we ment, Chapters 61 through 64 on the different ends of the
have tried to do, therefore, is to concentrate on a solid Lewy body dementias spectrum, and Chapters 65, 66 and
body of information which, although requiring some 68 covering fronto-temporal dementia and its subtypes,
updating in the future, is likely to remain the essential including semantic dementia. The general structure of this
core knowledge in the field for many years to come. book has been well honed over the four previous editions,
The book is divided into seven parts to reflect the and we have stuck largely to what seemed a popular and
complex nature of the topic. Part 1 deals with gen- successful formula. However, we have ensured that nearly
eral aspects of dementia, including its history, diag- all chapters have been updated; some established topics
nosis and assessment, and the use of investigations, have new authors taking an entirely fresh view, and we have
multi-­d isciplinary assessment and all aspects of its man- included new chapters on several key areas such as services,
agement. Part 2 focuses on mild cognitive impairment, managing co-morbidities, terminal care, the international
an increasingly important entity but one whose concep- challenge of dementia, neuroinflammation in Alzheimer’s
tual basis is changing as we discover more about early or disease and delirium.
prodromal pre-dementia states. Part 3 concentrates on We anticipate that target readership will be broad,
Alzheimer’s disease and includes discussions about risk reflecting the breadth of the book. It was never designed as
factors, underlying neurobiology, biomarkers and both a book to be read from cover to cover, and though we doubt
pharmacological and non-pharmacological management it will be, if it were it would probably be one of the quickest
as well as prospects for prevention. The well recognized ways of getting entirely up to speed with a complex and var-
but still poorly understood overlap between vascular and ied field. However, it is designed as a reference guide, to be
degenerative pathologies and the interactions between dipped into as and when needed. We anticipate that at least
the two are covered in Part 4, whilst Part 5 deals with some sections will be relevant and of interest to just about
vascular cognitive impairment and vascular dementia. all health and social care professionals who have contact
Part 6 contains chapters on Lewy body dementias, both with or an interest in people with dementia, including med-
dementia with Lewy bodies and dementia that occurs in ical specialists such as neurologists, psychiatrists and physi-
established Parkinson’s disease, while Part 7 discusses cians, nurses and allied health professionals, and trainees
other dementias, such as fronto-temporal dementias and within these groups. We hope that others will also find the
less common causes of dementia like Creutzfeld–Jakob book of value. For example, pre-clinical scientists who work
disease (CJD), progressive supranuclear palsy (PSP) and in this area will be able to learn more about the clinical rel-
multi system atrophy (MSA). This final part also covers evance and underlying biology of those with ­disorders they
the borderland between the dementias and functional are studying.

x
Preface xi

In choosing our authors, we have sought to achieve a renowned experts and those whom we consider to be the
balance between retaining sufficient authors from the pre- rising stars of the future.
vious editions to provide some continuity and including
some new contributors. We have also sought an authorship David Ames, Melbourne
to reflect a mixture of those who are already internationally Alistair Burns, Manchester
John T. O’Brien, Cambridge
Acknowledgements

We are grateful to our publishers for commissioning a fifth Chapter 42


edition of this book. The award of the best book of the year Taryn Silber, Ron Petersen and Yonas E. Geda thank
in the area of psychiatry for the fourth edition of this book Angelina Luti for her diligence in assembling the endnote
by the British Medical Association in 2011 may have had a library on biomarkers and proofing the manuscript. The
lot to do with this recommission. As usual, some chapters authors would also like to thank Ezra Geda for proofing
have been culled, some introduced, and virtually all have the manuscript.
been revised. We thank our patient and dedicated authors, Chapter 46
our secretaries and our publishing team for helping us put This chapter was originally written by Peter Lantos and
together what we hope is a clear account of everything useful Nigel Cairns and appeared as Chapter 37 in the second
known about dementia in 2016. If the sixth edition eventu- edition of Dementia. It was updated by Colin Masters in all
ates, it may be edited by a different team, as one of us has now subsequent editions.
retired from academic life and the other two are not getting
any younger, but we devoutly hope that this established text Chapter 51
will continue to be found useful by researchers and those Harold Hampel and Simone Lista were supported by
working with people with dementia for a long time to come. the AXA Research Fund, the Foundation Université
Pierre et Marie Curie and the ‘Fondation pour la
David Ames, Alistair Burns, John T. O’Brien Recherche sur Alzheimer’, Paris, France. The research
Melbourne, Manchester, Newcastle-upon-Tyne leading to these results has received funding from
December 2015 the program ‘Investissements d’avenir’ ANR-10-IAIHU-06.

Chapter 59
ADDITIONAL ACKNOWLEDGMENTS Raj Kalaria’s work has been supported by the Medical
Research Council (United Kingdom), Alzheimer’s
Chapter 24 Research, United Kingdom and the Dunhill Medical Trust,
Ian James thanks Angela Hope, who provided both United Kingdom.
comments and assisted with the preparation of this
chapter.
Chapter 38
Cleusa P. Ferri, Maëlenn Guerchet and Martin Prince
acknowledge, in memoriam, the valuable contribution
made to the previous version of this chapter (4th edition)
by Jim Jackson.

xii
Contributors

Dag Aarsland Newcastle University


Stavanger University Hospital Newcastle upon Tyne, United Kingdom
Stavanger, Norway
Martin M. Bednar
and Neuroscience and Pain Research Unit Pfizer Global
Division of Neurogeriatrics Research and Development
Department of Neurobiology Cambridge, Massachusetts
Care Sciences and Society, Karolinska Institute Alan Bensoussan
Stockholm, Sweden National Institute of Complementary Medicine
George S. Alexopoulos Western Sydney University
Weill Cornell Medical College Campbelltown, Australia
White Plains, New York Stephen Beres
Osvaldo P. Almeida Weill Cornell Medical College
Western Australian Centre for Health and Ageing White Plains, New York
School of Psychiatry and Clinical Neurosciences, Centre German Berrios
for Medical Research Emeritus Chair of the Epistemology of Psychiatry
University of Western Australia University of Cambridge
and Cambridge, United Kingdom
Department of Psychiatry
Royal Perth Hospital Betty S . Black
Perth, Australia Department of Psychiatry and Behavioral Sciences
Johns Hopkins Medical Institutions
June Andrews Baltimore, Maryland
Professor Emeritus
University of Stirling Kaj Blennow
Bridge of Allan, Scotland Clinical Neurochemistry Laboratory
Department of Psychiatry and Neurochemistry
Sylvaine Artero Institute of Neuroscience and Physiology
Institut National de la Santé et de la Recherche Médicale The Sahlgrenska Academy at the University of
(INSERM) Gothenburg
Neuropsychiatry, Epidemiological and Clinical Mölndal, Sweden
Research
and
Hôpital La Colombière
Montpellier, France The Torsten Söderberg Professorship in Medicine
Royal Swedish Academy of Sciences
Sube Banerjee
Stockholm, Sweden
Centre for Dementia Studies
Brighton and Sussex Medical School Stephen C. Bowden
University of Sussex Melbourne School of Psychological Sciences
Brighton, United Kingdom University of Melbourne
Melbourne, Australia
Robert Barber
Old Age Psychiatry Patricia A. Boyle
Institute of Neuroscience Department of Behavioral Science

xiii
xiv Contributors

Rush Alzheimer’s Disease Center Claudia Cooper


Rush University Medical Center Division of Psychiatry
Chicago, Illinois University College London
London, United Kingdom
Henry Brodaty
Dementia Collaborative Research Centre Jody Corey-Bloom
School of Psychiatry Department of Neurosciences
University of New South Wales University of California
Sydney, Australia San Diego, California

Kolbjørn Brønnick Sebastian J. Crutch


Psychiatric Division Dementia Research Centre
Stavanger University Hospital University College London
and London, United Kingdom
Network for Medical Sciences
Daniel Davis
University of Stavanger
Department of Public Health and Primary Care
Stavanger, Norway
University of Cambridge
Anna Burke Cambridge, United Kingdom
Banner Alzheimer’s Institute
Vari Drennan
University of Arizona College of Medicine
Health Services Research
Phoenix, Arizona
St. George’s University of London
William J. Burke and
Banner Alzheimer’s Institute Kingston University
University of Arizona College of Medicine London, United Kingdom
Phoenix, Arizona
Klaus P. Ebmeier
David Challis Department of Psychiatry
Personal Social Services Research Unit University of Oxford
University of Manchester and
Manchester, United Kingdom The Warneford Hospital
Oxford, United Kingdom
Dennis Chang
National Institute of Complementary Yonas E. Geda
Medicine Translational Neuroscience and Aging
Western Sydney University Program
Campbelltown, Australia Collaborative Research Building
Mayo Clinic
Edmond Chiu
Scottsdale, Arizona
Academic Unit for Psychiatry of Old Age
University of Melbourne Timo Erkinjuntti
Melbourne, Australia Clinical Neurosciences, Neurology
University of Helsinki
Phyllis Chua
and
Department of Psychiatry
Helsinki University Hospital
School of Clinical Sciences
Helsinki, Finland
Monash University
Clayton, Australia Aoife Fallon
Centre for Ageing, Neuroscience and the
and
Humanities
Statewide Progressive Neurological Disease Trinity College
Service Dublin, Ireland
Calvary Health Care Bethlehem
Cleusa P. Ferri
Caulfield South, Australia
Department of Psychobiology
David Conn Universidade Federal de São Paulo
Centre for Education and
Baycrest Health Sciences Hospital Alemao Oswaldo Cruz
University of Toronto Institute of Education and Health Sciences (IECS)
Toronto, Canada São Paulo, Brazil
Contributors xv

Nicola Filippini Neuroscience Research Australia


Department of Psychiatry Sydney, Australia
University of Oxford
Harald Hampel
and
AXA Research Fund and UPMC Chair
The Warneford Hospital
and
Oxford, United Kingdom
Sorbonne Universités
Caroline A. Fisher Université Pierre et Marie Curie
The Melbourne Clinic and
The Royal Melbourne Hospital Institut de la Mémoire et de la Maladie d’Alzheimer
Melbourne, Australia (IM2A)
Leon Flicker Institut du Cerveau et de la Moelle Épinière (ICM)
Western Australian Centre for Health and Ageing Département de Neurologie
Centre for Medical Research, School of Medicine and Hôpital de la Pitié-Salpêtrière
Pharmacology Paris, France
University of Western Australia Rowan Harwood
Perth, Australia Nottingham University Hospitals NHS Trust
Andrew Ford and
Western Australian Centre for Health and Ageing University of Nottingham
School of Psychiatry and Clinical Neurosciences and Nottingham, United Kingdom
Centre for Medical Research John Hodges
University of Western Australia NeuRA and UNSW
and Randwick, Australia
Department of Psychiatry
Royal Perth Hospital Clive Holmes
Perth, Australia Clinical and Experimental Science
University of Southampton
Paul T. Francis Southampton, United Kingdom
Wolfson Centre for Age-Related Diseases
King’s College London, Guy’s Campus Jane Hughes
London, United Kingdom Personal Social Services Research Unit
University of Manchester
Serge Gauthier Manchester, United Kingdom
Alzheimer’s Disease Research Unit
McGill Centre for Studies in Aging Julian C. Hughes
McGill University Northumbria Healthcare NHS Foundation Trust
Montréal, Canada and
PEALS (Policy, Ethics and Life Sciences) Research Centre
Linda A. Gerdner Newcastle University
Stanford Geriatric Education Center Newcastle upon Tyne, United Kingdom
Center for Education in Family and Community Medicine
Stanford University School of Medicine Steve Iliffe
Stanford, California Primary Care for Older People
University College London
Dilip Ghosh London, United Kingdom
Soho Flordis International (SFI)
St Leonards, Australia Paul Ince
Department of Neuroscience
Maëlenn Guerchet Royal Hallamshire Hospital
King’s College London Sheffield, United Kingdom
and
Centre for Global Mental Health Louisa Jackman
Health Services and Population Research Department of Clinical Psychology
Institute of Psychiatry, Psychology and Neuroscience Newcastle University
London, United Kingdom Newcastle upon Tyne, United Kingdom

Glenda Halliday Robin Jacoby


University of New South Wales Department of Psychiatry
and University of Oxford
xvi Contributors

The Warneford Hospital Janina Krell-Roesch


Oxford, United Kingdom Translational Neuroscience and Aging Program
Collaborative Research Building
Ian James
Mayo Clinic
Older People’s Psychology and Challenging Behaviour
Scottsdale, Arizona
Teams
Northumberland, Tyne and Wear NHS Trust Alexander F. Kurz
and Department of Psychiatry and Psychotherapy
Campus for Ageing and Vitality Klinikum rechts der Isar
Newcastle upon Tyne, United Kingdom Technische Universität München
Munich, Germany
Mehran Javeed
Department of Old Age Psychiatry Nicola T. Lautenschlager
North Manchester General Hospital Academic Unit for Psychiatry of Old Age, Department of
and Psychiatry
Manchester Mental Health and Social Care Trust University of Melbourne
Manchester, United Kingdom NorthWestern Mental Health, Melbourne Health
Parkville, Australia
Matthew Jones
Cerebral Function Unit Iracema Leroi
Salford Royal NHS Foundation Trust Department of Psychiatry/Honorary Consultant
Salford, United Kingdom Institute of Brain, Behaviour and Mental Health
and and
Institute of Brain Behaviour and Mental Health Manchester Academic Health Sciences Centre
University of Manchester University of Manchester
Manchester, United Kingdom Manchester, United Kingdom

Linus Jönsson Simon Lewis


Division of Neurogeriatrics Brain & Mind Centre
NCS, Department of Neurobiology Care Sciences and Sydney Medical School
Society Sydney, Australia
Karolinska Institutet Evelyn A. Lindsay
Stockholm, Sweden Department of Medicine (Neuroscience)
and Monash University
H. Lundbeck A/S Melbourne, Australia
Valby, Denmark
Simone Lista
Raj N. Kalaria AXA Research Fund and UPMC Chair
Institute of Neuroscience and
Newcastle University Sorbonne Universités
Campus for Ageing and Vitality Université Pierre et Marie Curie
Newcastle upon Tyne, United Kingdom Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A)
Institut du Cerveau et de la Moelle Épinière (ICM)
Joseph P.M. Kane
Département de Neurologie
Institute of Neuroscience
Hôpital de la Pitié-Salpêtrière
Newcastle University
Paris, France
Newcastle upon Tyne, United Kingdom
Gill Livingston
Milica G. Kramberger
Division of Psychiatry
Department of Neurology
University College London
University Medical Center Ljubljana
London, United Kingdom
Ljubljana, Slovenia
and Samantha Loi
Division of Neurogeriatrics Academic Unit for Psychiatry of Old Age
and Department of Psychiatry
Department of Neurobiology Care Sciences and Society University of Melbourne
Karolinska Institute Melbourne, Australia
Stockholm, Sweden and
Contributors xvii

NorthWestern Mental Health Bronwyn Moorhouse


Melbourne Health Royal Talbot Rehabilitation Centre
Parkville, Australia Kew, Australia

Sue Lord Sarah Shizuko Morimoto


Institute for Neuroscience Weill Cornell Medical College
Newcastle University White Plains, New York
Newcastle upon Tyne, United Kingdom
David Neary
Constantine G. Lyketsos Cerebral Function Unit, Greater Manchester
Department of Psychiatry and Behavioral Sciences Neurosciences Centre
John Hopkins Bayview Medical Center Salford Royal NHS Foundation Trust
Baltimore, Maryland Salford, United Kingdom

Clare E. Mackay and


Department of Psychiatry Institute of Brain, Behaviour and Mental Health
University of Oxford University of Manchester
and Manchester, United Kingdom
The Warneford Hospital
Peter J. Nestor
Oxford, United Kingdom
German Center for Neurodegenerative Diseases
David M.A. Mann Magdeburg, Germany
Clinical and Cognitive Neuroscience Research Group
Shirley Nurock
Institute of Brain, Behaviour and Mental Health
Former Carer
University of Manchester
London Area Coordinator of the UK
and
Alzheimer’s Society Research in Dementia
Salford Royal NHS Foundation Trust
(QRD) Network
Salford, United Kingdom
Council of Relatives to Assist in the Care of
Jill Manthorpe Dementia
Social Care Workforce Research Unit London, United Kingdom
King’s College London
John T. O’Brien
London, United Kingdom
Department of Psychiatry
Colin L. Masters School of Clinical Medicine
Florey Institute of Neuroscience and Mental Health University of Cambridge
Parkville, Australia Cambridge Biomedical Campus
Cambridge, United Kingdom
Maree Mastwyk
Florey Institute of Neuroscience and Mental Health Sid E. O’Bryant
Parkville, Australia Institute for Aging and Alzheimer’s Disease Research
Department of Internal Medicine
Ian G. McKeith
University of North Texas Health Science Center
Biomedical Research Building
Fort Worth, Texas
Newcastle University Institute for Ageing
Campus for Ageing and Vitality Daniel W. O’Connor
Newcastle upon Tyne, United Kingdom Department of Psychiatry
Monash University
Susanna Melkas
Melbourne, Australia
Clinical Neurosciences, Neurology
University of Helsinki Desmond O’Neill
and Centre for Ageing, Neuroscience and the Humanities
Helsinki University Hospital Trinity College
Helsinki, Finland Dublin, Ireland

Kirsten Moore Leonardo Pantoni


Marie Curie Palliative Care Research Department NEUROFARBA Department
Division of Psychiatry Neuroscience Section
University College London University of Florence
London, United Kingdom Florence, Italy
xviii Contributors

Joana B. Pereira Craig W. Ritchie


Division of Clinical Geriatrics Centre for Dementia Prevention
Department of Neurobiology, Care Sciences and
and Society Centre for Clinical Brain Sciences
Karolinska Institute University of Edinburgh
Stockholm, Sweden Edinburgh, United Kingdom

Nilika Perera and


Department of Old Age Psychiatry Scottish Dementia Clinical Research Network
Stepping Hill Hospital NHS Scotland
and Perth, United Kingdom
Pennine Care NHS Foundation Trust
Karen Ritchie
Manchester, United Kingdom
Institut National de la Santé et de la Recherche Médicale
Nicole Pesa (INSERM)
Dementia Collaborative Research Centre Neuropsychiatry, Epidemiological and Clinical Research
and Hôpital La Colombière
Centre for Healthy Brain Ageing Montpellier, France
University of New South Wales
Lynn Rochester
Kensington, Australia
Institute for Neuroscience
Ronald C. Petersen Newcastle University
Alzheimer’s Disease Research Center Newcastle upon Tyne, United Kingdom
Mayo Clinic
Rochester, Minnesota Jonathan D. Rohrer
National Hospital for Neurology and Neurosurgery
Anna Pink University College London Hospitals
Translational Neuroscience and Aging Program NHS Foundation Trust
Collaborative Research Building London, United Kingdom
Mayo Clinic
Scottsdale, Arizona Gustavo C. Román
Department of Neurology
Martin Prince Weill Cornell Medical College
King’s College London Houston Methodist Neurological Institute
and Houston, Texas
Centre for Global Mental Health, Health Services and
Population Research Christopher C. Rowe
Institute of Psychiatry, Psychology and Austin Health, Department of Nuclear Medicine and
Neuroscience Centre for PET
London, United Kingdom Heidelberg, Australia

Nitin Purandare (deceased) Tom C. Russ


Exeter University Centre for Dementia Prevention
Exeter, United Kingdom and
Centre for Clinical Brain Sciences
Peter V. Rabins
and
Department of Psychiatry and Behavioral Sciences
Centre for Cognitive Ageing and Cognitive
John Hopkins Medical Institutions
Epidemiology
Baltimore, Maryland
and
Michael S. Rafii Alzheimer Scotland Dementia Research Centre
Department of Neurosciences University of Edinburgh
University of California Edinburgh, United Kingdom
San Diego, California and
Glenn Rees Scottish Dementia Clinical Research Network
Alzheimer’s Disease International NHS Scotland
London, United Kingdom Perth, United Kingdom

Ruth Remington Karim Saad


Framingham State University Consultant in Old Age Psychiatry
Framingham, Massachusetts Clinical Network for Dementia
Contributors xix

NHS West Midlands Genevieve Z. Steiner


Coventry, United Kingdom National Institute of Complementary Medicine
Western Sydney University
Elizabeth Sampson
Campbelltown, Australia
Marie Curie Palliative Care Research Department
Division of Psychiatry Robert Stewart
University College London Department of Psychological Medicine
London, United Kingdom King’s College London
and
Greg Savage
Institute of Psychiatry, Psychology and Neuroscience
Department of Psychology
London, United Kingdom
ARC Centre of Excellence in Cognition and its
Disorders Elsdon Storey
Macquarie University Department of Medicine (Neuroscience)
Sydney, Australia Monash University
Melbourne, Australia
Simon J. Scalzo
Melbourne School of Psychological Sciences Joe Stratford
University of Melbourne Consultant Psychiatrist for Older People
Melbourne, Australia Stroud, United Kingdom

Katrin Seeher Daniel Strech


Dementia Collaborative Research Centre, School of Institute for History, Ethics and Philosophy of Medicine
Psychiatry Hannover Medical School
University of New South Wales Hannover, Germany.
Sydney, Australia
Sana Suri
Hugh Series Department of Psychiatry
Oxford Health NHS Foundation Trust University of Oxford
and and
University of Oxford Faculty of Law The Warneford Hospital
Oxford, United Kingdom Oxford, United Kingdom

Ajit Shah Caroline Sutcliffe


Department of Ageing, Ethnicity and Mental Health Personal Social Services Research Unit
University of Central Lancashire University of Manchester
Preston, United Kingdom Manchester, United Kingdom

Margie Smith Pierre N. Tariot


smartDNA Pty Ltd Banner Alzheimer’s Institute
MHTP Translational Research Facility University of Arizona College of Medicine
Clayton, Australia Phoenix, Arizona

John-Paul Taylor
Julie S. Snowden
Old Age Psychiatry, Institute of Neuroscience
Cerebral Function Unit
Newcastle University
Salford Royal NHS Foundation Trust
Newcastle upon Tyne, United Kingdom
Salford, United Kingdom
and Andrew Teodorczuk
Old Age Psychiatry, Institute of Neuroscience
Institute of Brain, Behaviour and Mental Health
Newcastle University
University of Manchester
and
Manchester, United Kingdom
Northumberland Tyne and Wear NHS Foundation
John Snowdon Trust
Sydney Medical School Newcastle upon Tyne, United Kingdom
University of Sydney
Alan Thomas
Sydney, Australia
Institute of Neuroscience
Kathleen A. Spangehl and
Translational Neuroscience and Aging Program Newcastle University Institute for Ageing
Mayo Clinic Newcastle University
Scottsdale, Arizona Newcastle upon Tyne, United Kingdom
xx Contributors

Jennifer Torr Robert S. Wilson


Department of Psychiatry Department of Behavioral Science and Department of
University of Melbourne Neurological Science
Melbourne, Australia Rush Alzheimer’s Disease Center
Rush University Medical Center
Anne Unkenstein
Chicago, Illinois
Department of Psychiatry
University of Melbourne Anders A. Wimo
Melbourne, Australia Division of Neurogeriatrics, NCS
Department of Neurobiology, Care Sciences and
Vyara Valkanova
Society
Department of Psychiatry
Karolinska Institutet
University of Oxford
Stockholm, Sweden
and
The Warneford Hospital Bengt Winblad
Oxford, United Kingdom Department of Neurobiology, Care Sciences and
Society
Jenny T. van der Steen
Karolinska Institutet
EMGO Institute for Health and Care Research
Stockholm, Sweden
VU University Medical Center
Amsterdam, the Netherlands Michael Woodward
Aged Care
Victor L. Villemagne
Austin Health
Department of Nuclear Medicine and Centre for PET
Victoria, Australia
Austin Health
and and
The Florey Institute of Neuroscience and Mental Health University of Melbourne
Heidelberg, Australia Melbourne, Australia
Jason D. Warren Paul Yates
Dementia Research Centre Department of Aged Care Services
Institute of Neurology Austin Health
University College London Heidelberg, Australia
London, United Kingdom
and
Flavie Waters University of Melbourne
School of Psychiatry and Clinical Neurosciences Melbourne, Australia
University of Western Australia
Crawley, Australia Genevieve S. Yuen
Weill Cornell Medical College
and
White Plains, New York
Clinical Research Centre
North Metropolitan Health Service Mental Health Henrik Zetterberg
Mt. Hawthorn, Australia Clinical Neurochemistry Laboratory
Department of Psychiatry and Neurochemistry
Alissa Westphal Institute of Neuroscience and Physiology
Department of Psychiatry The Sahlgrenska Academy at the University of
University of Melbourne Gothenburg
and Mölndal, Sweden
NorthWestern Mental Health, Melbourne Health
and
Victoria, Australia
Department of Molecular Neuroscience
Beverley Williams UCL Institute of Neurology
Aged Psychiatry Service London, United Kingdom
Caulfield Hospital
Caulfield, Australia
Part     1
Dementia: General aspects

1 Dementia: Historical overview 3


German Berrios
2 The lived experience of dementia 19
Shirley Nurock
3 Prevalence and incidence of dementia 24
Daniel W. O’Connor
4 What is dementia, and how do you assess it? Definitions, diagnostic criteria and assessment 33
Joseph P.M. Kane and Alan Thomas
5 Screening and assessment instruments for the detection and measurement of cognitive impairment 44
Leon Flicker
6 Neuropsychological assessment of dementia 52
Greg Savage
7 Neuropsychiatric aspects of dementia 59
Nilika Perera, Mehran Javeed, Constantine G. Lyketsos and Iracema Leroi
8 Measurement of behaviour disturbance, non-cognitive symptoms and quality of life 67
Ajit Shah
9 Cross-cultural issues in the assessment of cognitive impairment 79
Ajit Shah
10 Structural brain imaging 92
Robert Barber and John T. O’Brien
11 Functional brain imaging and connectivity in dementia 107
Klaus P. Ebmeier, Nicola Filippini, Clare E. Mackay, Sana Suri and Vyara Valkanova
12 Molecular brain imaging in dementia 119
Victor L. Villemagne and Christopher C. Rowe
13 Services for people with dementia 135
Sube Banerjee
14 Family carers of people with dementia 142
Katrin Seeher and Henry Brodaty
15 Dementia care in the community: Challenges for primary health and social care 161
Steve Iliffe, Jill Manthorpe and Vari Drennan
16 Managing people with dementia in the general hospital 172
Andrew Teodorczuk, Rowan Harwood and Elizabeth Sampson
17 The role of nursing in the management of dementia 183
Maree Mastwyk and Beverley Williams
18 Social work and care/case management in dementia 192
David Challis, Jane Hughes and Caroline Sutcliffe
19 Occupational therapy in dementia care 199
Alissa Westphal
20 Speech and language therapy in dementia assessment and management 208
Bronwyn Moorhouse and Caroline A. Fisher
2 Dementia

21 Role of the physiotherapist in the management of dementia 220


Sue Lord and Lynn Rochester
22 Therapeutic effects of music in persons with dementia 229
Linda A. Gerdner and Ruth Remington
23 Psychological, behavioural and psychosocial interventions for neuropsychiatric symptoms in dementia:
What works, what does not and what needs more evidence? 235
Gill Livingston and Claudia Cooper
24 Treating problem behaviours in dementia by understanding their biological, social and psychological causes 244
Ian James and Louisa Jackman
25 Treatments for behavioural and psychological symptoms in Alzheimer’s disease and other dementias 257
Anna Burke, William J. Burke and Pierre N. Tariot
26 Psychological approaches for the practical management of cognitive impairment in dementia 279
Anne Unkenstein
27 Sexuality and dementia 285
Joe Stratford
28 Residential care for people with dementia 292
David Conn, John Snowdon and Nitin Purandare
29 Design and dementia 304
June Andrews
30 Legal issues and dementia 308
Hugh Series and Robin Jacoby
31 Driving and dementia 314
Aoife Fallon and Desmond O’Neill
32 Quality of life in dementia: Conceptual and practical issues 325
Betty S. Black and Peter V. Rabins
33 Ethical issues 339
Julian C. Hughes and Daniel Strech
34 End-of-life and palliative care in dementia 344
Julian C. Hughes and Jenny T. van der Steen
35 Advanced dementia and care at the end of life 349
Kirsten Moore and Elizabeth Sampson
36 Alzheimer’s associations and societies 359
Henry Brodaty, Nicole Pesa and Glenn Rees
37 Health economic aspects of dementia 371
Anders A. Wimo, Linus Jönsson and Bengt Winblad
38 Global challenge of dementia: What can be done? 388
Cleusa P. Ferri, Maëlenn Guerchet and Martin Prince
39 The international challenge of dementia 402
Karim Saad
40 The pharmaceutical industry and dementia: How can clinicians, researchers and industry work together for the
betterment of people with dementia and their families? Continuous vigilance and transparency is the answer 411
Martin M. Bednar and Leon Flicker
1
Dementia: Historical overview

GERMAN BERRIOS

name different concepts and different behaviours); indeed,


1.1 MATTERS HISTORIOGRAPHICAL currently it is part of yet another convergence. How long this
current bundle will last is unclear. Historians are only too
Exploring why clinical textbooks often carry historical aware of the so-called presentistic fallacy (latest is b
­ estest)
chapters can be informative. One common reason for such and of how this distortion of reality tends to make research-
addition is fashion. Although for some time now such chap- ers feel that the current narrative is (1) superior to all past
ters are almost de rigueur, editors vary in what they want. ones; and (2) the truth.
Some just want lists of achievements as a celebration of Since the fourth edition of this textbook, no ground-
scientific progress and this task is best met by retired clini- breaking scholarly work has been published to challenge
cians. Others go further and ask for a sort of meta-analysis the historical hypotheses put forward in earlier versions of
of historical papers; in practice, this leads to requesting this chapter (e.g. Boller, 2008). If anything, developments
‘updated’ chapters for each new edition of the textbook. in the history and philosophy of science support its con-
Going further a third group of editors considers them a structionist perspective (Golinski, 1998). As mentioned
necessity. Their view is correct because symptoms and dis- above, clinical categories (whether symptoms or disorders)
orders are inherently historical; that is, objects constructed result from the bundling up of selected morsels of behav-
in a semantic space and made temporarily stable by means iour, explanatory concepts and words. Complex social and
of specific social narratives networks neurobiological cor- economic variables then determine whether the ensuing
relations. In the case in hand, history has shown that all bundle will last, and for how long. For reasons that have to
narratives about dementia have been: (1) born in specific do with the rhetoric of science, these social acts are sold as
historical convergences and (2) considered as ‘true’ during scientific acts. For example, it would be historically naive
the time they were socially predominant. to believe that the decision to consider a symptom-cluster,
such as Lewy Body dementia, as a ‘new disease’ is solely
1.1.1 CONVERGENCES based on the discovery of powerful, ineluctable and repli-
cable correlations (Perry et al., 1996). Given that there is no
Convergences are historical processes whereby words clear theory linking firstly the symptoms to each other, and
(names, terms) and concepts (definitions, accounts, expla- secondly the symptom bundle to the Lewy bodies them-
nations) are used to: (1) break up the complaints and selves, it is not unreasonable to believe that social variables
behaviour of certain individuals; and (2) select some of have also played a role in the acceptance of this new variant
the fragments on the basis that they constitute a mean- of dementia.
ingful bundle (Berrios, 2011). Convergences tend to be This is not a new state of affairs and science has always
made explicit in writings: (1) inscribed in the predomi- been made in the same way, for example, it was thus at
nant social media and (2) by authors who are seen as the the time Kraepelin constructed the concept of Alzheimer’s
spokesmen of fashionable cliques. Hence, although social disease (Berrios, 1990a). In fact, there is nothing wrong
usefulness tends to be the main determinant of the dura- with accepting that social forces shape scientific facts,
tion of convergences, they are rhetorically presented as a ­particularly in psychiatry. More to the point, accepting
scientific truth. such view would render neurobiologists less fundamen-
As this chapter will show, the word dementia has par- talist in their claims, and clinicians more useful to their
ticipated in a number of convergences (i.e. has been used to patients.

3
4 Dementia

Knowledge of the successive convergences that consti- modern writers confuse dementia with mania,
tute the history of dementia and of the current conver- which is a delusional state accompanied by dis-
gence should be a precondition to do scientific research. turbed behaviour (audace); these symptoms
For the purposes of this chapter, it should suffice to map are not present in subject with dementia who
these convergences since the eighteenth century, for exhibit foolish behaviour and cannot under-
example, from that implied in the work of Boissier de stand what they are told, cannot remember
Sauvages (1771) to the one propounded by Marie (1906). anything, have no judgment, are sluggish and
The former defined dementia as a generic term; the latter retarded …
saw in dementia a syndrome, which could be enacted in Physiology teaches that the vividness of
a variety of diseases each with its recognisable phenom- our understanding depends on the intensity
enology and putative neuropathology. Current research of external stimuli … in pathological states
on dementia remains inscribed in Marie’s epistemological these may be excessive, distorted or abolished;
frame. dementia results from abolition of stimuli which
may follow: 1. damage to the brain caused by
excessive usage, congenital causes or old age,
1.1.1.1 Words and concepts 2. failure of the spirit, 3. small volume of the
brain, 4. violent blows to the head causing brain
In Roman times, the word dementia was used to mean damage, 5. incurable diseases such as epilepsy,
‘being out of one’s mind, insanity, madness, folly’ (Lewis or exposure to venoms (Charles Bonnet reports
and Short, 1969). For example, in the first century bc, of a girl who developed dementia after being
Cicero (1969) (Tusculanan disputations, Book 3, para bitten by a bat) or other substances such as opi-
10) and Lucretius (1975) (De Rerum Natura, Book 1, line ates and mandragora.
704) used dementia as a synonym of madness. Indeed, Dementia is difficult to cure as it is related
up to the 1700s, states of cognitive and behavioural dete- to damage of brain fibres and nervous fluids; it
rioration of whatever origin that ended up in psychoso- becomes incurable in cases of congenital defect
cial incompetence were still being called by a variety of or old age … [otherwise] treatment must follow
names: amentia, dementia, imbecility, morosis, fatuitas, the cause …
anoea, foolishness, stupidity, simplicity, carus, idiocy, [The legal definition of dementia reads]:
dotage and senility. ‘Those in a state of dementia are incapable of
The term dementia first appears in the European ver- informed consent, cannot enter into contracts,
naculars during the seventeenth century. In Blancard’s sign wills, or be members of a jury. This is why
English dictionary (1726) it is used as an equivalent of anoea they are declared incapable of managing their
or ‘extinction of the imagination and judgment’ (p. 21). own affairs. Actions carried out before the dec-
By 1644, according to the Oxford English Dictionary, an laration of incapacity are valid unless it is dem-
adjectival form demented entered the English language. onstrated that dementia predated the action.
In his Spanish-French dictionary, Sobrino (1791) wrote: Ascertainment of dementia is based on
‘­
demencia = démence, folie, extravagance, égarement, examination of handwriting, interviews by
alienation d’esprit’ (p. 300). Rey (1995), in turn, states that magistrates and doctors, and testimony from
démence appeared in French in 1381 to refer to ‘madness, informants. Declarations made by notaries
extravagancy’ but that the adjective dément only came into that the individual was of sane mind whilst
currency by 1700. It would seem, therefore, that between signing a will are not always valid as they may
the seventeenth and eighteenth centuries derivatives of the be deceived by appearances, or the subject
Latin stem demens (without mind) were becoming estab- might have been in a lucid period. In regards
lished in most European vernaculars. As we shall see pres- to matrimonial rights, démence is not a suf-
ently the full medicalisation of dementia started after the ficient cause for separation, unless it is accom-
1750s. panied by aggression (furour). It is, however,
Evidence for an early medical usage of the term demen- sufficient for a separation of property, so that
tia is found in the French Encyclopaedia (Diderot and the wife is no longer under the guardianship
d’Alembert, 1765): of her husband. Those suffering from demen-
tia cannot be appointed to public positions or
Dementia is a disease consisting in a paralysis of receive privileges. If they became demented
the spirit characterized by abolition of the rea- after any has been granted, a coadjutor should
soning faculty. It differs from fatuitas, morosis, be appointed …’
stultitia and stoliditas in that in the latter there
is a weakening of understanding and memory; Although modern-sounding, the above definition must
and from delirium which is a temporary impair- be read with caution: its clinical description depends
ment in the exercise of the said functions. Some on contrasts and differences with delirium and a list of
Dementia: Historical overview 5

disorders, which are no more; its legal meaning is based on ‘Imbecility’ are bêtise (stupidity, foolishness), niaiserie
the old Roman accounts, and its mechanisms make use of (silliness) and démence; in Greek paranoia, and demen-
the camera obscura metaphor and assume the passive defi- tia, fatuitas and vecordia in Latin. The term is used to
nition of the mind that Condillac (who inspired the author refer to patients who are fools, (fous), imbeciles (imbécil-
of the article) had borrowed from John Locke. les), mentally weak (foibles d’esprit), mad (insensés).’ (p.
723). Boissier lists 12 subtypes of imbecility of which the
first one is the imbecility of the elderly (l’imbécillite de
1.1.1.2 Behaviours vieillard), also known as puerile state, drivelling or senile
madness, and which he explains thus: ‘Because of the
There are two ways of conceiving diseases in the history stiffness of their nervous fibres, old people are less sensi-
of medicine. According to the conventional view, medical tive to external impressions …’ (p. 724).
discoverers are like botanists exploring a new forest. Now In the Nosographie (1818; first edition 1798), Pinel dealt
and again they discover a ‘new species’ of plant, name it with cognitive impairment under amentia and morosis,
and offer it to the world. The plant has always been there, which he explains as a failure in the association of ideas
only that undiscovered. The act of discovery does not add leading to disordered activity, extravagant behaviour, super-
anything to its essence. The second view is that discover- ficial emotions, memory loss, difficulty in the perception of
ing a new disease is tantamount to constructing it out of an objects, obliteration of judgement, aimless activity, auto-
opaque mass of symptoms and complaints. The languages of matic existence and forgetting of words or signs to convey
description involved (whether phenomenological, anatomi- ideas. He also referred to démence sénile (para 116). Pinel
cal or molecular) actually create the boundaries for the new did not remark upon the difference between congenital and
disease. These are called the ontological and constructionist acquired dementia (Pinel, 1806).
views, respectively. The above entries summarize well the views on
Taken these views into account, we can say that asking dementia before the nineteenth century. There was, first
the frequent question ‘where were the patients with demen- of all, a legal meaning according to which dementia
tia type a, b or c before the twenty-first century’? (when the was a state of non-imputability due to mental incompe-
proper groupings have at long last been achieved!) reflects a tence (of whatever origin). In France, this was enshrine
belief in the ontological view, in the view that twenty-first in Article 10 of the Napoleonic Code: ‘There is no crime
century types of dementia have always existed except that when the accused is in a state of dementia at the time of
earlier medics were not clever enough to describe them the alleged act’ (Code Napoléon, 1808). Secondly, there
properly. The keen clinical-historian, wearing anachronistic
was a clinical meaning. This could be very general (i.e.
spectacles can indeed find in the literature of the seventeenth
a synonym of madness) or specific, that is a clinical con-
and eighteenth century (and indeed of earlier periods) cases
dition, which was differentiable from mania (which, at
that ‘sound as if’ they had dementia a, b or c. The thinking
the time, described as any state of acute excitement be
here is similar to that seen in pathographic diagnosis: Did
it schizophrenic, hypomanic or organic) and delirium
St. Theresa of Jesus have epilepsy, Dr. Johnson Gilles de la
(which referred, more or less, to what goes on nowa-
Tourette syndrome or Mozart Asperger’s?
days under the same name). Although chronic, demen-
For example, in relation to ‘Stupidity or Foolishness’,
tia could still be reversible, affect individuals of any age
Thomas Willis (1684) wrote ‘although it chiefly belongs to
and be a final common pathway, that is the end deficit for
the rational soul, and signifies a defect of the intellect and
many other mental disorders. This created a template for
judgement, yet it is not improperly reckoned among the
the alienists of the nineteenth century.
diseases of the head or brain; for as much as this eclipse of
the superior soul proceeds from the imagination and the
memory being hurt, and the failing of these depends upon
the faults of the animal spirits, and the brain itself’ (p. 209). 1.2 THE NINETEENTH CENTURY
Willis suggested that stupidity might be genetic (original, as
when ‘fools beget fools’) or caused by ageing (‘some at first 1.2.1 BRAIN CHANGES AND AGEING
crafty and ingenious, become by degrees dull, and at length
foolish, by the mere declining of age, without any great Since the beginning of the nineteenth century, cases had
errors in living;) (p. 211) or other causes such as ‘strokes or been described of brain softening followed by cognitive fail-
bruising upon the head’, ‘drunkenness and surfeiting’, ‘vio- ure. For example, Rostan (1823) divided 98 subjects with
lent and sudden passions’ and ‘cruel diseases of the head’ brain softening due to scurvy into three groups: simple,
such as epilepsy. abnormal and complicated, the latter two showing mental
The same is the case with Boissier de Sauvages (1771), symptoms. These occurred before, during and after the soft-
one of the great classificators of the eighteenth century. ening itself; thus, senile dementia and insanity might pre-
Order three (eighth class) of his Nosographie Methodique cede the brain changes. Related to stroke, Rostan described
encompasses delirium, paraphrosyne, imbecility, cognitive failure and attacks of insanity suggesting that
melancholia, demonomania and mania. Synonyms of these symptoms were ‘a general feature … not a positive
6 Dementia

sign of localisation’ (pp. 214–215). Durand-Fardel (1843) (standard deviation = 10.9); seven being, in fact, cases of
provided an account of the relationship between softening general paralysis of the insane showing grandiosity, disin-
and insanity, warning that softening was used to refer both hibition, motor symptoms, dysarthria and terminal cog-
to a disease (stroke) and to a state of the brain. Psychiatric nitive failure. There also was included a 20-year-old girl
complications were acute and long term, the former includ- who in modern terms might have suffered from a catatonic
ing confusion, depression, irritability, acute insanity and syndrome; and a 40-year-old woman with pica, cognitive
loss of mental faculties (p. 139); the latter, of gradual onset, impairment and space-occupying lesions in her left hemi-
included impairment of memory, poverty of thinking and a sphere and cerebellum (Esquirol, 1838). Although the mean
regression to infantile forms of behaviour, features that led age of this sample and the absence of cases of senile demen-
to ‘true dementia’ (pp. 327–328). tia may simply reflect a selection bias or the much shorter
Although J.H. Jackson (1875) wrote ‘softening … as a life expectancy operating in Esquirol’s day, it is more likely
category for a rude clinical grouping was to be deprecated’ to reflect the view that age was not an important variable for
(p. 335), he proceeded to follow Durand-Fardel’s classifica- the definition of dementia. Together with irreversibility, age
tion suggesting that mental symptoms might follow stroke only became a defining criterion by the second half of the
immediately or wait for days and even months. Jackson nineteenth century (only in some forms of dementia).
recognized that major cognitive failure may ensue, and saw Aware of the importance of clinical description like his
this as an instance of dissolution, according to which emo- teacher Esquirol, Calmeil wrote: ‘It is not easy to describe
tional symptoms were considered as release phenomena (for dementia, its varieties and nuances; because its complica-
an analysis of dissolution see Berrios, 1991). Lastly, Jackson tions are numerous … it is difficult to choose its distinctive
suggested that anxiety, stress and irritability might be har- symptoms’ (p. 71). Dementia followed chronic insanity and
bingers of stroke. brain disease, and was partial or general. Calmeil was less
A major difference can be noticed between the eigh- convinced than his co-student Georget that all dementias
teenth century views of dementia and those set down a cen- were associated with alterations in the brain.
tury later when this category was divided up into subtypes In regard to senile dementia, Calmeil remarked: ‘There
with the so-called ‘senile type’ referring to irreversible states is a constant involvement of the senses, elderly people can
of cognitive impairment affecting the elderly. Amentia, be deaf, and show disorders of taste, smell and touch; exter-
until then used as a synonym of dementia was redefined as nal stimuli are therefore less clear to them, they have little
a ‘psychosis with sudden onset following severe, often acute memory of recent events, live in the past, and repeat the
physical illness or trauma’ (Meynert, 1890). This does not same tale; their affect gradually wanes away …’ (p. 77). A
mean that the syndromatic view of the dementias had been keen neuropathologist, Calmeil concluded that there was no
abandoned, indeed, the so-called vesanic dementias (i.e. ter- sufficient information on the nature and range of anomalies
minal states for all manner of mental disorders) continued found in the skull or brain to decide on the cause of demen-
well into the beginning of the twentieth century. tia (pp. 82–83) (Calmeil, 1835).
A Ghent alienist, thinking in Flemish and writing in
1.2.2 DEMENTIA IN SOME COUNTRIES French, Guislain (1852) reported that in dementia: ‘All
intellectual functions show a reduction in energy, exter-
1.2.2.1 France nal stimuli cause only minor impression on the intel-
lect, imagination is weak and uncreative, memory absent
In his doctoral thesis, Esquirol (1805) used the word and reasoning pathological … There are two varieties of
dementia to refer to loss of reason, as in démence acciden- dementia … one affecting the elderly (senile dementia of
tal, démence mélancolique; and proceeded to distinguished Cullen) the other younger people. Although confused with
between acute, chronic and senile dementia. Acute demen- dementia, idiocy must be considered as a separate group’
tia was short-lived, reversible and followed fever, haemor- (p. 10). In dementia, ‘the patient has no memory, or at
rhage and metastasis; chronic dementia was irreversible least is unable to retain anything … impressions evapo-
and caused by masturbation, melancholia, mania, hypo- rate from his mind. He may remember names of people
chondria, epilepsy, paralysis and apoplexy; lastly, senile but cannot say whether he has seen them before. He does
dementia resulted from ageing, and consisted in a loss of the not know what time or day of the week it is, cannot tell
faculties of understanding (Esquirol, 1814). Esquirol’s final morning from evening, or say what 2 and 2 add to … he
thoughts on dementia were influenced by his controversy has lost the instinct of preservation, cannot avoid fire or
with Bayle (1822) who via his notion of chronic arachnoidi- water and is unable to recognize dangers; has also lost
tis propounded an anatomical (organic) view of all forms of spontaneity, is incontinent of urine and faeces and does
insanity and scorned Pinel’s views that some vesanias might not ask for ­a nything, he cannot even recognize his wife or
develop in a psychological space (Bayle, 1826). children …’ (p. 311) (Guislain, 1852).
Together with his student Georget, Esquirol supported Because in the past the mentally ill: ‘Had been catego-
a descriptivist approach, at least in relation to some forms rized only in terms of a [putative] impairment of their men-
of mental disorder. He reported 15 cases of ‘dementia’ tal faculties …’ (p. 2), Morel (1860) endeavoured to develop
(seven males and eight females) with a mean age of 34 years a taxonomy that distinguished between occasional and
Dementia: Historical overview 7

determinant causes of mental disorder (p. 251) and sug- apprehension. Numerous examples of this dis-
gested six clinical groups: hereditary, toxic, associated with ease, or decay of the mental powers are to be
the neuroses, idiopathic, sympathetic and dementia. met within every receptacle containing a con-
In regards to the latter, Morel (1860) believed that: siderable assemblage of deranged persons …
incoherence is either a primary disease, arising
… if we examine dementia (amentia, progres- immediately from the agency of exciting causes
sive weakening of the faculties) we must accept on a constitution previously health, or it is a sec-
that it constitutes a terminal state. There will, of ondary affection, the result of other disorders
course, be exceptional insane individuals who, of the brain and nervous system which, by their
until the end, preserve their intellectual facul- long duration or severity, give rise to disease in
ties; the majority, however, are subject to the the structure of those organs … secondary inco-
law of decline. This results from a loss of vital- herence or dementia follows long-protracted
ity in the brain … Comparison of brain weights mania, attacks of apoplexy, epilepsy or paraly-
in the various forms of insanity shows that the sis, or fevers attended with severe delirium. This
heavier weights are found in cases of recent decay of the faculties has been termed fatuity
onset. Chronic cases show more often a gen- or imbecility, and it has been confounded with
eral impairment of intelligence (dementia). Loss idiotism, which in all its degrees and modifica-
in brain weight – a constant feature of dementia tions is a very different state … (Prichard, 1835,
– is also present in ageing, and is an expression pp. 83–85).
of decadence in the human species. [There are]
natural dementia and that dementia resulting The same can be said of the views expressed by Bucknill
from a pathological state of the brain … some and Tuke in their popular textbook:
forms of insanity are more prone to end up in
dementia (idiopathic) than others … it could Dementia may be either primary or consecu-
be argued that because dementia is a terminal tive; acute or chronic. It may also be simple or
state it should not be classified as a sixth form complicated; it is occasionally remittent but
of mental illness … I must confess I sympathise rarely intermittent. It is primary when it is the
with this view, and it is one of the reasons why I first stage of the mental disease of the patient;
have not described the dementias in any detail and when this occurs, it is, perhaps, one of
… on the other hand from the legal and patho- the most painful forms of insanity; the patient
logical viewpoints, dementia warrants separate often being acutely sensible of a gradual loss
treatment … (pp. 837–38). of memory, power of attention and executive
ability. At this period, the distinction is often
Morel’s view was in keeping with his own theory of well marked between the strictly intellectual
‘degeneration’ (Morel, 1857; Pick, 1989), that is, in tainted and affective disorder … dementia is much
pedigrees, successive generation show worse forms of men- more frequently consecutive, that is the conse-
tal disorder, ending up in dementia. Morel believed that quence of other diseases of the mind. Thus dur-
there were no specific brain alterations in dementia. This ing 44 years, while 277 cases of mania and 215
was challenged by L.F. Marcé (1863) in a series of articles on of melancholia were admitted at the Retreat,
the neuropathology of senile dementia. only 48 of dementia were admitted during the
same period; yet, at the end of that term, there
1.2.2.2 Great Britain were remaining in the institution, 20 patients in
a state of dementia out of 91 inmates. Mania
There is no space in this chapter to analyse with the same very often degenerates into dementia; as also
level of detail the evolution of the concept of dementia in do melancholia and monomania … it should be
other European countries although it can be said that it observed, that the term dementia may be, and
followed similar lines. Views in England, for example, were sometimes is, too indiscriminately employed.
mainly derivative from French ones. In a popular textbook, All writers of authority agree in representing
and following Pinel, Esquirol and Calmeil, Prichard included impairment of the memory as one of the earli-
a category, which he called ‘incoherence or dementia’: est symptoms of dementia; but we believe cases
are occasionally classed under incipient demen-
[It] is a very peculiar and well-marked form tia, in which close observation would show that
of mental disorder. The mind in this state is the memory is unimpaired. … It is often rather
occupied, without ceasing, by unconnected a torpid condition of the mind, falling under the
thoughts and evanescent emotions; it is incapa- division ‘apathetic insanity, which ought not to
ble of continued attention and reflection, and at be confounded with dementia …’ (Bucknill and
length loses the faculty of distinct perception or Tuke, 1858, pp. 117–119).
8 Dementia

1.2.2.3 Germany 1.2.3 THE FRAGMENTATION OF


DEMENTIA
Similar concepts are found in German-speaking nations
and the views of Heinroth, Feuchtersleben, Griesinger and During the second half of the nineteenth century, and
Kahlbaum were influential until the beginning of the sec- based on the clinical observations and reconceptualiza-
ond half of the nineteenth century. Heinroth (1975) used the tion carried out by the French, German and English writ-
term dementia in a very broad sense to refer to a state of ers mentioned above, dementia starts to be considered as a
mind, which might accompany or follow other mental dis- syndrome and hence could be attached to a variety of disor-
orders, that is, as vesanic dementia, a term which late in the ders. The primary classification was to be between primary
century was to become very popular, particularly in France. and secondary, the latter including all the vesanic demen-
This concept, which is not related to age, is redolent of the tias, that is states of defect that could follow any severe form
later notion of secondary dementia, that is the state of psy- of insanity. An increased use of light microscopy during
chosocial and cognitive incompetence that might follow a the second half of the century led to the view that primary
functional psychosis. forms of dementia could be caused by degenerations of
Feuchtersleben’s (1847) usage is even more general. He cerebral parenchyma or by arteriosclerosis. By 1900, senile,
uses dementia as a synonym of madness and may refer to arteriosclerotic, infectious and traumatic forms of demen-
forms of acute madness with and without accompanying tia had been reported (Berrios and Freeman, 1991). Mixed
idiocy. There is only one form of dementia, which he refers forms, such as dementia praecox were also suggested reflect-
as moria and considered as more or less chronic and more ing Kahlbaum’s view that mental disorders appeared dur-
or less cognitive. Although the patient may end up in a state ing specific biological transitions. The list of parenchymal
of idiocy he can also show lucid intervals. Once again, age is forms became longer after the inclusion of states of degen-
of no relevance to moria and hence one must conclude that eration due to alcoholism, epilepsy, myxoedema and lead
Feuchtersleben is referring to a form of vesanic dementia. poisoning. The history of some of these forms will be dis-
Griesinger’s nosology is not altogether clear and has cussed presently.
often been interpreted as being based on the belief that there
is only one form of madness (unitary psychoses concept), 1.2.3.1 Vesanic dementias
which may go through at least three stages: depression (as
in melancholia), exaltation (as in mania) and weakness (as The term vesanic dementia began to be used after the 1840s
in chronic madness and dementia). In the second edition to refer to the clinical states of cognitive disorganisation fol-
of his great work, Griesinger (1861/1867) insists that the lowing insanity (Berrios, 1987); its meaning, has changed
states of mental weakness ‘do not constitute primary but pari passu with psychiatric theory. According to the uni-
consecutive forms of insanity’ (p. 319). This suggests that he tary insanity notion, vesanic dementia was a terminal stage
is also referring to a form of vesanic dementia although he (after mania and melancholia); according to degeneration
includes under this large class all the forms of mental handi- theory, it was the final expression of a corrupted pedigree;
cap where no preliminary primary forms of madness can and according to post-1880s nosology, a final common
be recognized. Under the heading ‘dementia’, Griesinger pathway to some insanities. Vesanic dementias were revers-
includes mental disorders fundamentally caused by a ‘gen- ible, and could occur at any age; risk factors such as old age,
eral weakness of the mental faculties’ including loss of emo- lack of education, low social class, bad nutrition, etc., accel-
tions. Age is not a factor in the development of dementia or erated the progression of the dementia or impeded recovery
apathetic dementia and hence it must also be concluded that (p. 597) (Ball and Chambard, 1881).
Griesinger is referring to vesanic dementia. The vesanic dementias included cases of melancho-
The work of Kahlbaum, particularly his important book lia with marked cognitive impairment. Under the term
of 1863 on the definition and classification of mental dis- démence mélancolique Mairet (1883) reported cases of
orders marks the beginning of a new era in psychiatry. His depressed patients with cognitive impairment who on post-
incorporation of time as a variable in the analysis of mad- mortem showed alterations in the temporal lobe; this led
ness (longitudinal definition) and his view that period of life Mairet to hypothesize that the affected sites were related to
is relevant to the form of the disease remain the pillars of feelings, and to suggest that nihilistic delusions appeared
psychiatric nosology to this day. The concept of dementia when the lesion spread to the cortex (Berrios, 1985a).
is dealt with in the third section of Kahlbaum’s book (1863) Mairet’s cases, some of which would now be called Cotard’s
under the name of aphrenia. This clinical category refers syndrome showed psychomotor retardation, refused food
to states of mental impotence (Zustand geistiger Impotenz) and died in stupor (Cotard, 1882; Berrios and Luque, 1995).
(p. 153), which Kahlbaum equates to the old German notion Other cases got better and are redolent of what later on was
of Blödsinn. After complaining that neither the Greeks nor called depressive pseudodementia.
Latin writers managed to specify a term for this condition, Another contributor to the understanding of cogni-
he insists that a word is needed to refer to states of cognitive tive impairment in the affective disorders was George
and behavioural incompetence such as those seen in demen- Dumas (1894) who suggested that it was ‘mental fatigue
tia terminalis (Berrios, 1996). that explained the psychological poverty and monotony
Dementia: Historical overview 9

of melancholic depressions’ and that the problem was In general, there is little evidence that alienists considered
not ‘an absence but a stagnation of ideas’; i.e. he was, general paralysis as a paradigm disease, i.e. a model for all
therefore, the first to explain the disorder as a failure in other mental diseases. It can even be said that the new ‘dis-
performance. ease’ created more problems than it solved (for a discussion
The word pseudodementia, however, had originated of this issue see Berrios, 1985a).
in a different clinical tradition and was first used by Carl
Wernicke to refer to ‘a chronic hysterical state mimicking 1.2.3.3 Arteriosclerotic dementia
mental weakness’ (Bulbena and Berrios, 1986). In the 1950s
it was given a lease of life by writers such as Madden et al. Old age was considered as an important factor in the devel-
(1952), Anderson et al. (1959) and Kiloh (1961). Current opment of arteriosclerosis (Berrios, 1994) and a risk factor
usage remains ambiguous as it relates to three clinical in diseases such as melancholia (Berrios, 1991). By 1910,
situations: a real (albeit reversible) cognitive impairment there was a trend to include all dementias under ‘men-
accompanying some psychoses, a parody of such impair- tal disorders of cerebral arteriosclerosis’ (Barrett, 1913).
ment and the cognitive deficit of delirium (Bulbena and Arteriosclerosis, might be generalized or cerebral, inherited
Berrios, 1986). or acquired and caused by syphilis, alcohol, nicotine, high
blood pressure and ageing. In those genetically predisposed,
1.2.3.2 General paralysis of the insane cerebral arteries were considered as thinner and less elas-
tic. Arteriosclerosis caused mental changes by narrowing of
Bayle (1822) described under the name arachnitis chro- arteries and/or reactive inflammation. The view that arte-
nique cases of what later was to be called ‘general paraly- riosclerotic dementia resulted from a gradual strangulation
sis of the insane’. Whether this ‘new phenomenon’ resulted of blood supply to the brain was also formed during this
from ‘a mutation in the syphilitic virus towards the end period; consequently, emphasis was given to prodromal
of the eighteenth century’ is unclear (p. 623) (Hare, 1959). symptoms and strokes were but the culmination of a pro-
Equally dubious is the claim that its discovery reinforced cess started years before.
the belief of alienists in the anatomo-clinical view of men- Some opposed this view from the beginning. For
tal disease (Zilboorg, 1941). In fact, it took more than 30 example, Marie (1906) claimed that such explanation was
years for general paralysis to gain acceptance as a ‘separate’ a vicious circle as alienists claimed both that: ‘Ageing was
disease. Bayle’s ‘discovery’ was more important in other caused by arteriosclerosis and the latter by ageing’ (p. 358),
way, namely, in that it challenged the cross-sectional view of and Walton (1912) expressed serious doubts from the his-
­disease; in the words of Bercherie (1980): ‘For the first time topathological point of view. The frequent presence in
in the history of psychiatry there was a morbid entity which ­post-mortem of such changes also concerned pathologists
presented itself as a sequential process unfolding itself into who worried that they could not ‘safely exclude cerebral
successive clinical syndromes’ (p. 75). arteriosclerosis of greater or less degree in any single case’ of
By the 1850s, no agreement had yet been reached as to senile dementia (p. 677) (Southard, 1910). Based on a review
how symptoms were caused by the periencephalite chro- of these arguments, Olah (1910) concluded that there was no
nique diffuse (as general paralysis was known at the time). such a thing as ‘arteriosclerotic psychoses’. But the ‘chronic
Three clinical types were recognized: manic-ambitious, global ischaemia’ hypothesis won the day, and it was to
melancholic-hypochondriac and dementia; according to the continue well into the second half of the twentieth century.
unitary view, all three constituted stages of a single disease, For some it became a general explanation; for example,
the order of their appearance depending on the progress of North and Bostock (1925) reported a series of 568 general
the cerebral lesions. Baillarger (1883), however, sponsored psychiatric cases in which around 40% suffered from arte-
a dualist view: ‘Paralytic insanity and paralytic dementia rial disease, which—according to the authors—was even
are different conditions’. It is clear that the debate had less responsible for schizophrenia. The old idea of an apoplectic
to do with the nature of the brain lesions than with how form of dementia, however, never disappeared.
mental symptoms and their contents were produced in gen-
eral: How could the ‘typical’ content of paralytic delusions 1.2.3.4 Apoplectic dementia
(grandiosity) be explained? Since the same mental symp-
toms could be seen in all manner of conditions, Baillarger Apoplectic dementia achieved its clearest enunciation in the
(1883) believed that chronic periencephalitis could only work of Benjamin Ball (Ball and Chambard, 1881). Organic
account for the motor signs—mental symptoms ‘there- apoplexy resulted from bleeding, softening or tumour and
fore, having a different origin’ (p. 389). The absence of a might be ‘followed by a notable decline in cognition, and
link between lesion and symptom also explained why some by a state of dementia which was progressive and incur-
patients recovered. able … of the three, localized softening (ramollissement en
The view that general paralysis might be related to syphi- foyer) caused the more severe states of cognitive impair-
lis (put forward by Fournier, 1875) was resisted. Indeed, the ment’ (p. 581). Ball believed that prodromal lapses of cog-
term pseudo-general paralysis was coined to refer to cases nition (e.g. episodes of somnolence and confusion with
of infections causing psychotic symptoms (Baillarger, 1889). automatic behaviour, for which there was no memory after
10 Dementia

the event) and sensory symptoms were caused by athero- two factors: cerebral arteriosclerosis and cyclothymic pre-
matous lesions. Visual hallucinations, occasionally of a morbid personality. Lafora (1935) emphasized the role of
pleasant nature, were also common. After the stroke, per- cerebrovascular pathology, and claimed that disinhibition
sistent cognitive impairment was frequent. Post-mortem and presbyophrenic behaviour were caused by a combina-
studies showed in these cases softening of ideational areas tion of senile and atherosclerotic changes. Burger-Prinz and
of cortex and white matter. Ball also suggested a laterality Jacob (1938), however, questioned the view that cyclothy-
effect (p. 582) in that right hemisphere strokes led more mic features were a necessary precondition. Bessière (1948)
often to dementia whereas left hemisphere ones caused per- claimed that presbyophrenia was a syndrome found in con-
plexity, apathy, unresponsiveness and a tendency to talk to ditions such as senile dementia, brain tumours, traumatic
oneself (p. 583). Following Luys, he believed that some of psychoses and confusional states. More recently, it was sug-
these symptoms resulted from damage to corpus striatum, gested that presbyophrenia was a subform of Alzheimer’s
insular sulci and temporal lobe. During Ball’s time atten- disease characterized by a severe atrophy of locus caeruleus
tion also shifted from white to red softening. Charcot (1881) (Berrios, 1985b).
wrote on cerebral haemorrhage (the new name for red soft- One of the features of presbyophrenia was confabula-
ening): ‘Having eliminated all these cases, we find ourselves tion. This complex symptom has not quite found a place
in the presence of a homogeneous group corresponding to in psychopathology (Berrios, 2000a). Two phenomena are
the commonest form of cerebral haemorrhage. This is, par included under the name confabulation. The first type
excellence, sanguineous apoplexy … as it attacks a great concerns ‘untrue’ utterances of subjects with memory
number of old people, I might call it senile haemorrhage’ impairment; often provoked or elicited by the interviewer,
(p. 267). these confabulations are accompanied by little conviction
and are believed by most clinicians to be caused by the
1.2.3.5 Presbyophrenia and confabulation (conscious or unconscious) need to ‘cover up’ for some
memory deficit. Researchers wanting to escape the ‘inten-
The word presbyophrenia was coined by Kahlbaum (1863) tionality’ dilemma have made use of additional factors
to name a subtype of the paraphrenias (insanities occurring such as presence of frontal lobe pathology, dysexecutive
during periods of biological change). Presbyophrenia was a syndrome, difficulty with the temporal dating of memo-
form of paraphrenia senilis characterized by amnesia, dis- ries leading to an inability temporally to string out mem-
orientation, delusional misidentification and confabulation. ory data etc.
Ignored for more than 30 years, the term reappeared in the The second type concerns confabulations with fantastic
work of Wernicke, Fischer and Kraepelin. content and great conviction as seen in subjects with func-
Wernicke’s classification of mental disorders was based tional psychoses and little or no memory deficit. Less is
on his theory of the three-partite relational structure con- said in the neuropsychological literature about this group
sciousness (outside world, body and self) (Lanczik, 1988). although (at least in the case of schizophrenics) it has been
Impairment of the link between consciousness and outside correlated with a bad performance on frontal lobe tests. It
world led to presbyophrenia, delirium tremens, Korsakoff’s is our belief that this group remains of crucial importance
psychosis and hallucinosis. Amongst the features of pres- to psychiatrists. Little is known about the epidemiology of
byophrenia, Wernicke included confabulations, disorienta- either type of confabulation.
tion, hyperactivity, euphoria and a fluctuating course; acute In clinical practice, these two ‘types’ can be found in
forms resolved without trace, chronic ones merged with combination. It remains unclear why so many patients with
senile dementia (Berrios, 1986). Korsakoff’s psychosis or frontal lobe disorder, in spite of the
In France, Rouby (1911) suggested that presbyophre- fact that they do meet the putative conditions for confabula-
nia was a final common pathway for cases suffering from tion (amnesia, frontal lobe damage, difficulty with the dating
Korsakoff’s psychosis, senile dementia or acute confusion. of memories etc.) do not confabulate. Furthermore, confab-
Truelle and Bessière (1911) proposed, in turn, that it might ulations have also been reported in subjects with lesions in
result from a toxic state caused by liver or kidney failure. the non-dominant hemisphere and in the thalamus.
Kraepelin (1910) lumped presbyophrenia together with the Under different disguises, the covering up or gap ­filling
senile and presenile insanities, and after comparing them hypothesis is still going strong. Although superficially
with Korsakoff’s patients reported that presbyophrenic ­plausible, it poses a serious conceptual problem in regards
patients were older, free from polyneuritis and history of to the issue of awareness of purpose: If full awareness is pre-
alcoholism, and showed hyperactivity and elevated mood. sumed, then it is difficult to differentiate confabulations
Ziehen (1911) wrote that ‘their marked memory impair- from lying; if no awareness is presumed then the semantics
ment contrasts with the relative sparing of thinking’. Oskar of the concept of purpose is severely stretched and confabu-
Fischer (1912) suggested that disseminated cerebral lesions lations cannot be differentiated from delusions.
were the essential anatomical substratum of presbyophrenia. The received view of confabulations also neglects the
During the 1930s, two new hypotheses emerged. clinical observation that confabulations (particularly pro-
Bostroem (1933) concluded that in clinical terms presbyo- voked ones!) do occur in dialogical situations: For exam-
phrenia resembled mania, suggesting an interplay between ple, the way in which the patient is asked questions may
Dementia: Historical overview 11

increases the probability of his producing a confabulation.


This suggests that the view that confabulations are a disor- 1.3 THE TWENTEITH CENTURY
der of a putative narrative function found in normal human
subjects must be taken seriously. It is hypothesized here that 1.3.1 ALZHEIMER’S DISEASE
this trait is normally distributed in the population. In the
absence of adequate epidemiological information, research Alzheimer’s disease has become a prototypical form of
efforts should be directed at mapping the distribution of dementia. From this point of view, a study of its origins
this narrative (or confabulatory) capacity in the community should throw light on the evolution of the concept of
at large. Only then it will be possible to understand the sig- dementia in general. The writings of Alzheimer, Fischer,
nificance of its disorders. In the long term, this approach Fuller, Lafora, Bonfiglio, Perusini, Ziveri, Kraepelin and
will prove more heuristic than unwarranted speculation others feel deceptively fresh, and this makes anachronis-
based on few anecdotal cases. tic reading inevitable. However, the psychiatry of the late
Reported in relation to clinical conditions other than nineteenth century is a foreign country: Concepts such as
memory deficit, confabulation-like behaviours can already dementia, neurone, neurofibril and plaque were then still in
be found in the clinical literature of the second half of the process of construction and meant different things to differ-
nineteenth century. Sully (1895) suggested that such behav- ent people. A discussion of these issues is beyond the scope
iours might be related to a psychological function whose of this chapter (for this see Berrios, 1990a).
role was filling gaps in the flow of our lives and explained
why ‘our image of the past is essentially one of an unbro-
ken series of conscious experiences’. Sully believed that
1.3.1.1 The neuropathology of dementia
this function also intervened when memory failed: ‘Just as before Alzheimer
the eye sees no gap in its field of vision ­corresponding to
Enquiries into the brain changes accompanying demen-
the ‘blind’ spot of the retina, but carries the impression
tia started during the 1830s but consisted in descriptions
over this area, so memory sees no lacuna in the past, but
of external appearance (Wilks, 1865). The first important
carries its image of conscious life over each of the forgot-
microscopic study was that of Marcé (1863) who described
ten spaces’ (p. 282).
cortical atrophy, enlarged ventricles and softening. The
Kraepelin (1886–1887) reported typical cases of con-
vascular origin of softening was soon ascertained (Parrot,
fabulation associated with inter alia general paraly-
1873), but the distinction between vascular and parenchy-
sis of the insane, melancholia and dementia; and later
mal factors had to await until the 1880s. From then on,
on suggested that pseudomemories could be a symptom
microscopic studies concentrated on cellular death, plaques
of paraphrenia (without cognitive impairment), and
and neurofibrils.
described a clinical variety called paraphrenia confabu-
lans (Kraepelin, 1919). Under schizophrenic akzessorischen
Gedachtnisstörungen, Bleuler (1911) discussed three related 1.3.1.2 Alzheimer and his disease
phenomena: Gedächnisillusionen (illusions or distortions
of memory), identifizierenden Erinnerungstäuschungen Alzheimer (1907) reported the case of a 51-year-old woman,
(memory falsifications based on identification), and with cognitive impairment, delusions, hallucinations, focal
Erinnerungshalluzinationen (memory hallucinations). Of symptoms, and whose brain showed plaques, tangles and
the former he wrote: ‘Memory illusions often constitute the arteriosclerotic changes. The existence of neurofibrils had
main material for the construction of delusions in para- been known for some time (DeFelipe and Jones, 1988; Barret,
noids. The entire previous life of the patient may be changed 1913); for example, that in senile dementia ‘the destruction
in his memory in terms of this complex’ (p. 115). On this of the neurofibrillae appears to be more extensive than
definition, it is difficult, on the basis of their intrinsic fea- in the brain of a paralytic subject’ (Bianchi 1906, p. 846).
tures, to distinguish illusion of memory from confabula- Fuller (1907) had remarked in June 1906 (i.e. 5 months
tion. Indeed, symptom-naming is determined by whether before Alzheimer’s report) on the presence of neurofibrillar
schizophrenia or ‘organic disorder’ is the associated disease. bundles in senile dementia (p. 450). Likewise, the associa-
Unsuccessfully, Bleuler (1911) tried to establish a differenti- tion of plaques with dementia was not a novelty: Beljahow
ation on the basis of mechanism: ‘Until now, and in contrast (1889) had reported them in 1887, and so had Redlich and
to the views of some authors, I have not observed confabu- Leri few years later (Simchowicz, 1924); in Prague, Fischer
lation as it appears in organic cases; e.g. memory halluci- (1907) gave an important paper in June 1907 pointing out
nations that fill in memory gaps, which at first appear at a that miliary necrosis could be considered as a marker of
(usually external) given moment and mostly adapt them- senile dementia.
selves to such an occasion (p. 117). Kleist (1960) described Nor was the syndrome described by Alzheimer new:
patients with ‘progressive confabulosis’ as ‘cheerful, expan- States of persistent cognitive impairment affecting the
sive and with little in the way of thought or speech disorder’ elderly, accompanied by delusions and hallucinations were
(p. 211); and Leonhard (1957) redescribed the condition as well known (Marcé, 1863; Krafft-Ebing, 1873; Crichton-
‘confabulatory euphoria’. Brown, 1874; Marie, 1906). As a leading neuropathologist,
12 Dementia

Alzheimer was aware of this work. Did he then mean to (fretta battezzate) as “Alzheimer’s disease”’ (p. 378). He went
describe a new disease? The answer is that it is most unlikely on to say that this state is only a variety of senile dementia.
he did, his only intention having been to point out that such Simchowicz (1924), who had worked with Alzheimer, wrote
a syndrome could occur in younger people (Alzheimer, ‘Alzheimer and Perusini did not know at the time that the
1911). This is confirmed by commentaries from those who plaques were typical of senile dementia [in general] and
worked for him: Perusini (1911) wrote that for Alzheimer believed that they might have discovered a new disease’
‘these morbid forms do not represent anything but atypical (p. 221). These views, from men who lived in Alzheimer’s
form of senile dementia’ (p. 143). and Kraepelin’s time, must be taken seriously (for a detail
discussion of these issues see Berrios 1990a).
Of late there has been an attempt to rewrite the history
1.3.1.3 Naming the disease of Alzheimer’s disease. After having been lost for years, the
Kraepelin (1910) coined the term in the eighth edition of case notes of Auguste D., the first patient with Alzheimer’s
his handbook: At end of the section on ‘senile dementia’ disease were found in the late 1990s (Maurer and Maurer,
he wrote: ‘The autopsy reveals, according to Alzheimer’s 1998). As mentioned above the original report by Alzheimer
description, changes that represent the most serious form (1907) clearly stated that Auguste suffered from severe con-
of senile dementia … the Drusen were numerous and fusion, delusions, hallucinations, focal symptoms and on
almost one third of the cortical cells had died off. In their post-mortem her brain showed plaques, tangles and most
place instead we found peculiar deeply stained fibrillary importantly arteriosclerotic changes. As if to confirm
bundles that were closely packed to one another, and the ontology of Alzheimer’s disease, Graeber et al. (1998)
seemed to be remnants of degenerated cell bodies … The reported that in tissue sections belonging to this case they
clinical interpretation of this Alzheimer’s disease is still confirmed the presence of neurofibrillary tangles and amy-
confused. Whilst the anatomical findings suggest that loid plaques. Most interestingly, and given that vascular
we are dealing with a particularly serious form of senile changes are currently not supposed to be a diagnostic crite-
dementia, the fact that this disease sometimes starts rion, these authors decided to correct Alzheimer’s own his-
already around the age of 40 does not allow this supposi- topathological study and state that Auguste’s brain showed
tion. In such cases we should at least assume a “senium no arteriosclerotic lesions. Furthermore, they report that
praecox” if not perhaps a more or less age-independent the apolipoprotein E (ApoE) genotype of Auguste was ε3/
unique disease process’. He went on to call it Alzheimersche ε3, ‘indicating that mutational screening of the tissue is
Krankheit (p. 356). feasible’.
On the basis of these findings, can one say that back in
1910 Kraepelin was after all right in claiming that Alzheimer
1.3.1.4 Reception of the disease had ‘discovered’ a new disease? The answer has to be that
one must judge his decision in terms of what Kraepelin
Alzheimer (1911) showed surprise at Kraepelin’s interpreta- knew at the time and of the academic pressure he was under.
tion, and always referred to his ‘disease’ as Erkrankungen He knew what the Alzheimer’s report stated in 1907, what
(in the medical language of the 1900’s a term softer than the latter might have verbally added, and upon Kraepelin’s
Krankheit, term used by Kraepelin). Others also expressed perusal of Auguste’s case notes, which were requested by
doubts. Fuller (1912), whose contribution to this field him from Frankfurt (indeed this is the reason why they
has been sadly neglected, asked ‘why a special clinical were lost for such a long time). In clinical terms, Auguste
­designation—Alzheimer’s disease—since, after all, they are was not a ‘typical’ case. At 51 she had delusions, halluci-
but part of a general disorder’ (p. 26). Hakkboutsch and nations and Alzheimer’s reported arteriosclerotic changes.
Geier (1913), in Russia, saw it as a variety of the involu- None of these features is mentioned in Kraepelin’s original
tion psychosis. Simchowicz (1911) considered ‘Alzheimer’s claim. The question is, why?
disease’ as only a severe form of senile dementia. Ziehen It is also interesting that the so-called ‘second case’ of
(1911) did not mention the disease in his major review of Alzheimer’s (Graeber et al., 1997), Johann F., reported
senile dementia. In a meeting of the New York Neurological in 1911 is now considered to have suffered a plaques-only
Society, Ramsay Hunt (Lambert, 1916) asked Lambert, the form of Alzheimer’s disease. The same authors suggest that
presenter of a case of ‘Alzheimer’s disease’ that ‘he would knowledge of this case may have encouraged Kraepelin
like to understand clearly whether he made any distinction to report the discovery by Alzheimer’s of a new disease.
between the so-called Alzheimer’s disease and senile demen- Unfortunately, there is no evidence that Kraepelin knew of
tia other than … in degree and point of age’. Lambert agreed this case when he was writing the relevant section of the
suggesting that, as far as he was concerned, the underlying eighth edition of his Lehrbuch. More to the point is that he
pathological mechanisms were the same (Lambert, 1916). knew of Perusini’s review (1909) as he himself had asked the
Lugaro (1916) wrote: ‘For a while it was believed that a cer- young Italian assistant to collect four cases. In addition to
tain agglutinative disorder of the neurofibril could be con- Auguste D (which includes more information than that pro-
sidered as the main “marker” (contrassegno) of a presenile vided by Alzheimer in 1907), Perusini reviewed the cases
form [of senile dementia], which was “hurriedly baptized” of Mr. R.M., a 45-year-old basket maker who had epileptic
Dementia: Historical overview 13

seizures, Mrs. B.A. a 65-year-old who had marked clinical During the same period, Liepmann, Stransky and
features of myxoedema and Schl. L, a 63-year-old who had Spielmeyer had described similar cases with aphasia
suffered from syphilis since 1870, and was Romberg-positive and circumscribed cerebral atrophy (Mansvelt, 1954); so
and had a pupillary syndrome and heard voices. Since these much so that Urechia and Mihalescu felt tempted to name
are likely to have been the cases on which Kraepelin based the syndrome Spielmeyer’s disease (Caron, 1934). This did
his decision to construct the new disease, it would be inter- not catch on, however, and in two classical papers on what
esting that the neuropathology and neurogenetics of the he coined the term Pick’s disease, Carl Schneider (1927,
other three cases (Auguste’s has already been done) is also 1929) constructed a new profile for this condition by sug-
investigated. gesting that it evolved in three stages – the first with a
disturbance of judgement and behaviour, the second with
1.3.2 PICK’S DISEASE AND THE FRONTAL localized symptoms (e.g. speech) and the third with gen-
DEMENTIAS eralized dementia. He recognized rapid and slow forms,
the former with an akinetic and aphasic subtypes and a
Dementias believed to be related to frontal lobe pathol- malignant course, and the latter with a predominance
ogy have once again become fashionable, and authors of plaques (probably indistinguishable from Alzheimer’s
often invoke the name of Arnold Pick (Niery et al., disease).
1988). However, when the great Prague neuropsychiatrist
described the syndrome named after him, all he wanted 1.3.3 DEMENTIA SINCE WORLD WAR I
was to draw attention to a form of localized (as opposed
to diffuse) atrophy of the temporal lobe (Pick, 1892). This By the 1920s, the description and classification of the
alteration was to give rise to a dysfunction of language and dementias had notably changed. This is well illustrated
praxis, and be susceptible to diagnosis during life. Pick by comparing the first (1869) and fifth (1918) editions
believed that lobar atrophies constituted a stage in the evo- of the Official Nomenclature of Diseases of the Royal
lution of the senile dementias. College of Physicians. In 1869, dementia (Condition 107)
The story starts, as it should, before Pick. Louis Pierre is classified as acute and chronic and is grouped with
Gratiolet (1854) renamed the cerebral lobes after the respec- paralysis of the insane (Condition 108), amentia (idiocy)
tive sections of overlying skull: Thus anterior became frontal (Condition 109) and insipientia (imbecility) (Condition
lobe. He made no assumption as to the function of the ‘ante- 110). In 1918, dementia is grouped with dementia prae-
rior extremity of the cerebral hemisphere’. Phrenologists, cox (also called schizophrenia) and divided into primary
however, did and related reflective and perceptive functions and secondary (the latter originating from organic cere-
(qualitatively defined) to the forehead (Anonymous, 1832) bral disease other than syphilis, arterial disease, senile,
(on the science of Phrenology: See Combe, 1873; Lanteri- epilepsy and injury).
Laura, 1970). Modular assumptions (i.e. a one-to-one cor- Continental views are well illustrated by the popular
relation between mental function and brain site) involving Lehrbuch der Psychiatrie written by Eugen Bleuler whose
the frontal lobes started only during the 1860s, following English, French and Spanish translations did a great deal
reports on dysfunction of language in lesions of the fron- to uniformize psychiatric nomenclature and classifica-
tal lobes (Broca, 1861; Henderson, 1986). These claims ran tion in both Europe and the United States. Bleuler (1920)
parallel to those of Jackson’s that the cerebral cortex was listed four main categories: dementia paralytica, prese-
the general seat of personality and mind (Jackson, 1894). nile and senile dementia, arteriosclerotic dementia and
Meynert (1885) believed that ‘the frontal lobes reach a high presbyophrenia. ‘Vesanic dementia’, the category used to
state of development in man’ but still defined mental disor- name a final common pathway for all chronic psychoses
ders as diseases of the forebrain (by which he meant ‘pros- was all but abandoned. By the end of the Second World
encephalon’ or human brain as a whole). War, Presbyophrenia also was sounded its dead knell.
Neither in the first (case of focal senile atrophy and Lastly, Bini (1948) published his great book on Le Demenze
aphasia in a man of 71; Pick, 1892) nor in the second case Presenili where he reported 37 cases studied in detail and
report (a woman of 59 with generalized cortical atrophy, offered the classificatory scheme for the dementias that has
particularly of the left hemisphere; Pick, 1901) did Pick lasted until today.
actually inculpate the frontal lobes as responsible for the The stage was being prepared for Roth’s (1955) classi-
symptomatology. A link with the frontal lobes was only cal paper on The Natural History of Mental Disorder in Old
made in his fourth case report (a 60-year-old man with Age where he explored the clinical context in which the
‘bilateral frontal atrophy’; Pick, 1906). Which of these dementias occurred and concluded that the results: ‘Suggest
cases should, therefore, be considered as the first with that affective psychosis, late paraphrenia and acute confu-
Pick’s disease? At the time, in fact, no one thought that sion are distinct from the two main causes of progressive
Pick had described a new disease; Barrett (1913) con- dementia in old age; senile and arteriosclerotic psychosis.
sidered the two first cases of Pick’s as atypical forms of They also provide some support for the validity of the clini-
Alzheimer’s disease, and Ziehen (1911) did not see any- cal distinction drawn between senile and arteriosclerotic
thing special in them. psychosis’ (p. 295). This paper offered not only a working
14 Dementia

classification of the mental disorders of all age but also a Thus far, biological therapies based on the neuroscien-
vindication of the real place of the vesanic dementias and in tific narrative of dementia have been less successful than
doing so set the basis for a new professional specialism, the expected. This has led to some broadening of therapeutic
psychiatry of old age. approach, and the earlier emphasis on a pure cognitive par-
adigm has given way to interest in behavioural failures such
as self and personality deterioration, and emotional and
volitional involution.
1.4 CONCLUSIONS As a collateral issue, promises of finding a ‘cure’ for
dementia have generated some speculation on the eventual
The history of the word dementia must not be confused finding of an elixir of eternal youth. This has unavoidably
with that of the concepts or behaviours involved. By the led to asking the deontological question about the duties of
year 1800, two definitions of dementia were recognized the generation that has discovered such elixir to the follow-
and both had psychosocial incompetence as their central ing ones.
concept: In addition to cognitive impairment, the clinical So far, the most hopeful response to the crisis created by
definition included other symptoms such as delusions and the need to care for the growing number of persons affected
hallucinations. Irreversibility and old age were not features with dementia has been the positive and creative attitude of
of the condition, and in general dementia was considered to carers, both professional and lay, and the search for solutions
be a terminal state to all sorts of mental, neurological and in fields beyond neuroscience. Their good will, patience and
physical conditions. creativity needs to be encouraged and supported by all, and
The adoption of the anatomo-clinical model of disease by mostly by the body politics.
nineteenth-century alienists changed this. Questions were
asked then as to the neuropathological basis of dementia REFERENCES
and this, in turn, led to readjustments in its clinical descrip-
tion. The history of dementia during the nineteenth century Alzheimer, A. (1907). Über eine eigenartige Erkrankung
is, therefore, the history of its gradual attrition. Stuporous der Hirnrinde. Allgemeine Zeitschrift für
states (then called acute dementia), vesanic dementias and Psychiatrie und Psychisch-Gerichtlich Medizine,
localized memory impairments, were gradually reclassified, 64: 146–148.
and by 1900 the cognitive paradigm, i.e. the view that the Alzheimer, A. (1911). Über eigenartige Krankheitsfälle
essential feature of dementia was intellectual impairment, des späteren Alters. Zeitschrift für die gesamte
was established (Berrios, 1989). Neurologie und Psychiatrie, 4: 356–385.
From then on, efforts were made to explain other symp- Anderson, E.W., Threthowan, W.H. and Kenna, J.C. (1959).
toms such as hallucinations, delusions and mood and An experimental investigation of simulation and
behavioural disorders as epiphenomena, and as unrelated pseudodementia. Acta Psychiatrica et Neurologica
to whatever the central mechanism of dementia was. There Scandinavica, 34 (Suppl. 132): S1.
has also been a fluctuating acceptance of the parenchymal Anonymous (1832). An exposure of the unphilosophical
and vascular hypotheses, the latter leading to the descrip- and unchristian expedients adopted by antiphre-
tion of arteriosclerotic dementia. The separation of the ves- nologists, for the purpose of obstructing the moral
anic dementias and of the amnestic syndromes led to the tendencies of phrenology. A review of John Wayte’s
realisation that age and ageing mechanisms were impor- book. The Phrenological Journal and Miscellany, 7:
tant, and by 1900 senile dementia became the prototype of 615–622.
the dementias; by 1970, Alzheimer’s disease had become the Baillarger, J. (1883). Sur la théorie de la paralysie générale.
flagship of the new approach. During the last few years, the Annales Médico-Psychologiques, 35 (18–52):
cognitive paradigm has become an obstacle, and a gradual 191–218.
re-expansion of the symptomatology of dementia is fortu- Baillarger, J. (1889). Doit-on dans la Classification des
nately taking place (Berrios, 1989; 1990b). Maladies Mentales Assigner une Place á Part aux
It is against this clinical and classificatory context that Pseudo-paralysies Générales? Annales Médico-
the mental disorders in old age in general, and the demen- Psychologiques, 41: 521–525.
tias in particular have continued their development. The Ball, B. and Chambard, E. (1881). Démence. In
latter were different disorders, had a different outcome A. Dechambre and Lereboullet, L. (eds.), Dictionnaire
and hence had a different aetiology. As the neuroscien- Encyclopédique des Sciences Médicales. Paris,
tific narratives became predominant, classifications of the France: Masson, pp. 559–605.
dementias based on biological markers gradually replaced Barrett, A.M. (1913). Presenile, arteriosclerotic and senile
older ones based on clinical phenomenology. This process disorders of the brain and cord. In W.A. White and
is going on and from the perspective of the history and Jelliffe, S.A. (eds.), The Modern Treatment of Nervous
epistemology of dementia it is difficult to predict where it and Mental Diseases. London, England: Kimpton,
will go. pp. 675–709.
Dementia: Historical overview 15

Bayle, A.L.J. (1822). Recherches sur les Maladies Mentales. Bini, L. (1948). Le Demenze Presenile. Roma, Italy: Edizioni
Paris, France: Thése de Médecine. Italiane.
Bayle, A.L.J. (1826). Traité Des Maladies du Cerveau. Paris, Blancard, S. (1726). The Physical Dictionary Wherein
France: Gabon et Compagnie. the Terms of Anatomy, the Names and Causes of
Beljahow, S. (1889). Pathological changes in the brain Diseases, Chirurgical Instruments, and Their Use, Are
in dementia senilis. Journal of Mental Science, Accurately Described. London, England: John and
35: 261–262. Benjamin Sprint.
Bercherie, P. (1980). Les Fondements de la Clinique. Paris, Bleuler, E. (1911). Dementia Praecox. Leipzig, Germany:
France: La Bibliothéque ’d’Ornicar. Deuticke. (English Translation: Dementia Praecox,
Berrios, G.E. (1985a). ‘Depressive pseudodementia’ or ‘mel- 1950, New York, NY: International Universities Press).
ancholic dementia’: A 19th century view. Journal of Bleuler, E. (1920). Lehrbuch der Psychiatrie, Third Edition.
Neurology, Neurosurgery and Psychiatry, 48: 393–400. Heidelberg: Springer.
Berrios, G.E. (1985b). Presbyophrenia: Clinical aspects. Boissier de Sauvages, F. (1771). Nosologie Methodique,
British Journal of Psychiatry, 147: 76–79. dans laquelle les Maladies sont rangées par Classes,
Berrios, G.E. (1986). Presbyophrenia: The rise and fall of a suivant le Systême de Sydenham, & l’order des
concept. Psychological Medicine, 16: 267–275. Botanistes. Paris, France: Hérissant le Fils.
Berrios, G.E. (1987). History of the functional psychoses. Boller, F. (2008). History of dementia. In C. Duyckaerts
British Medical Bulletin, 43: 484–498. and Litvan, I. (eds.), Dementia: Handbook of Clinical
Berrios, G.E. (1989). Non-cognitive symptoms and Neurology, Vol. 89, Third Series. New York: Elsevier,
the diagnosis of dementia. Historical and ­clinical pp. 3–13.
aspects. British Journal of Psychiatry, 154 Bostroem, A. (1933). Über Presbyophrenie. Archiv für
(Suppl. 4): S11–S16. Psychiatrie und Nervenkrankenheiten, 99: 339–354.
Berrios, G.E. (1990a). Alzheimer’s disease: A conceptual Broca, P. (1861). Perte de la Parole, Ramollissement
history. International Journal of Geriatric Psychiatry, 5: Chronique et Destruction Partielle du Lobe Anterieur
355–365. Gauche du Cerveau. Bulletin de la Société de
Berrios, G.E. (1990b). Memory and the cognitive paradigm Anthropologie Paris, 2: 235–238.
of dementia during the 19th century: A conceptual Bucknill, J.C. and Tuke, D.H. (1958). A Manual of
history. In R. Murray and Turner, T. (eds.), Lectures on Psychological Medicine. London, England: John
the History of Psychiatry. London, England: Gaskell, Churchill.
pp. 194–211. Bulbena, A. and Berrios, G.E. (1986). Pseudodementia:
Berrios, G.E. (1991). Affective disorders in old age: A Facts and figures. British Journal of Psychiatry, 148:
conceptual history. International Journal of Geriatric 87–94.
Psychiatry, 6: 337–346. Burger-Prinz, H. and Jacob, H. (1938). Anatomische und
Berrios, G.E. (1994). The psychiatry of old age: A con- klinische Studien zur senilen Demenz. Zeitschrift
ceptual history. In J. Copeland, Abou-Saleh, M. für die gesamte Neurologie und Psychiatrie, 161:
and Blazer, D. (eds.), The Principles and Practice 538–543.
of Geriatric Psychiatry. Chichester, England: Wiley, Calmeil, L.F. (1835). Démence. In Dictionnaire de
pp. 11–16. Médicine on Repertoire General des Sciences
Berrios, G.E. (1996). The classification of mental disorders: Médicales, Second Edition. Paris, France: Bechet,
Part III. History of Psychiatry, 7: 167–182. pp. 70–85.
Berrios, G.E. (2000a). Confabulations. In G.E. Berrios and Caron, M. (1934). Etude Clinique de la Maladie De Pick.
Hodges, J.R. (eds.), Memory Disorders in Clinical Paris, France: Vigot Fréres.
Practice. Cambridge: Cambridge University Press, Charcot, J.M. (1881). Clinical Lectures on Senile and
pp. 348–368. Chronic Diseases. London, England: The New
Berrios, G.E. (2011). Convergences that are no more. Sydenham Society.
History of Psychiatry, 22: 133–136. Cicero, M.T. (1969). Tusculanan Disputations, Translated by
Berrios, G.E. and Freeman, H. (eds.) (1991). Alzheimer and J.E. King. Vol. XVIII. Cambridge, MA: Loeb Classical
the Dementias. London, England: Royal Society of Library.
Medicine. Code Napoléon (1808). Edition Originale et Seule
Berrios, G.E. and Luque, R. (1995). Cotard’s delusion or Officielle. Paris, France: L’Imprimerie Impériale.
syndrome? A conceptual history. Comprehensive Combe, G. (1873). Elements of Phrenology. Edinburgh,
Psychiatry, 36: 218–223. Scotland: MacLachlan and Stewart.
Bessière, R. (1948). La Presbyophrénie. L’Encéphale, 37: Cotard, J. (1882). Du délire des negations. Archives de
313–342. Neurologie, 4: 152–170; 282–296.
Bianchi, L. (1906). A Textbook of Psychiatry. London, Crichton-Browne, J. (1874). Senile dementia. British
England: Baillière, Tindall and Cox. Medical Journal, 1: 601–603; 640–643.
16 Dementia

DeFelipe, J. and Jones, E.G. (eds.) (1988). Cajal on the Guislain, J. (1852). Leçons Orales sur les Phrénopathies.
Cerebral Cortex: An Annotated Translation of the Paris, France: Gand, L. Hebbelynck.
Complete Writings. Oxford: Oxford University Press. Hakkébousch, B.M. and Geier, T.A. (1913). De la Maladie
Diderot, D. and d’Alembert, J.L.R. (eds.) (1765). d’Alzheimer. Annales Médico-psychologiques,
Encyclopédie ou Dictionnaire Raisonné des 71: 358.
Sciences, des Arts et des Métieres, par une Hare, E. (1959). The origin and spread of Dementia
Societé de Gens de Lettres, Vol. 4, A. Paris, Paralytica. Journal of Mental Science, 105: 594–626.
France: Briasson, David, Le Breton, Durand, Heinroth, J.C. (1975). Textbook of Disturbances of Mental
pp. 807–808. Life, Baltimore, MD: Johns Hopkins University Press.
Dumas, G. (1894). Les États Intellectuels dans la Henderson, V.H. (1986). Paul Broca’s less heralded
Mélancolie. Paris, France: Alcan. ­contributions to aphasia research. Historical per-
Durand-Fardel, M. (1843). Traité du Ramollissement du spective and contemporary relevance. Archives of
Cerveau. Paris, France: Baillière. Neurology, 43: 609–612.
Esquirol, E. (1805). Des Passions. Paris, France: Didot Jackson, J.H. (1875). A lecture on softening of the brain.
Jeune. Lancet, 106 (2714): 335–339.
Esquirol, E. (1814). Démence. In Dictionaire des Sciences Jackson, J.H. (1894). The factors of insanities. Medical
Médicales, par une Societé de Médicins et de Press and Circular, II: 615–625.
Chirurgiens, Paris, France: Panckouke, pp. 280–293. Kahlbaum, K.L. (1863). Die Gruppierung der psychischen
Esquirol, E. (1838). Des Maladies Mentales, Paris, France: Krankheiten, Danzig: A. W. Kafemann.
Baillière. Kiloh, L.G. (1961). Pseudo-dementia. Acta Psychiatrica
Feuchtersleben, E. (1847). The Principles of Medical Scandinavica, 37: 336–351.
Psychology, Translated by H.E. Lloyds and Babington, Kleist, K. (1960). Schizophrenic symptoms and cerebral
B.G. London, England: Sydenham Society. pathology. Journal of Mental Science, 106: 246–255.
Fischer, O. (1907). Miliare Nekrosen mit drusigen Kraepelin, E. (1886–1887). Über Erinnerungsfälschungen.
Wucherungen der Neurofibrillen, eine regelmaessege Archiv für Psychiatrie und Nervenkrankheiten, 17:
Verandaerung der Hirnrinde bei seniler Demenz. 830–843; 18: 199–239, 395–436.
Monatsschrift für Psychiatrie und Neurologie, 22: Kraepelin, E. (1910). Psychiatrie: Ein Lehrbuch für
361–372. Studierende und Ärzte, Leipzig, Germany: Johann
Fischer, O. (1912). Ein weiterer Beitrag zur Klinik und Ambrosius Barth.
Pathologie der presbyophrenen Demenz. Zeitschrift für Kraepelin, E. (1919). Dementia Praecox, Translated by R.M.
die gesamte Neurologie und Psychiatrie, 12: 99–135. Barclay and Robertson, G.M. Edinburgh: Livingstone.
Fournier, A. (1875). Syphilis du Cerveau. Paris, France: Krafft-Ebing, R. (1873). De la Démence Sénile. Annales
Baillière. Médico-psychologiques, 34: 306–307.
Fuller, S.C. (1907). A study of the neurofibrils in dementia Lafora, G.R. (1935). Sobre la Presbiofrenia sin Confa­
paralytica, dementia senilis, chronic alcoholism, cere- bulaciones. Archivos de Neurobiologia, 15: 179–211.
bral lues and microcephalic idiocy. American Journal Lambert, C.I. (1916). The clinical and anatomical features of
of Insanity, 63: 415–468. Alzheimer’s disease. Journal of Mental and Nervous
Fuller, S.C. (1912). Alzheimer’s disease (senium praecox): Disease, 44: 169–170.
The report of a case and review of published cases. Lanczik, M. (1988). Der Breslauer Psychiater Carl Wernicke.
Journal of Nervous and Mental Disease, 39: 440–455; Sigmaringen: Thorbecke.
536–557. Lanteri-Laura, G. (1970). Histoire de la Phrenologie. Paris,
Golinski, J. (1998). Making Natural Knowledge. Cambridge, France: Presses Universitaires de France.
England: Cambridge University Press. Leonhard, K. (1957). Aufteilung der endogenen Psychosen.
Graeber, M.B., Kosel, S., Egensperger, R. et al. (1997). Berlin, Germany: Akademie-Verlag.
Rediscovery of the case described by Alois Alzheimer Lewis, C.T. and Short, C. (1969). A Latin Dictionary (First
in 1911: Historical, histological and molecular genetic Edition, 1879). Oxford: At the Clarendon Press.
analysis. Neurogenetics, 1: 73–80. Lucretius (1975). De Rerum Natura, Translation by W.H.D.
Graeber, M.B., Kosel, S., Grasbon-Frodl, E. et al. (1998). Rouse. Cambridge: Harvard University Press.
Histopathology and APOE phenotype of the first Lugaro, E. (1916). La psichiatria tedesca nella storia e
Alzheimer’s disease patient. Neurogenetics, 1: nell’attualita. Rivista di Patologia Nervosa e Mentale,
223–238. 21: 337–386.
Gratiolet, L.P. (1854). Mémoires sur les Plis Cérébraux de Madden, J.J., Luhan, J.A., Kaplan L.A. et al. (1952). Non-
l’Homme et des Primates. Paris, France: Bertrand. dementing psychoses in older persons. Journal of the
Griesinger, W. (1861/1967). Mental Pathology and American Medical Association, 150: 1567–1570.
Therapeutics, Translated by C.L. Robertson and Mairet, A. (1883). De la Démence mélancolique. Paris,
Rutherford, J. London, England: Sydenham Society. France: Masson.
Dementia: Historical overview 17

Mansvelt, J. (1954). Pick’s Disease. Enchede: van der Loeff. Pick, A. (1901). Senile Hirnatrophie als Grundlage von
Marcé, L.V. (1863). Recherches cliniques et anatomo- Herderscheinungen. Wiener Clinische Wochenschrift,
Pathologiques sur la démence senile et sur les 14: 403–404.
différences qui la separent de la paralysie générale. Pick, A. (1906). Über einen weiterer Symptomenkomplex
Gazette Médicale de Paris, 34: 433–435; 467–469; im Rahmen der Dementia senilis, bedingt durch
497–502; 631–632; 761–764; 797–798; 831–833; umschriebene sträkere Hirnatrophie (gemische
855–858. Apraxie). Monatschrift für Psychiatrie und Neurologie,
Marie, A. (1906). La Démence. Paris, France: Doing. 19: 97–108.
Maurer, K. and Maurer, U. (1998). Alzheimer: Das Leben Pick, D. (1989). Faces of Degeneration. Cambridge:
eines Arztes und die Karriere einer Krankheiten. Cambridge University Press.
Munich, Germany: Piper. Pinel, Ph. (1806). A Treatise on Insanity, Translated by D.D.
Meynert, T. (1885). Psychiatry. A Clinical Treatise on Davis. Sheffield, England: Cadell and Davies.
Diseases of the Fore-brain, Translated by B. Sachs. Pinel, Ph.(1818). Nosographie Philosophique, Sixth Edition.
New York, NY: Putnam. Paris, France: Brosson (first published: 1798).
Meynert, T. (1890). Amentia. In Klinische Vorlesungen über Prichard, J.C. (1835). A Treatise on Insanity. London,
Psychiatrie auf Wissenschaftlichen Grundlagen, für England: Sherwood, Gilbert and Piper.
Studierende und Ärzte, Juristen und Psychologen. Rey, A. (1995). Dictionnaire Historique de la Langue
Vienna, Austria: Braumüller. Française, Vol. 2. Paris, France: Dictionnaire Le
Morel, B.A. (1857). Traité des Dégénérescences Physiques Robert.
Intellectuelles et Morales de l’Espèce Humaine. Paris, Rostan, L. (1823). Recherches sur le Ramollissement du
France: Baillière. Cerveau, Second Edition. Paris, France: Bechet.
Morel, B.A. (1860). Traité des Maladies Mentales. Paris, Roth, M. (1955). The natural history of mental disorder in
France: Masson. old age. British Journal of Psychiatry, 101: 281–301.
Niery, D., Snowden, J.S., Northen, B. and Goulding, Rouby, J. (1911). Contribution á l’étude de la Presbyo­
P. (1988). Dementia of frontal lobe type. Journal phrénie, Thèse de Médicine, Paris, France: E. Nourris.
of Neurology, Neurosurgery and Psychiatry, Royal College of Physicians (1869). The Nomenclature of
51: 353–361. Diseases, First Edition. London, England: Golbourn.
North, H.M. and Bostock, F. (1925). Arteriosclerosis Royal College of Physicians (1918). The Nomenclature
and mental disease. Journal of Mental Science, of Diseases, Fifth Edition. London, England: His
71: 600–601. Majesty’s Stationery Office.
Olah, G. (1910). Was kann man heute unter Schneider, C. (1927). Über Picksche Krankheit. Monatschrift
Arteriosklerotischen Psychosen verstehen? Psychiatrie für Psychiatrie und Neurologie, 65: 230–275.
Neurologie Wochenschrift, 52: 532–533. Schneider, C. (1929). Weitere Beiträge zur Lehre von der
Parrot, J. (1873). Cerveau. VIII Ramollissement. In Pickschen Krankheit. Zeitschrift für die gesamte
A. Dechambre and L. Lereboullet (eds.), Dictionnaire Neurologie und Psychiatrie, 120: 340–384.
Encyclopédique des Sciences Médicales, Vol. 14. Simchowicz, T. (1911). Histologische Studien über die
Paris, France: Mason and Asselin, pp. 400–431. Senile Demenz. Histologische und Histopatholog­
Perry, R., McKeith, I. and Perry, E. (eds.) (1996). Dementia ischen Arbeiten über der Grosshirnrinde, 4: 267–444.
with Lewy Bodies. Cambridge: Cambridge University Simchowicz, T. (1924). Sur la signification des plaques
Press. séniles et sur la formule sénile de l’écorce cérébrale.
Perusini, G. (1909). Über klinish und histologisch eigen- Revue Neurologique, 31: 221–227.
artige psychische Erkrankungen des späteren Sobrino, F. (1791). Aumentado O Nuevo Diccionario De
Lebensalters. In F. Nissl and Alzheimer, A. (eds.), Las Lenguas Española, Francesa Y Latina. Leon de
Histologische und histopatologische Arbeiten, Francia: J.B. Delamolliere.
Vol 3. pp. 297–351. Southard, E.E. (1910). Anatomical findings in ‘senile
Perusini, G. (1911). Sul valore nosografico di alcuni rep- dementia’: A diagnostic study bearing especially on
erti istopatologici caratteristiche per la senilitá. the group of cerebral atrophies. American Journal of
Rivista Italiana di Neuropatologia, Psichiatria ed Insanity, 61: 673–708.
Elettroterapia, 4: 193–213. Sully, J. (1895). Illusions. A Psychological Study, Fifth
Pick, A. (1892). Über die Beziehungen der senilen Edition. London, England: Kegan Paul.
Hirnatrophie zur Aphasie. Prager Medicinische Truelle, V. and Bessière, R. (1911). Recherches sur la
Wochenschrift, 17: 165–167. (Translation: Berrios G.E. Presbyophrénie. L’Encéphale, 6: 505–520.
and Girling D.M. (1994) Introduction to and transla- Walton, G.L. (1912). Arteriosclerosis probably not an
tion of on the relationship between senile cerebral important factor in the etiology and prognosis of
atrophy and aphasia by Arnold Pick. History of involution psychoses. Boston Medical and Surgical
Psychiatry, 5: 539–549). Journal, 167: 834–836.
18 Dementia

Wilks, S. (1865). Clinical notes on atrophy of the brain. Ziehen T. (1911). Les Démences. In A. Marie A (ed.), Traité
Journal of Mental Science, 10: 381–392. International de Psychologie Pathologique, Vol. 2.
Willis T. (1684). Practice of Physick, Translated by Paris, France: Alcan, pp. 281–381.
S. Pordage. London, England: T. Dring, C. Harper Zilboorg G. (1941). A History of Medical Psychology. New
and J. Leigh, pp. 209–214. York, NY: Norton.
2
The lived experience of dementia

SHIRLEY NUROCK

was not going to turn out as we had envisaged. I felt desper-


2.1 OUR STORY ately sad for him and initially wondered if it could be a mis-
take, that he was being forgetful on purpose and would soon
‘What do you mean, Alzheimer’s disease? Surely revert to his usual charming, distracted self. Denial soon
he’s too young!’ turned to alarm as his memory deteriorated noticeably,
then, selfishly, to anger. Never voiced, but I was angry with
Apparently not. With these words (over the telephone) our him for abandoning us in this way. What would become of
lives were turned upside-down. My handsome, caring hus- us? What would become of me? In my forties, my life now
band Leonard, a GP here in London, in his fifties, had been on hold.
diagnosed with Alzheimer’s disease (AD), our three chil- As we cried ourselves to sleep I embarked on that roll-
dren then in their early teens. ercoaster of emotions that was to last over 15 years. I think
Yes, he had been more than usually forgetful over the last
he felt humiliated and, aware that his mental faculties were
couple of years, infuriating me sometimes by mislaying keys
failing, became increasingly frustrated and would mutter
or turning up at the wrong time or wrong place, but onset was
about throwing himself off the bridge. I genuinely believed
insidious and I was pre-occupied with my work, our hectic
he had a period of good quality life and love ahead and could
lifestyle, the children and their decisions about schools, uni-
always reassure him that he had much to live for and three
versities, and choice of subjects and unthinkingly accepted
affectionate children nearing adulthood. I had no choice but
his explanation of merely being stressed at work. This was
to give up my part-time work painting children’s pictures,
the late l980s and a new GP contract was coming into place
that involved computerization and appointment of practice just at a time when numbers of commissions were soaring.
managers; not concepts he was familiar with. Subsequently I recall panicking and frantically rushing
The neurologist advised he stop work immediately, stop round reorganizing our finances, signing new Wills, Powers
driving, just stay at home and keep as healthy as possible of Attorney and arguing with the Pensions Department
and don’t move house, that would be the worst thing to do; about entitlement.
‘Oh, and join the Alzheimer’s Society. That was it. He did. Imparting the news to friends and family was a relief, no
Or rather we did. Although aware that this was not good more making excuses for Leonard’s lapses of memory and
news, I was unfamiliar with the diagnosis and he seemed increasing forgetfulness over names and faces. Most just
not to grasp its wider implications. looked bewildered and sadly many of our medical friends
Given that his practice list was over 12,000, his assertion just vanished. Were they afraid it was contagious, sorry they
that only one or two of his patients may had AD seemed an couldn’t make him better, or what? When effusive letters of
underestimate. We lived in a five-storey narrow town house condolence came from them some 15 years later, I was not in
and the advice about not moving ultimately turned out to be forgiving mood. A brave few turned out to be true friends,
one of the factors that led to earlier institutionalization than visiting no matter how distressing they found our situation.
should have been the case. The children accepted what I told them and although
I laid hands on as many books as I could (this was before always gentle with their father, I was aware that they
the age of universal Internet access) and slowly the full hor- retreated more into their own lives and friends. It was
ror of the prognosis began to sink in; that this was a termi- particularly hard for the two youngest, still at school and
nal illness, possibly I was going to be looking after him for living at home. The youngest, our son, shy at the best of
years, our happy and charmed life with our loving family times, was consumed with embarrassment when I would
19
20 Dementia

turn up at school concerts or parents’ evenings, husband- found by the police were traumatic. For two years I slept on
in-tow because there was nowhere else to leave him. If we a mattress on the floor by his side of our bed so that when
were out as a family in public places our son had to help he got up to wander in the middle of the night he tripped
his father to the toilet. I appreciate how hard it was for him. and woke me. Thus I could prevent him walking out of the
Embarrassment became a feature of our lives and as one bedroom door and falling down the stairs. Locking doors,
by one the children left home for pastures new and friends removing electric plugs, hiding basin plugs, turning off light
deserted us, loneliness became another. Even my husband switches, night-lights everywhere, flushing the toilet every
noticed the latter and one day, not having spoken for weeks, time I went past, all became routine.
clearly asked ‘Where they all gone?’ I knew but hadn’t the Eventually our GP, fed up with seeing me asking for more
heart to tell him. sleeping pills (for myself) said he would refer us to a private
Still active, he sought the outdoors and would spend psychiatrist. Great, I thought, perhaps something could be
hours pacing the nearby park. Within a year or two he was done about my husband’s agitation. It was only after two
becoming a danger crossing roads so I would accompany sessions that I realized I was the patient, that he thought I
him; in fact I have never walked so much in my life as those was depressed. I was furious. Yes, I probably was depressed
years. Often I would drive to the river and we would walk but could not coping with my husband’s illness be one rea-
along the towpath to look at boats seeking a tranquillity not son why?
possible in the city centre. From the outset, being under 65 years of age, Leonard
At home he invariably followed me round the house, was not eligible to be seen by a geriatrician, but this psychia-
sat next to me when I was speaking on the telephone and trist did refer us to social services and an appointment was
accompanied me to the bathroom, unwilling to let me out of made for an assessment at the mental health day hospital
his sight. Touching, and I understood why, but desperately and in turn this led to some helpful visits from an occupa-
lonely and starved of company and good conversation, I felt tional therapist.
I was going crazy and remember on occasion pushing him Anti-depressants helped me, but I was aware he was
away from my side. depressed too. The psychiatrist’s suggestion that I try him
He never saw a doctor, he was never ill in the way of on my pills was a disaster – he became totally disoriented.
having colds or flu. No one seemed to think I could be I took matters into my own hands and in a convoluted way
finding life hard, it was as if I didn’t exist, I had no status. found a retired psychologist from the neuro-rehabilitation
Even as a doctor’s wife, I was unaware that there should be unit at The National Hospital and subsequently my husband
help available and that someone other than the none-too- attended regularly for over a year – again privately. When I
helpful GP should know of our situation. That no expres- collected him he was different, happier, it was worth any-
sion of sympathy, concern or encouragement ever came thing to see him like that even if the effects lasted only a few
from the medical profession was something I found hard hours. Helpfully I was given feedback on what he seemed
to bear. unable to express to me, as the time he knew I was going
In desperation I tried to access drug trials, enquired away for a week and had found a private respite home for
at centres of research in the London area and followed up him just outside London. Evidently he thought I would leave
every relevant journal article, but to no avail. When eventu- him there and never come back.
ally he was invited to an assessment his Mini-Mental State Local day-centres were totally unsuitable. How could I
Examination score was below the criteria for eligibility. leave him sitting in front of a TV with a rug over his knees
Failed again, although of course that trial came to nothing. in the company of 80-year-olds? It took months to track
All those years ago an eminent neurologist assured me not down a private centre run by a charity that had received
to worry about the remote possibility of the children inher- a good review in an Alzheimer’s Society magazine. When
iting the condition since within 20 years there would be a a place became available I was relieved to discover two
cure. Where is it? other younger men attending. I had to convince my pro-
During the early years we took some holidays abroad. testing husband that he was going there to help, he was
He loved the sun and I discovered that cruises were ideal; a ­doctor after all. The three became good friends and
we had our own cabin, he couldn’t get lost – well he could, although by this stage none could read, I am told some
we both did, but couldn’t go far – he could pace the deck, amusing c­onversations ensued on the subject of cur-
visit new places, watch films. The motion of the ship and rent affairs, but it didn’t matter, it gave them a sense of
the wide sea proved almost hypnotic. I never knew what companionship.
our fellow passengers made of us. I recall him asking the The downside was that it was on the other side of
captain of the ship whether he was going to Leningrad London, so Tuesdays and Thursdays I had a frantic rush
too! These holidays were hard work mentally, but, on bal- to get him up and dressed for the hour’s drive through
ance, worthwhile, a last attempt to lead as normal a life as the rush-hour traffic. I then ended up the opposite side
possible. of London with insufficient time easily to drive home and
A couple of years later agitation was added to the equa- back to collect him.
tion and I was forced on alert 24-hours a day. I still man- By then I had decided I needed to know why I felt the
aged to lose him once and those three hours until he was way I did (dreadful) and enrolled on a part-time external
The lived experience of dementia 21

Diploma in Psychology at London University. Lectures were suggested something to calm him down. It was haloperidol.
in the evenings so I had to leave our son to give my husband When we returned a month later I said he seemed worse,
supper and see him into bed. Subsequently I found a good could barely talk or walk now and was showing signs of
public library near the day centre and would spend those incontinence. The dose was doubled.
days struggling with essays. If only I had known then what I know now. In my igno-
When he was in long-term care I went on to gain an MSc rance I was unaware for whom haloperidol was meant or
in Gerontology. At the back of my mind was the thought what the side-effects were, and to this day I blame myself
that when all this was over I could have years ahead and for his rapid descent into severe dementia. After only a few
would have to do something with my life. Sadly, going back weeks he became a bent, drooling, shuffling shell of his for-
to my former occupation was out of the question. During mer self, without facial expression and with involuntary foot
my years as a carer I have completely lost any creative ability jerking movements that were to stay with him for the rest of
and although I still paint as a hobby can no longer produce his days. I was distraught, no one seemed to listen to me and
imaginative children’s designs. I felt powerless to help my poor husband. I had become a
At first I didn’t even realize I was a so-called ‘carer’. I bystander watching horror after horror being heaped upon
was using occasional sitters from a local private agency – in him as he hurtled down the dark tunnel of AD. It was heart-
fact we went straight from hiring baby-sitters to husband- breaking, it was obscene, haloperidol yet another trigger to
sitters, sometimes they were the same girls. Eventually, after institutionalization.
much form-filling, social services allocated us home respite One day as I collected Leonard from the day centre the
for three hours a week. This only worked well during a six manager took me aside and said sorry they couldn’t have
month ‘pilot’ when a carer with specific dementia train- him back if he carried on like this, he hadn’t sat down all
ing looked after him and I could go out with an easy mind. day, refused to eat, was falling and disrupting the oth-
Usually it was a different carer each week and I spent the ers. When we arrived home I phoned the GP and for the
time away from home fretting and worrying. first and only time asked for a home visit. A young locum
In time a voluntary organization in the Borough offered doctor arrived, took one look at him shuffling round the
a 2-hour free sitting service once a week. The first after- living-room in tight circles, leaning sideways at an alarm-
noon a lovely Jamaican nurse arrived and I went off to ing angle and foaming at the mouth and said ‘Take him
the dentist. When I returned my husband was wander- to A & E.’
ing about the house causing havoc and no sign of her. Midsummer’s Eve and, if I had but known it, this was
Evidently he had told her to go and, not being obliged to to be his Last Journey. He never came home again to live.
stay, she left. She had dreadlocks and beads in her hair and The taxi driver surveyed us suspiciously as I spread a plas-
I think he was scared of her. When I tactfully tried to ask tic sheet on the seat. It wasn’t a heart attack or another fit
the office whether they had someone else, we were accused as the locum suspected and after a few days on an acute
of being racist – no good telling them that my husband medical ward he was transferred to the chaotic in-patient
was not racist, that you cannot rationalize with someone assessment ward at the Mental Health unit from where,
with dementia and, if he was scared, no amount of talking despite my protests, it was eventually recommended he go
would convince him otherwise. We went to the bottom of into long-term care.
the waiting list. The last years of his life in first a home for the elderly
Two years later we must have reached the top again and mentally ill, where within a few weeks, he broke a hip, and
for a few blissful months a kindly lady would arrive at 8 p.m. a few months later the other hip (no one saw these incidents
alternate Tuesdays and beg me to enjoy an evening out with happen) were a nightmare. I had been told I would feel only
friends. Night out, you’re kidding! Friends! What friends? I relief when he went into care. In fact it was harder. I was
would take a large glass of wine and a sleeping pill and shut not in control of his life, how understanding staff were, how
myself in a bedroom until 8 o’clock next morning when she they handled him, what he was given to eat and I hated to
would knock on my door with my husband showered and see him wearing someone else’s clothes. AD had robbed him
dressed. of his dignity and he had always been an immensely digni-
Those carers who say they find their role rewarding and fied man.
spiritually uplifting fill me with disbelief. I could never see I spent hours with him each day trying to provide some
it like that, only as an appalling tragedy with catastrophic quality of life and the sort of interaction never given in the
consequences for him and all who loved him and in the Home. After breaking the second hip he never got back on
absence of any pastoral concern from within our commu- his feet again so when he broke a femur – apparently due
nity I turned away from religion altogether. Laughter was to staff’s poor skills with the hoist – I refused to let him
conspicuously absent; obviously my fault that unlike other return there and he was eventually found a bed in continu-
carers I was unable to find amusing any of the incidents in ing National Health Service care in a private nursing home
our day-to-day life, it was just unremitting hard work laced not too far from where we lived.
with utter despair. I had to accept that nowhere would be perfect or pro-
The agitation became worse and worse over the next vide the standard of care I wished for him, but I found
couple of years. Eventually the unsympathetic geriatrician it impossible to forgive the poor quality of his and the
22 Dementia

other residents’ lives. To the Home Manager’s annoyance more antibiotics, just let him go in peace.’ Ten days later he
I employed some private carers; a strong man able to push died, with myself, our son and son-in-law by his side. His
the wheelchair up the hill to the park weather permitting; a journey through a living hell was over, a farewell that had
delightful Irish lady to sit with him occasionally – and sing; lasted nearly 16 years. Watching him being wheeled out to
a caring Russian girl to give him tea on Saturdays so that the waiting ambulance on a cold drizzly February Friday
I could see my children outside the care home setting and was the bleakest day of my life.
visit my parents. Leonard, these you have missed: watching our children
And my parents are still alive now, my father 98. They embarking on their paths to adulthood, through univer-
loved their son-in-law, their honorary doctor they called sity, their careers, celebrating their successes. You were
him, and couldn’t accept what was happening to him, so out there, but not there. You saw both the girls on their wed-
of turn. Neither could bear to visit him in the Home, so con- ding days wearing their beautiful white dresses because
sequently saw little of me during those years as my time was we stopped by the Home on our way to the ceremony, but
spent beside Leonard. I don’t know if you recognized them or understood the
I doubt being GP to a care home with so many frail momentousness of the day. You never had the pleasure
elderly people is every doctor’s dream, but the ability to of walking your daughters up the aisle or knowing your
diagnose concomitant illnesses of old age in non-verbal delightful sons-in-law and daughter-in-law. Our son grew
patients is key to their quality of life. On one occasion I had up not knowing you for the brilliant, kind and caring man
been insisting to the GP for over two weeks that something you had once been.
was very wrong, that it couldn’t ‘still be an allergy’. It was You would have adored the grandchildren, five of them
for me to telephone a former dermatologist colleague of now and, incredibly, twins on the way. You always doted
my husband’s who that same day diagnosed bullous pem- on our children, how much more time you would have
phigoid and started him on steroids. I had always believed had in retirement to enjoy the next generation. I trea-
nature should be allowed to take its course, but in another sure the photograph of our first grandchild Sam on your
24 hours, undiagnosed, he would have died the most appall- lap just a few weeks before you died. You were staring
ing death as those massive blisters spread down his throat ­w ide-eyed at this little baby; I so hope you knew it was
and other orifices. yours.
How could I feel other than anger and helplessness? And our children … I still cannot fathom the extent
Interestingly that year on steroids he reverted to something to which their lives have been affected. The day after he
resembling his old self; smiles and occasional laughter not graduated from Cambridge our son started his first job – in
heard for years. When I suggested he stay on a low-dose Scotland – as far away from home as possible. Now they are
given the feelings of wellbeing they promoted, I was told all married, all back in London and seem happy, although
this wasn’t ethical. What about all those ethical issues ill- the spectre of AD, unspoken, must linger on occasion.
prepared family carers are forced to confront at all stages of Rightly or wrongly, I tried to shield them from the caring
the disease? What is ethical about the way some care homes role, feeling that it should not be allowed to dominate their
treat people with dementia? lives and I would never ever wish or expect them to look
Much as I disliked the Home and literally had to after me should I succumb in the future. I tried so hard
grit my teeth as I rang the entry bell each day, I always to be a good wife and carer, mother and father, daughter
longed to see my husband, to touch him and prattle on and eventually grandmother, but in the end was so over-
with my usual monologue, my mental state so closely tied whelmed, so torn in a myriad directions, I am not sure I
with how he was. If he was unwell, in pain, or looking have succeeded as any of these.
­d istressed I would go home in the evening feeling dis- Current research is trying to emphasize the positive
traught. If he was alert and calm I felt lighter and slept aspects of caring. Few coping 24/7 with challenging behav-
better. The wording on the birthday cake one year said iour, incontinence, social services and unhelpful carers
Happy 88th Birthday; he was 68. Numbed and aghast in are going to accept being told it is a positive and satisfac-
equal measure, I couldn’t protest. Sadness was ever pres- tory experience. However if carers survive the physical and
ent, but now it was weariness, wondering what disaster emotional turmoil of a ‘caring career’ there is evidence that
would befall him next and an overriding sense of fear that we emerge more confident, more empowered to deal with
engulfed me. statutory authorities. Literally, we have fought a battle and
‘What do you mean, MRSA? How?’ The Home man- come through it.
ager had no idea how or where, and gradually over the In my case the positive legacy includes teaching medi-
next few months with a series of chest infections, sores, cal students, postgraduate clinical psychologists and nurses
and endless courses of antibiotics, Leonard went down- about the impact of dementia on families, active involve-
hill. He looked so unhappy and anguished, painfully thin, ment on many major dementia research projects and the first
just bones really, and had a greatly diminished quality carer to be awarded a research grant by the UK Alzheimer’s
of life. I had always worried how I would know when the Society. I continue my interest because our children are
end was approaching. Suddenly I knew. I had to force the nearing middle age and, who knows, potentially at risk in
nurse out of the room as I pleaded with the GP ‘please no the next decade or two.
The lived experience of dementia 23

The mental and physical costs are long-lasting: for me,


anxiety, chronic insomnia and backache from all those years 2.2 POSTSCRIPT
of ‘lifting’. The year before he died I spent weeks in hospital
and recuperating from legionella and have never felt com- My caring career started over twenty years ago. Awareness
pletely well since. Feeling totally knocked out, I remember of dementia has increased exponentially with the predicted
sobbing by his side, convinced he would outlive me yet. The explosion of numbers likely to be affected in the future.
toll on family relationships; with my parents (who in some Research inches forward and more is known about the
obscure way I think blame him for ruining their daughter’s benefits of psychological interventions. We already have
life); and wider family members who all had their own views evidence on what optimum care looks like and the special
on how I was looking after him and what I should, or should needs of younger people with dementia. We family carers
not, be doing for the best. The social legacy is immense in believe there needs to be a change of attitude, more effort
terms of work, lifestyle, relationships and, of course, finances. and the necessary funding to translate that evidence into
However, Life goes on: new friends, new hobbies, some delivering better quality care and training as the main route
amazing fellow carers, travel, a degree of contentment I to improving life for those suffering now and until such
never believed possible to achieve again. But tears are never time as effective symptom relief or a cure becomes available.
far away and I would do anything to wind the clock back 25 The latter of course remains the ultimate goal.
years and be sharing happy memories with my beloved hus- Our turbulent journey, just one of millions being acted
band as we grow old together, rather than rushing around out across the globe, may serve as a caution to all those pro-
the country telling how it was. fessionals involved in caring for this group.
3
Prevalence and incidence of dementia

DANIEL W. O’CONNOR

not confirmed by a large controlled trial of HRT as a treat-


3.1 SCOPE OF EPIDEMIOLOGY ment of dementia, perhaps because women taking HRT
were healthier and at lower risk (Almeida and Flicker,
Epidemiology is the study of the distribution and determinants 2005). Apart from selection bias, reasons for discrepancies
of disease in human populations. It maps the frequency of dis- between analytic and experimental studies include con-
ease and identifies people at higher or lower risk of contracting founding by pre-existing cognitive impairment, insufficient
particular conditions. Once risk factors are confirmed, their treatment dosing and limited follow-up (Coley et al., 2008).
impact can be reduced. Studies linking cigarette smoking Since dementia develops over many decades, interventions
with lung cancer, and hypertension with stroke, sparked suc- might need to be deployed decades earlier to be effective.
cessful campaigns to limit smoking and lower blood pressure. Particular risk factors for AD and vascular dementia
Ideally, epidemiological studies will identify reversible trig- (VaD) are addressed in Chapter 44 and Chapters 57 through
gers to Alzheimer’s disease (AD) and other dementias. 60. This chapter focuses instead on the starting point of
Links between life experience and disease are identi- epidemiological knowledge, namely studies of the preva-
fied by means of observational, analytic and experimental lence and incidence of dementia and its various subtypes.
approaches. Observational studies entail large-scale sur- Wherever possible, information comes from community
veys to measure the frequency of dementia and to note cor- surveys rather than clinical settings.
relations with sociodemographic, biometric and lifestyle
variables. Surveys, while costly and time-consuming, are
essential in conditions like dementia. Many cases go unrec-
ognized by primary and specialist health services, even 3.2 METHODOLOGICAL ISSUES
in high-income countries, and clinical registers are there-
fore not reliable sources of data (Iliffe and Robinson, 2009; The prevalence of dementia measures the proportion of peo-
Knopman et al., 2011). ple within a population who meet diagnostic specifications.
Analytic studies contrast the backgrounds of people with Incidence measures the numbers of new cases of dementia
dementia (drawn from community surveys or clinical prac- within a defined period. Prevalence studies are simpler: people
tice) with those of matched controls to identify points of dif- are divided into ‘cases’ of dementia and ‘non-cases’ at a single
ference that might, on further study, prove to be risk factors. point in time. Some will have developed dementia recently:
Case–control studies are economical but accurate matching others have had it for many years. A community might there-
is problematic and associations will emerge by chance if fore have a higher than usual prevalence because its residents
questions cover hundreds of putative causes. To complicate with dementia survive longer. Incidence studies entail two
matters, people with dementia cannot report accurately on surveys, a year or more apart, to determine the annual con-
exposure. Information must therefore be sought from rela- version of former ‘non-cases’ to ‘cases’. They have greater sci-
tives whose knowledge is imperfect and subject to bias. entific value since rates are not confounded by survival.
Both types of study, observational and analytic, gener- The first surveys of mental disorder date from the
ate hypotheses to be tested in experimental trials, some- 1950s. Lin (1953), who supplemented brief evaluations of
times with unexpected results. The finding in some (but Taiwanese residents with information from relatives and
not all) case–control studies that hormone replacement hospitals, concluded that only 0.5% of people aged over 60
therapy (HRT) conferred protection against dementia was years had ‘senile dementia’. By contrast, Essen-Moller (1956)

24
Prevalence and incidence of dementia 25

diagnosed 28% of Swedes aged over 70 years with ‘mild or responsible. In reality, differences might stem largely or
severe dementia’ based on detailed personal assessments by solely from methodological discrepancies. The few studies
skilled clinicians. This 60-fold discrepancy might reflect in which identical research protocols were applied to dispa-
genuine differences in dementia prevalence, but method- rate populations are discussed in Section 3.7.
ological factors were almost certainly to blame.
Even today, there is no commonly agreed protocol to 3.2.1 SCREENING
screen, assess and diagnose dementia in community popu-
lations. As a result, prevalence rates can rarely be compared Population surveys are so expensive that most researchers
directly. There is little disagreement in advanced cases of use a brief cognitive test to select respondents for detailed
dementia: discrepancies arise more commonly in mild or evaluation. Of the many tests available, the 30-point Mini-
borderline cases. Since early dementia merges impercepti- Mental State Examination (MMSE) (Folstein et al., 1975),
bly with the normal age-related cognitive decline, the divid- which takes only 10 minutes or so to administer and requires
ing line will vary depending on investigators’ definitions no specialist equipment, is used most commonly in high-
and assessment tools. income countries (Ismail et al., 2010). In a meta-analysis
By way of illustration, Table 3.1 summarizes the meth- of 21 community and primary studies, 83% of people with
ods employed in three large, well-documented reports from dementia scored at or below the selected cut-point (usually
a regional South Indian city (Mathuranath et al., 2010), a 23 points) while 87% of ‘normals’ scored above it (Mitchell,
regional centre in Western Turkey (Arslantaş et al., 2009) 2009). However, only a third of those ‘failing’ the MMSE met
and a representative sample of older South Koreans (Kim et criteria for dementia so further investigation is mandatory.
al., 2011). The studies varied in important respects (research Since the MMSE tests reading, writing and arithmetic,
personnel, participants’ education, screening tests, screen- performance is shaped by education. In the United Kingdom,
ing cut-points and special assessments) making it difficult to 11% of ‘normal’ older people who left school before their fif-
compare one with another. Diagnostic criteria, while appar- teenth birthday scored 23 points or less compared with only
ently identical, might also be operationalized quite differ- 3% of those with higher education. Conversely, the same
ently (Seshadri et al., 2011). Reported dementia prevalence cut-point missed 27% of dementias in better educated people
rates in age group 70–74 years varied from 2.6% in India to compared with only 12% in those with limited education
5.2% in South Korea and 9.8% in Turkey (albeit with wide (O’Connor et al., 1989c). Adjusting cut-points to reflect age-
confidence intervals). If true, further investigation is war- and education-specific norms seems not to improve validity
ranted to determine what genetic or lifestyle factors are in literate communities (Cullen et al., 2005). A better strategy

Table 3.1 Comparison of methods and results in three dementia prevalence studies
Place India South Korea Turkey
First author (year) Mathuranath (2010) Kim (2011) Arslantaş (2009)
Respondents (N) 2466 6141 3100
Sampling frame Census Census Census
Sample, age (years) Aged over 55 Aged over 65 Aged over 55
Institutions Not applicable Included Included
Education 21% ≤ 4 years education 16% no education 18% illiterate
Screening test Addenbrooke’s Cognitive MMSE MMSE
Examination
Informant interviews Some Yes Yes
Investigations Clinical interview, Clinical interview, Clinical interview,
neuropsychology, neuropsychology, neuropsychology,
physical examination, laboratory tests, imaging laboratory tests, imaging
laboratory tests, imaging
Diagnostic criteria DSM-IV, NINCDS-ADRDA, DSM-IV, NINCDS-ADRDA, DSM-IV, NINCDS-ADRDA,
Hachinski Ischemia Scale NINCDS-AIREN, other NINCDS-AIREN, other
Dementia types AD, VaD AD, VaD, LBD AD, VaD, other
Research personnel Interviewers, psychologists, Interviewers, nurses, Clinicians
neurologists psychologists, psychiatrists
Dementia prevalence, 70–74 2.6% 5.2% 9.8%
years
Abbreviations: MMSE, Mini-Mental State Examination; DSM-IV, Diagnostic and Statistical Manual of Mental Disorders, 4th Edition;
AD, Alzheimer’s disease; LBD, Lewy bodies dementia; VaD, vascular dementia.
26 Dementia

in two-phase studies is to evaluate all who score below the Revised (DSM-III-R) and Clinical Dementia Rating criteria
chosen cut-point together with a proportion of high scorers to vignettes of older people identified in clinics and com-
to reduce the risk of missing early or atypical dementias. munity surveys, between-centre agreement was generally
The MMSE is not an appropriate screening tool in coun- high. As expected, agreement rates were lower at the border
tries where older people are often illiterate and innumer- of ‘normal ageing’ and ‘mild dementia’ and for frail, deaf or
ate. An alternative instrument, the Community Screening depressed participants (O’Connor et al., 1996).
Instrument for Dementia which combines culturally sensi- The critical role of diagnostic glossaries was shown by
tive cognitive test items with an informant interview, has Erkinjuntti et al. (1997) who used data from a multistage sur-
performed well in epidemiological surveys in India, China, vey of 10,000 older Canadians to illustrate how criteria (and
Latin America and Africa and is available in a shortened also assessment procedures) shape prevalence rates. Only
form (Prince et al., 2011). 3% of those surveyed met the International Classification of
In high-income countries, up to half of all people with Diseases, 10th Edition (ICD-10) criteria for dementia com-
moderate and severe dementia will be missed if residents pared with 29% on DSM-III-R. Similar anomalies emerged
of long-stay hospitals and aged care facilities are excluded in studies in middle-income countries where prevalence
(O’Connor et al., 1989b). With respect to geographic scope, rates based on 10/66 dementia specifications were roughly
some investigators approach all eligible residents of a suburb double those based on inferred Diagnostic and Statistical
or town. Others employ complex strategies to select represen- Manual of Mental Disorders, 4th Edition (DSM-IV) criteria
tative residents of entire cities, states or countries. A smaller (Prince et al., 2012).
locale promotes cooperation, reduces costs and makes Community diagnoses based on thorough clinical
­follow-up easier. Larger scale studies ensure that findings can evaluations and informant histories have acceptable clini-
be generalized to the whole of a region or nation. The choice cal validity. In a large British survey, only 3 of 56 people
of approach is driven by study resources and objectives. diagnosed with mild dementia by the Cambridge Mental
Survey response rates are an issue if participants dif- Disorders of the Elderly Examination (CAMDEX) were
fer significantly from non-participants. Very active older judged not to have dementia 2 years later (O’Connor et al.,
people can be difficult to contact while, conversely, people 1991). Factors contributing to premature diagnoses included
with dementia are sometimes shielded by carers. In one U.S. deafness, depression and unstable diabetes mellitus.
study, older people who declined to participate in a survey Validity is best confirmed by neuropathological exami-
were twice as likely to have been diagnosed with dementia nation but gathering post-mortem material in community
as participants (Knopman et al., 2014). populations is challenging. In a comparison of well-assessed
community and clinic research participants, community-
3.2.2 DIAGNOSIS based subjects were less likely to have pure AD but more
likely to have cortical infarctions and mixed pathologies
Ideally, clinical assessment comprises a mental status exami- (Schneider et al., 2009).
nation, cognitive battery, informant interview, neurological Given all these issues, it is desirable to apply identical
examination, laboratory tests and neuroimaging with inputs assessment and diagnostic procedures when comparing
from physicians, psychiatrists and neuropsychologists, but dementia rates in setting with another. This occurs infre-
not all studies meet this standard (Prince et al., 2013). quently. Only the geriatric mental status schedule (Copeland
Informant interviews are essential. Since dementia et al., 1991) and the 10/66 Dementia Research Group proto-
impairs memory and insight, information must be sought col (Prince et al., 2007) have been deployed across multiple
from a relative or carer about the subjects’ personal details, communities and countries.
cognition, functional status, medical and psychiatric his-
tory and medications. These details help to confirm the
diagnosis of dementia, especially in its early stages, and to 3.3 DEMENTIA
distinguish AD from other disorders. Informants’ reports
correlate highly with other markers of cognitive and func- 3.3.1 PREVALENCE
tional capacity: under-reporting and over-reporting are
remarkably uncommon (O’Connor et al., 1989a). Small num- If dementia is truly more common in one part of the world
bers of respondents forbid contact with informants or have or in people from a particular background, knowledge of
no surviving relatives. One stand-alone, structured infor- the responsible social or environmental triggers might lead
mant interview, the Informant Questionnaire on Cognitive to preventative or mitigating interventions.
Decline in the Elderly (IQCODE), has sound psychometric In the first of several meta-analyses, Jorm et al. (1987)
properties barely affected by education (Jorm, 2004). took findings from 22 reports to construct a mathemati-
If research findings are to carry weight, diagnostic criteria cal model relating dementia prevalence to age and study
must be interpreted similarly in different centres. In a study characteristics. Rates varied widely but there was a con-
of five research centres in Australia, Europe and the United sistent trend for prevalence to rise exponentially with age
states in which clinicians were asked to apply Diagnostic with a doubling every 5.1 years (Table 3.2). This was con-
and Statistical Manual of Mental Disorders – 3rd Edition, firmed by Hofman et al. (1991) who reanalysed data from
Prevalence and incidence of dementia 27

Table 3.2 Prevalence rates (%) for dementia by age evidence came from the Bronx Aging Study in which the
derived from 22 studies risk of dementia grew steadily with age, but with a slowed
rate of growth from age 80 (Hall et al., 2005). The matter
Age (Years)
is not completely settled though. In a study of Californian
65–69 1.4 nonagenarians, dementia incidence rates climbed steadily
70–74 2.8 over the 4-year follow-up period with very high rates among
75–79 5.6 centenarians (Corrada et al., 2010).
80–84 10.5 In the few studies to date that tracked changes in demen-
85–89 20.8 tia incidence rates using consistent assessment and diagnos-
90–94 38.6 tic methods over periods of one to two decades, there were
95–99 144.3 no significant changes in rates in China (Li et al., 2007) and
the Netherlands (Schrijvers et al., 2012).
Source: Data from Jorm, A.F. et al., Acta Psychiatrica Scandinavica,
76, 465–479, 1987.
3.3.3 SURVIVAL
12 European studies conducted between 1977 and 1989 in
which dementia was defined using DSM-III or equivalent The best estimates of survival come from follow-up of newly
criteria. No major differences emerged between countries. diagnosed cases since prevalence surveys capture only sur-
More recently, a meta-analysis of data from 147 studies vivors. In the Leipzig Longitudinal Study that followed new
across most parts of the world found a prevalence of 5%–7% cases of dementia for 3 years, 51% of participants died com-
over age 60 in most regions. There were pointers to higher pared with 19% of non-impaired controls. Male sex, age,
than average rates in Latin America and lower ones in West frailty and dementia severity increased mortality risk fur-
sub-Saharan Africa but the similarities between regions ther (Gühne et al., 2006). In the Cache County study, the
were more striking than any differences (Prince et al., 2013). risk of death was 3.5 times higher than normal for people
Two studies with strikingly high prevalence rates hint at with dementia aged 75–84 and 7.3 times higher for those
an association between dementia and social deprivation. In aged 65–74 (Tschanz et al., 2004).
the first, aged residents of three Arab–Israeli villages were
assessed using modified cognitive tests and informant inter-
views. A quarter met DSM-IV criteria for dementia, three 3.4 ALZHEIMER’S DISEASE
times more than in comparable Jewish–Israeli communi-
ties. Risk increased with age, female gender and illiteracy 3.4.1 PREVALENCE
(Bowirrat et al., 2001). In Australia, where many indigenous
people have limited education and multiple medical co- Table 3.3 presents findings from a meta-analysis of AD rates
morbidities, the prevalence of dementia above age 65 was using data from 11 European studies conducted in the 1990s
27% based on detailed, culturally appropriate assessments (Lobo et al., 2000). As before, rates rise sharply with age.
(Smith et al., 2008). By contrast, rates were very low in Cree Corrada et al. (1995) applied logistic regression to iden-
Indian settlements in northern Canada where older people tify methodological factors associated with variability in
remain actively engaged in fishing, trapping and crafts 15 published papers from Europe, United States, Japan and
(Hendrie et al., 1993). China. After adjusting for age, higher rates tended to be
Three groups of investigators tracked dementia preva- reported in studies that included mild cases and used labo-
lence rates over time using identical (or at least very similar) ratory tests to aid in diagnosis. Lower rates emerged from
methods within a defined geographic area. Rates were sig- studies that used brain scans and cerebral ischaemia scores.
nificantly lower in the UK study, pointing to improvements This is not surprising. Studies that can afford laboratory
in cognitive health over a 20-year period (Matthews et al., tests are likely to be more thorough in other respects too
2013). However, prevalence remained stable with time in and studies using brain scans might attribute cases, rightly
a 10-year study of an African-American community (Hall or wrongly, to visible evidence of vascular pathology.
et al., 2009) and a 20-year Swedish study (Qiu et al., 2013).
3.4.2 INCIDENCE
3.3.2 INCIDENCE
In a refinement of an earlier meta-analysis, Ziegler-Graham
If it is true that prevalence increases exponentially with age, et al. (2008) found that incidence rates doubled every
varying only in its time of onset, everyone will be affected 5.5 years quite consistently in 27 studies from Asia, Europe
at some point in their life. This hypothesis, which has major and the Americas. Base rates varied somewhat between
public health implications, was tested by Ritchie and Kildea regions but not to a statistically significant degree. Findings
(1995) who analysed findings from 12 studies with data over from three recent incidence surveys in the United States
age 80, adequate sampling procedures and clear diagnostic (Miech et al., 2002), Brazil (Nitrini et al., 2004) and Canada
methods. Their logistic model showed a flattening of growth (Tyas et al., 2006) lay within the bounds identified in the
at age 95, suggesting that dementia is not inevitable. Further meta-analyses.
28 Dementia

Table 3.3 Prevalence rates (%) of Alzheimer’s disease in a Table 3.4 Prevalence rates (%) of vascular dementia in a
meta-analysis of 11 European studies meta-analysis of 11 European studies

Age (Years) Males Females Age (Years) Males Females


65–69 0.6 0.7 65–69 0.5 0.1
70–74 1.5 2.3 70–74 0.8 0.6
75–79 1.8 4.3 75–79 1.9 0.9
80–84 6.3 8.4 80–84 2.4 2.3
85–89 8.8 14.2 85–89 2.4 3.5
90+ 17.6 23.6 90+ 3.6 5.8
Source: Data from Lobo, A. et al., Neurology, 54 (Suppl. 5), S4–S9, Source: Data from Lobo, A. et al., Neurology, 54 (Suppl. 5), S4–S9,
2000. 2000.

3.4.3 SURVIVAL Americans (Borenstein et al., 2014). By contrast, Japanese


men in Hawaii, whose lifestyle is more authentically Asian,
Younger onset adds to the lethality of AD. In a follow-up study had VaD rates similar to those in Japan (White et al, 1996).
of 108 incident cases identified in the Baltimore Longitudinal
Study of Aging, diagnosis was associated with a 39% reduc- 3.5.2 INCIDENCE
tion in median life span for people aged 90 years compared
with 67% for those aged 65 years (Brookmeyer et al., 2002). Incidence rates vary widely from study to study for the rea-
sons outlined earlier but are generally much lower than for
AD. Rates look to be higher in Asian populations than North
American ones (Fratiglioni et al., 1999), more in older stud-
3.5 VASCULAR DEMENTIA ies than recent ones (Homma and Hasegawa, 2000).

3.5.1 PREVALENCE
3.6 OTHER DEMENTIAS
Estimating the prevalence of VaD is difficult: definitions
and assessment methods vary too widely to give confidence While most attention has been paid to AD and VaD, demen-
in study findings. In a review by Rocca and Kokmen (1999), tia due to other causes accounted for 11% of cases in the
for example, prevalence rates were higher in studies that 25 European prevalence surveys summarized by Fratiglioni
combined ‘mixed dementia’ with VaD and based diagnoses et al. (1999).
on radiologists’ reports of brain scans. In a meta-analysis of 18 community surveys, there was
In a meta-analysis by Lobo et al. (2000), rates were lower a wide variation in prevalence rates of Lewy body demen-
than those for AD (Table 3.4). This fits with findings from tia, depending on the investigators’ diagnostic criteria and
community autopsy series in the United Kingdom and thresholds. Prevalence rates ranged from 0% to 5% (mean
United States showing a preponderance of AD pathol- 0.52%) at age 65 years and over, with Lewy body demen-
ogy, though with high admixtures of vascular abnormali- tia accounting for between 0% and 20% (mean 4.6%) of
ties (Neuropathology Group of the MRCCFA Study, 2001; all identified dementias (Vann Jones and O’Brien, 2014).
Schneider et al., 2009). Frontotemporal dementia is discussed in the following.
Rates of VaD look to be higher in Asia. When Fratiglioni Dementias due to potentially reversible causes are rare. In
et al. (1999) compared findings from 25 European studies a U.S. case file review of 560 new cases of dementia identified
with five from Japan, Korea, India and China, the propor- in the Rochester Epidemiology Project, non-­degenerative
tions of cases ascribed to VaD were 28% in Europe and causes were identified in 30% of young-onset patients but
38% in Asia. It was diagnosed twice more often than AD only 5% in the older-onset group. The most common causes
in a series of Japanese studies but ratios were similar in two were chronic mental illness, brain tumours, alcoholism and
Chinese studies (Graves et al., 1996). cerebral anoxia (Knopman et al., 2006). No cases were iden-
Caucasian–Asian differences might be real, not artifac- tified of dementia due to normal pressure hydrocephalus,
tual. In one rigorous, 7-year Japanese study in which diag- subdural haematoma, hypothyroidism, vitamin B12 defi-
noses were confirmed whenever possible by post-mortem ciency or neurosyphilis.
examination, 61% of dementias in men were attributed
to vascular disease, a much higher rate than observed 3.6.1 EARLY-ONSET DEMENTIA
in equivalent studies in Europe and the United States
(Yoshitake et al., 1995). Similar findings were reported in Dementia before 65 years of age is too rare to warrant a
a more recent study by Ikeda et al. (2001). Lifestyle is prob- door-to-door survey. Instead, frequency estimates are
­
ably more influential than genes: rates of VaD in elderly derived from checks of memory clinics, hospital records and
Japanese-Americans were similar to those of other North diagnostic registers. Given the gravity of these conditions
Prevalence and incidence of dementia 29

for carers and service providers, many cases are likely to be In a rural community in northern India, only 1% of peo-
captured in this way though socially marginalized people ple aged over 65 met DSM-IV criteria for dementia despite
can evade detection for lengthy periods. In a London study low literacy levels when assessed in detail using tools modi-
in which 227 cases nominated by general practitioners fied from an earlier U.S. survey. Gender and literacy made
(GPs), specialists and community services were reviewed no difference (Chandra et al., 1998). In a 2-year follow-up,
in detail, diagnoses in age group 45–64 years were as fol- incidence rates were about a third those found in the United
lows: AD (35%), VaD (18%), frontotemporal dementia (15%), States (Chandra et al., 2001) suggesting that northern Indian
alcohol-related dementia (14%) and other conditions (18%) lifestyles are protective.
including Lewy body dementia, Huntingdon’s disease, mul-
tiple sclerosis, corticobasal degeneration, Down’s syndrome
and unspecified conditions (Harvey et al., 2003).
The 80 cases identified in the same age group in a Sydney 3.8 FUTURE TRENDS
linkage study had a different spread of conditions: alcohol-
related dementia (25%), AD (15%), frontotemporal dementia According to burden of disease estimates, dementia con-
(9%), VaD (11%) and other or unspecified conditions (40%) tributes to more years lived with disability in later life than
(Withall et al., 2014). stroke, musculoskeletal disorders, cardiovascular disease
Frontotemporal dementia appears to be more common and cancer (WHO, 2003). Given population ageing, the rap-
than previously thought. In a sustained community-based idly growing numbers of people with dementia will therefore
search, Dutch neurologists and nursing home physicians impose great demands on families and social, residential and
identified 245 cases of whom 40 came to autopsy. The medical services throughout the world. To map this global
median age of onset was 58 years with a range of 33–80 impact, Brookmeyer et al. (2007) applied a complex math-
years. The prevalence rose from 3.6 per 100,000 at age 50–59 ematical model of incidence, progression and survival in AD
to 9.4 at age 60–69 years and then fell to 3.8 at 70–79 years to projected world populations. These very rough estimates
(Rosso et al., 2003). Incidence rates have since been reported suggest that the numbers of AD will quadruple between 2006
in the United Kingdom, with numbers similar to those for and 2050 from 27 to 106 million. Numbers in world areas
early-onset AD (Mercy et al., 2008). might conceivably be as follows: 6 million in Africa, 9 million
Irrespective of cause, early-onset dementia resulted in in North America, 11 million in Latin America, 16 million
much increased mortality in a Dutch memory clinic cohort. in Europe and 63 million in Asia. Modelling suggests that if
Over a short follow-up period (mean 2 years), the risk of interventions could delay both disease onset and progression
death was 43 times higher than for controls of the same age. by just 1 year, there would be nine million fewer cases over-
For dementia with later onset, by contrast, the risk was only all. Prevention is therefore of paramount importance.
three times higher (Keodam et al., 2008).

3.9 CONCLUSIONS
3.7 COMPARATIVE STUDIES
Surveys of dementia prevalence and incidence provide
Putative socio-environmental risk factors will be identified varying rates, most probably because of differences in study
more confidently if identical case-finding tools are applied methodology. Future studies will be easier to interpret now
to groups with vastly different social, educational, dietary, that many investigators are using similar, and sometimes
occupational and medical exposures. identical, approaches to assessment and diagnosis.
Revealing findings emerged from a direct comparison Alzheimer, vascular and other pathologies often coex-
of dementia in elderly Nigerian-Africans and African- ist in advanced old age. Seemingly clear-cut diagnoses in
Americans. This decade-long study applied identical prevalence, incidence and survival studies must therefore
screening tests and diagnostic evaluations comprising neu- be interpreted cautiously.
ropsychology, informant interviews, personal evaluations Dementia might be more common in certain places and
and CT brain scans to residents of Ibadan, Nigeria and cultures. It is important to compare incidence (not preva-
Indianapolis, United States (Hendrie et al., 1995). Contrary lence) rates of AD, VaD and so on in aged people from
to expectation, the age-standardized prevalence rate of AD widely differing backgrounds. Observations of very low
was lower in Nigerians aged over 65 years than African rates of dementia in Nigeria and northern India might
Americans (1.4% versus 6.2%). Rates for dementia of all prove to be of great significance.
causes were lower too (2.3% versus 8.2%). This disparity
was not due to greater life expectancy in the United States. REFERENCES
In a 5-year follow-up, age-standardized annual incidence
rates of AD were still lower in Ibadan, perhaps because of Almeida, O.P. and Flicker, L. (2005). Association between
lower weight, blood pressure, glucose and cholesterol levels hormone replacement therapy and dementia: Is it time
(Hendrie et al., 2001). to forget? International Psychogeriatrics, 17: 155–164.
30 Dementia

Arslantaş, D., Özbabalik, D., Metintaş, S. et al. (2009). Folstein, M.F., Folstein, S.E. and McHugh, P.R. (1975).
Prevalence of dementia and associated risk fac- Mini-Mental State: A practical method for grading the
tors in middle Anatolia, Turkey. Journal of Clinical cognitive state of patients for clinicians. Journal of
Neuroscience, 16: 1455–1459. Psychiatric Research, 12: 189–198.
Borenstein, A.R., Wu, Y., Bowen, J.D. et al. (2014). Fratiglioni, L., De Ronchi, D. and Aguera-Torres, H. (1999).
Incidence rates of dementia, Alzheimer disease Worldwide prevalence and incidence of dementia.
and vascular dementia in the Japanese American Drugs and Aging, 15: 365–375.
population in Seattle, WA. Alzheimer’s Disease and Graves, A.B., Larson, E.B., Edland, S.D. et al. (1996).
Associated Disorders, 28: 23–29. Prevalence of dementia and its subtypes in the
Bowirrat, A., Treves, T.A., Freidland, R.P. and Korczyn, A.D. Japanese American population of King County,
(2001). Prevalence of Alzheimer’s disease in an elderly Washington State: The Kame project. American
Arab population. European Journal of Neurology, 8: Journal of Epidemiology, 144: 760–771.
119–123. Gühne, U., Matschinger, H., Angermeyer, M.C. and Riedel-
Brookmeyer, R., Corrada, M.M., Curriero, F.C. and Heller, S.G. (2006). Incident dementia cases and mor-
Kawas, C. (2002). Survival following a diagnosis of tality: Results of the Leipzig Longitudinal Study of the
Alzheimer’s disease. Archives of Neurology, 59: Aged. Dementia and Geriatric Cognitive Disorders,
1764–1767. 22: 185–193.
Brookmeyer, R., Johnson, E., Ziegler-Graham, K. and Hall, K.S., Gao, S., Baiyewu, O. et al. (2009). Prevalence
Arrighi, H.M. (2007). Forecasting the global burden rates for dementia and Alzheimer’s disease in African
of Alzheimer’s disease. Alzheimer’s and Dementia, 3: Americans: 1992 versus 2001. Alzheimer’s and
186–191. Dementia, 5: 227–233.
Chandra, V., Ganguli, M., Pandav, R. et al. (1998). Hall, C.B., Verghese, J., Sliwinski, M. et al. (2005). Dementia
Prevalence of Alzheimer’s disease and other demen- incidence may increase more slowly after age 90:
tias in rural India: The Indo–US study. Neurology, 51: Results from the Bronx Aging Study. Neurology, 65:
1000–1008. 882–886.
Chandra, V., Pandav, R., Dodge, H.H. et al. (2001). Harvey, R.J., Skelton-Robinson, M. and Rossor, M.N.
Incidence of Alzheimer’s disease in a rural community (2003). The prevalence and causes of demen-
in India. Neurology, 57: 985–989. tia in people under the age of 65 years. Journal
Coley, N., Andrieu, S., Gardette, V. et al. (2008). of Neurology, Neurosurgery and Psychiatry, 74:
Dementia prevention: Methodological explanations 1206–1209.
for inconsistent results. Epidemiologic Reviews, 30: Hendrie, H.C., Hall, K.S., Pillay, N. et al. (1993). Alzheimer’s
35–66. disease is rare in Cree. International Psychogeriatrics,
Copeland, J.R.M., Dewey, M.E. and Saunders, P. (1991). 5: 5–14.
The epidemiology of dementia: GMS-AGECAT stud- Hendrie, H.C., Ogunniyi, A., Hall, K.S. et al. (2001).
ies of prevalence and incidence, including studies in Incidence of dementia and Alzheimer disease in 2
progress. European Archives of Psychiatry and Clinical communities: Yoruba residing in Ibadan, Nigeria and
Neuroscience, 240: 212–217. African Americans residing in Indianapolis, Indiana.
Corrada, M., Brookmeyer, R. and Kawas, C. (1995). Journal of the American Medical Association, 285:
Sources of variability in prevalence rates of 739–747.
Alzheimer’s disease. International Journal of Hendrie, H.C., Osuntokun, B.O., Hall, K.S. et al. (1995).
Epidemiology, 24: 1000–1005. Prevalence of Alzheimer’s disease and dementia
Corrada, M., Brookmeyer, R., Paganini-Hill, A. et al. (2010). in two communities: Nigerian Africans and African
Dementia incidence continues to increase with age in Americans. American Journal of Psychiatry, 152:
the oldest old: The 90+ study. Annals of Neurology, 1485–1492.
67: 114–121. Hofman, A., Rocca, W.A., Brayne, C. et al. (1991). The
Cullen, B., Fahy, S., Cunningham, C.J. et al. (2005). prevalence of dementia in Europe: A collaborative
Screening for dementia in an Irish community sample study of 1980–1990 findings. International Journal of
using MMSE: A comparison of norm-adjusted versus Epidemiology, 20: 736–748.
fixed cut-points. International Journal of Geriatric Homma, A. and Hasegawa, K. (2000). Epidemiology of
Psychiatry, 20: 371–376. vascular dementia in Japan. In E. Chiu, L. Gustafson,
Erkinjuntti, T., Ostbye, T., Steenhuis, R. and Hachinski, V. D. Ames and M.F. Folstein (eds.), Cerebrovascular
(1997). The effect of different diagnostic criteria on Disease and Dementia: Pathology, Neuropsychiatry
the prevalence of dementia. New England Journal of and Management. London: Martin Dunitz, pp. 33–46.
Medicine, 337: 1667–1674. Iliffe, S., Robinson, L., Brayne, C. et al. (2009). Primary care
Essen-Moller, E. (1956). Individual traits and morbid- and dementia: 1. Diagnosis, screening and disclo-
ity in a Swedish rural population. Acta Psychiatrica sure. International Journal of Geriatric Psychiatry, 24:
Scandanavica, 100 (Suppl.): 1–160. 895–901.
Prevalence and incidence of dementia 31

Ikeda, M., Hokoishi, K., Maki, N. et al. (2001). Increased Miech, R.A., Breitner, J.C.S., Zandi, P.P. et al. (2002).
prevalence of vascular dementia in Japan: A com- Incidence of AD may decline in the early 90s for men,
munity-based epidemiological study. Neurology, 57: later for women. Neurology, 58: 209–218.
839–844. Mitchell, A.J. (2009). A meta-analysis of the accuracy of
Ismail, Z., Rajji, T.K. and Shulman, K.I. (2010). Brief cogni- the mini-mental state examination in the detection of
tive screening instruments: An update. International dementia and mild cognitive impairment. Journal of
Journal of Geriatric Psychiatry, 25: 111–120. Psychiatric Research, 43: 411–431.
Jorm, A.F. (2004). The informant questionnaire on cog- Neuropathology Group of the Medical Research Council
nitive decline in the elderly (IQCODE): A review. Cognitive Function and Ageing Study (2001).
International Psychogeriatrics, 16: 275–293. Pathological correlates of late-onset dementia in a
Jorm, A.F., Korten, A.E. and Henderson, A.S. (1987). The multi-centre, community based population in England
prevalence of dementia: A quantitative integration and Wales. Lancet, 357: 169–175.
of the literature. Acta Psychiatrica Scandinavica, 76: Nitrini, R., Caramelli, P., Herrera, E. et al. (2004). Incidence
465–479. of dementia in a community-dwelling Brazilian popu-
Keodam, E.L.G.E., Pijnenburg, Y.A.L., Deeg, D.J.H. et lation. Alzheimer’s Disease and Associated Disorders,
al. (2008). Early-onset dementia is associated with 18: 241–246.
higher mortality. Dementia and Geriatric Cognitive O’Connor, D.W., Blessed, G., Cooper B. et al. (1996).
Disorders, 26: 147–152. Cross-national interrater reliability of dementia
Kim, K.W., Park, J.H., Kim, M.H. et al. (2011). A nation- diagnosis in the elderly and factors associated with
wide survey on the prevalence of dementia and mild disagreement. Neurology, 47: 1194–1198.
cognitive impairment in South Korea. Journal of O’Connor, D.W., Pollitt, P.A., Brook, C.P.B. and Reiss, B.B.
Alzheimer’s Disease, 23: 281–291. (1989a). The validity of informant histories in a com-
Knopman, D.S., Petersen, R.C., Cha, R.H. et al. (2006). munity study of dementia. International Journal of
Incidence and causes of nondegenerative nonvascu- Geriatric Psychiatry, 4: 203–208.
lar dementia: A population-based study. Archives of O’Connor, D.W., Pollitt, P.A., Hyde, J.B. et al. (1989b)
Neurology, 63: 218–221. The prevalence of dementia as measured by
Knopman, D.S., Petersen, R.C., Rocca, W.A. et al. (2011). the Cambridge Mental Disorders of the Elderly
Passive case-finding for Alzheimer’s disease and Examination. Acta Psychiatrica Scandinavica, 79:
dementia in two U.S. communities. Alzheimer’s and 190–198.
Dementia, 7: 53–60. O’Connor, D.W., Pollitt, P.A., Treasure, F.P. et al. (1989c).
Knopman, D.S., Roberts, R.O., Pankrantz, V.S. et al. (2014). The influence of education, social class and sex on
Incidence of dementia among participants and Mini-Mental State scores. Psychological Medicine, 19:
nonparticipants in a longitudinal study of cogni- 771–776.
tive aging. American Journal of Epidemiology, 180: O’Connor, D.W., Pollitt, P.A., Jones, B.J. et al. (1991).
414–423. Continued clinical validation of dementia diag-
Li, S., Yan, F., Li, G. et al. (2007). Is the dementia rate nosed in the community using the Cambridge
increasing in Beijing? Prevalence and incidence of Mental Disorders of the Elderly Examination. Acta
dementia 10 years later in an urban elderly popula- Psychiatrica Scandinavica, 83: 41–45.
tion. Acta Psychiatrica Scandinavica, 115: 73–79. Prince, M., Acosta, D., Ferri, C.P. et al. (2011). A brief
Lin, T.Y. (1953). A study of the incidence of mental disor- dementia screener suitable for use by non-
der in Chinese and other cultures. Psychiatry, 16: specialists in resource poor settings: The cross-
313–336. cultural derivation and validation of the brief
Lobo, A., Launer, L.J., Fratiglioni, L. et al. (2000). Community Screening Instrument for Dementia.
Prevalence of dementia and major sub-types in International Journal of Geriatric Psychiatry, 26:
Europe: A collaborative study of population-based 899–907.
cohorts. Neurology, 54 (Suppl. 5): S4–S9. Prince, M., Acosta, D., Ferri, C.P. et al. (2012). Dementia
Mathuranath, P.S., Cherian, P.J., Mathew, R. et al. (2010). incidence and mortality in middle-income countries
Dementia in Kerala, South India: Prevalence and and associations with indicators of cognitive reserve:
influence of age, education and gender. International A 10/66 Dementia Research Group population-based
Journal of Geriatric Psychiatry, 25: 290–297. cohort study. Lancet, 380: 50–58.
Matthews, F.E., Arthur, A., Barnes, L.E. et al. (2013). A Prince, M., Bryce, R., Albanese, E. et al. (2013). The global
two-decade comparison of prevalence of dementia in prevalence of dementia: A systematic review and
individuals aged 65 years and older from three geo- meta-analysis. Alzheimer’s and Dementia, 9: 63–75.
graphical areas of England. Lancet, 382: 1405–1412. Prince, M., Ferri, C.P., Acosta, D. et al. (2007). The proto-
Mercy, L., Hodges, J.R., Dawson, K. et al. (2008). Incidence cols for the 10/66 Dementia Research Group popula-
of early-onset dementias in Cambridgeshire, United tion-based research programme. BMC Public Health,
Kingdom. Neurology, 71: 1496–1499. 7 (1): 1.
32 Dementia

Qiu, C., von Strauss, E., Bäckman, L. et al. (2013). Twenty- Tschanz, J.T., Corcoran, C., Skoog, I. et al. (2004).
year changes in dementia occurrence suggest Dementia: The leading predictor of death in
decreasing incidence in central Stockholm, Sweden. a defined elderly population. Neurology, 62:
Neurology, 80: 1888–1894. 1156–1162.
Ritchie, K. and Kildea, D. (1995). Is senile dementia “age- Tyas, S.L., Tate, R.B., Wooldrage, K. et al. (2006).
related” or “ageing-related”? Evidence from meta- Estimating the incidence of dementia: The impact of
analysis of dementia prevalence in the oldest old. adjusting for subject attrition using health care utili-
Lancet, 346: 931–934. zation data. Annals of Epidemiology, 16: 477–484.
Rocca, W.A. and Kokmen, E. (1999). Frequency and distri- Vann Jones, S.A. and O’Brien, J.T. (2014). The prevalence
bution of vascular dementia. Alzheimer Disease and and incidence of dementia with Lewy bodies: A
Associated Disorders, 13 (Suppl. 3): S9–S14. systematic review of population and clinical studies.
Rosso, S.M., Kaat, L.D., Baks, T. et al. (2003). Psychological Medicine, 44: 673–683.
Frontotemporal dementia in the Netherlands: Patient White, L., Petrovich, H., Ross, W. et al. (1996). Prevalence of
characteristics and prevalence estimates from a dementia in older Japanese-American men in Hawaii:
population-based study. Brain, 126: 2016–2022. The Honolulu-Asia Aging Study. Journal of the
Schneider, J.A., Aggarwal, N.T., Barnes, L. et al. (2009). The American Medical Association, 276: 955–960.
neuropathology of older persons with and without Withall, A., Draper, B., Seeher, K. and Brodaty, H. (2014).
dementia from community versus clinic cohorts. The prevalence and causes of younger onset
Journal of Alzheimer’s Disease, 18: 691–701. dementia in Eastern Sydney, Australia. International
Schrijvers, E.M.C., Verhaaren, B.F.J., Koudstaal, P.J. et al. Psychogeriatrics, 26: 1955–1965.
(2012). Is dementia incidence declining? Trends in WHO (2003). World Health Report: Shaping the Future.
dementia incidence since 1990 in the Rotterdam Geneva, Switzerland: World Health Organization.
study. Neurology, 78: 1456–1463. Yoshitake, T., Kiyohara, Y., Kato, I. et al. (1995). Incidence
Seshadri, S., Beiser, A., Au, R. et al. (2011). Operationalising and risk factors of vascular dementia and Alzheimer’s
diagnostic criteria for Alzheimer’s disease and disease in a defined elderly Japanese population:
other age-related cognitive ­impairment—Part 2. The Hisayama study. Neurology, 45: 1161–1168.
Alzheimer’s and Dementia, 7: 35–52. Ziegler-Graham, K., Brookmeyer, R., Johnson, E. and
Smith, K., Flicker, L., Lautenschlager, N.T. et al. (2008). High Arrighi, H.M. (2008). Worldwide variation in the
prevalence of dementia and cognitive impairment in doubling time of Alzheimer’s disease incidence rates.
indigenous Australians. Neurology, 71: 1470–1473. Alzheimer’s and Dementia, 4: 316–323.
4
What is dementia, and how do you assess it?
Definitions, diagnostic criteria and assessment

JOSEPH P.M. KANE AND ALAN THOMAS

and senile dementia (Reisberg, 2006). Kraepelin (1899), who


4.1 INTRODUCTION had referred to presenile dementia 8 years before Alzheimer’s
findings, considered presenile dementia to represent a rare
While many of the defining characteristics of the dementia illness occurring exclusively in patients under 65 years old,
concept have been recognized for over 100 years, develop- and senile dementia to be the expression of cerebral arte-
ments in recent decades have both influenced, and been riosclerosis in patients over 65 (Reisberg, 2006). Alzheimer
influenced by, the way we think about dementia. The discov- had reservations over whether his findings represented a
ery of biomarkers and the development of individual ­criteria distinct clinicopathological entity, not least because of sub-
for aetiologies like Alzheimer’s disease (AD) (Dubois et al., sequent identification of neurofibrillary tangles in patients
2007), frontotemporal dementia (FTD) (Rascovsky et al., with senile dementia, but this perception of a dichotomy
2011) and dementia with Lewy bodies (DLB) (McKeith persisted until 1965 when Corsellis and Evans (1965) failed
et al., 2005) have sought to refine the diagnosis, but they to find a marked association between arteriosclerosis and
also present pressures on clinicians to go beyond dementia senile dementia, and continued to be reflected as separate
to its causal subtypes. There are also additional challenges entities in modern classification until 1980 (Reisberg, 2006).
from an ageing population, the emergence of mild cognitive Arguably, Kraepelin’s writings have contributed more to
impairment (MCI) as a diagnostic entity and its relation- the modern conception of dementia than any other single
ship to ageing-related change, and wider social, political body of work. In addition to recognizing dementia’s psycho-
and economic pressures. pathological heterogeneity, he placed senile dementia at the
This chapter discusses dementia as both a concept and extreme of a continuum of age-associated mental weakness
a diagnostic label, exploring the main characteristics of that included impaired comprehension and mental flex-
recently published diagnostic criteria. It discusses some of ibility, memory lapses (particularly for recent events) and
the challenges faced in translating these criteria from the disturbed attention. Critically, he recognized senile demen-
research environment into clinical practice, before outlin- tia as a disorder that had a significant impact on everyday
ing the process of clinical assessment. For detailed informa- living and independence, and which continues to form a
tion about the history of the concept of dementia, the reader cornerstone of both classification and diagnosis (Kurz and
is referred to Chapter 1. Lautenschlager, 2013).

4.2 HISTORICAL BACKGROUND 4.3 THE ADVENT OF CLASSIFICATION

Alzheimer’s observation of plaques and neurofibrillary tan- Changing conceptualizations of dementia since the lat-
gles in the brain of Auguste D (Alzheimer, 1907), a 51-year- ter half of the twentieth century are illustrated and influ-
old woman, and the subsequent baptism of his eponymous enced most clearly by the criteria listed in the Diagnostic
disorder by Emil Kraepelin (1910) effectively saw the cre- and Statistical Manual of Mental Disorders (DSM) and the
ation of two conceptual entities: presenile dementia, or AD, International Classification of Diseases (ICD).

33
34 Dementia

The first two editions of DSM, together with ICD-8 and but increasing recognition of common neuropathological
-9 (American Psychiatric Association, 1952, 1968; World features of presenile and senile dementia, together with
Health Organization, 1974, 1978), provided only a brief its classification in DSM-III provoked exponential growth
generalized description of ‘organic brain syndrome’ (OBS), in research into the disorder and wider recognition as an
leaving considerable scope for interpretation by clinicians. aetiology.
‘A mental disorder caused by diffuse impairment of brain DSM-IV (American Psychiatric Organization, 1994) dis-
function and manifested by impaired cognitive functions’, posed of OBS entirely as the term was felt to imply incor-
OBS encompassed both delirium (acute brain syndrome) rectly that ‘non-organic’ mental disorders do not have a
and dementia (chronic brain syndrome), distinguishing biological basis.
them primarily on the basis of reversibility, but it excluded It retained the conceptual framework laid out by DSM-
focal impairment and failed to acknowledge the existence III in necessitating multiple cognitive deficits, including
of reversible dementias. Impairment in functioning of suf- memory impairment, but brought about an important
ficient severity to ‘interfere grossly with the capacity to meet conceptual change in lowering the functional diagnostic
the ordinary demands of life’ was employed as a means of threshold of impairment from ‘sufficient to interfere with
severity rather than a requisite for the diagnosis itself. social or occupational functioning’ to ‘sufficient to cause
While DSM-III (American Psychiatric Association, limitations in complex activities’; limitation of activities
1980) retained the concept of an OBS, it was the first of such as work and managing finances were now considered
the systems to introduce dementia as a unitary concept, as sufficient evidence of a functional impact on functioning,
describing it as one of 10 OBS subtypes that also included and therefore dementia diagnosis. Rather than excluding
intoxication, withdrawal, organic personality syndrome cases where symptoms arose in the context of clouding of
and organic hallucinosis (Fox, 1983). DSM-III was a land- consciousness, DSM-IV specified that symptoms must arise
mark text, selling 500,000 copies in the United States in in the absence of delirium itself, not least as it had been rec-
14 languages; use for the first time extended far beyond that ognized that clouding of consciousness could occur in DLB.
of psychiatry into legal practice, government agencies, the Thus, successive criteria have defined dementia through
insurance industry and social services (George et al., 2011). specification of a number of core criteria, many of which can
It ushered in a ‘diagnostic revolution’ that transformed the be traced back to the work of Kraepelin and his predeces-
clinical conception of all psychiatric disorders from a broad sors: cognitive impairment, representing a decline in previ-
dimensional one to a more categorical approach, bringing ous abilities, adversely affecting everyday functioning, and
its authors, the American Psychiatric Association, to the differentiated from acute and transient cognitive impair-
forefront of the field of psychiatric nosology (George et al., ment (delirium). Although subsequent classification systems
2011). DSM-III provoked unprecedented debate and con- have expanded their descriptions of cognitive impairment
troversy that has, perhaps, escalated with the publication of beyond that of Kraepelin, amnesia, accompanied by impair-
each subsequent edition. ment in at least one other cognitive domain, has remained
DSM-III marked a more defined, operationalized the hallmark of the cognitive decline of dementia, and the
approach to diagnosis, evident in changes to criteria for focus of attention for clinicians and researchers. Revision
dementia. It specified memory impairment as a necessary and refinement of the boundaries representing functional
criterion and required impairment in multiple domains, impairment, however, has led to a successive lowering of
with deficits in ‘higher cortical functions’ (aphasia, apraxia, thresholds for dementia, recognizing impairment in com-
or agnosia), impairment of judgement or a change in per- plex tasks, rather than in more basic activities of living, as
sonality required in addition to amnesia. It sought to dif- being sufficient for a diagnosis. Clarification of both cogni-
ferentiate dementia from delirium by including clouding of tive and functional parameters has remained a key objective
consciousness and the presence of specific organic factors of recent dementia diagnostic criteria, such as DSM-5 and
as exclusion criteria. The threshold for functional impair- the National Institute on Aging-Alzheimer’s Association
ment appeared to diminish with inclusion of the require- (NIA-AA) criteria.
ment for impairment of sufficient severity ‘to interfere with
social or occupational functioning’. ICD-10 (World Health
Organization, 1992), although less circumspect in defining
functional changes, required the presence of a disturbance
4.4 DSM-5 AND NIA-AA CRITERIA
in multiple higher cortical functions; memory was included,
but not necessary. An ageing global population has led to an increasing preva-
Neither DSM-I nor II had referred to AD, but by describ- lence of dementia, and to a growing recognition of cogni-
ing primary degenerative dementia and multi-infarct tive decline and dementia as a continuum that encompasses
dementia, DSM-III reintroduced these terms to the scien- pre-dementia syndromes, their diagnosis and treatment.
tific and clinical lexicon (Boller and Forbes, 1998; George Historically common underlying causes of cognitive impair-
et al., 2011). Boller and Forbes (1998) note the paucity ment, such as B12 deficiency, hypothyroidism and neuro-
of AD in research literature during the 1970s, with only syphillis, have disappeared with easy identification and
42 papers published using ‘Alzheimer’ as a keyword in 1975, effective treatment, while some new, but rare causes, such as
What is dementia, and how do you assess it? Definitions, diagnostic criteria and assessment 35

human immunodeficiency virus (HIV) and its related cog- dementia’ (Ganguli et al., 2011) and to propose a uniform
nitive impairment have produced new challenges to clinical framework for defining all dementing disorders. These DSM
and research communities. A greater understanding of the criteria recognize that although categorization is necessary
aetiologies underpinning dementia, aided by advances in and helpful, a continuum exists between normal cognitive
diagnostic techniques, together with disease-specific treat- function and dementia, with MCI (or mild neurocognitive
ments targeting these aetiologies, have led to greater efforts disorder in DSM language) lying between these categories.
to define pathological disorders. While in the past, the The creation of the broad category of ‘neurocognitive dis-
fierce competition between Kraepelin’s Munich and Arnold order’ reflects this, with subcategories of mild and major
Pick’s Prague laboratories was said to be amongst the for- neurocognitive disorders comparable to MCI and dementia,
mer’s motivation to classify and name AD, the international respectively, albeit with important differences between the
research community today is characterized by a more col- DSM categories and their NIA-AA counterparts. Here, we
laborative spirit, aided by advances in communications and focus on definitional differences in dementia between the
transport, and exemplified by the revised internationally two.
agreed consensus criteria for AD, DLB, Parkinson’s disease
dementia (PDD) and FTD, all published in recent years. 4.4.1 COGNITIVE IMPAIRMENT
Although consensus groups continue to develop crite-
ria, there remains a need for operationalized criteria for Although DSM-5’s major NCD definition retains many
the overarching diagnosis of dementia itself, particularly of the characteristics of NIA-AA’s description, changes to
as the legal and social implications remain as pertinent as both cognitive and functional aspects of the criteria repre-
they were in antiquity. There is also increasing recognition sent a significant broadening of the scope of the diagnosis.
that in the oldest-old, multiple pathologies are the norm, Where DSM-IV, and indeed most conceptualizations of
not the exception, and that clinically distinguishing a sin- dementia since Kraepelin, required the presence of cogni-
gle aetiology in such people is very difficult (Savva et al., tive decline in more than one domain, one of which must be
2009; Jellinger and Attems, 2010). Both the recent DSM-5 memory impairment, neither NIA-AA nor DSM-5 require
(American Psychiatric Organization, 2013) and NIA-AA the presence of amnesia as part of their evidence of cogni-
(McKhann et al., 2011) criteria have attempted to address tive decline. This demonstrates a common shift away from
this, seeking to define dementia first, before considering memory impairment as the central feature for diagnosis,
underlying subtypes, a method expected to be mirrored and perhaps a move away from the use of AD as the blue-
in ICD-11, scheduled for 2017. Both classification systems print for conceptualization of dementia, a criticism which
demonstrate an effort to clarify functional impairment has long been levelled at dementia diagnostic criteria.
and neurocognitive domains better, and in doing so, the However, while NIA-AA criteria still require the pres-
boundaries of normal ageing, MCI and dementia. Although ence of cognitive impairment in at least two of five listed
some disagreement exists between the two systems over the domains, in keeping with DSM-IV conceptualization, a
importance of impairment in a single cognitive domain, DSM-5 diagnosis of major NCD requires impairment in
the diminishing significance of memory impairment in one cognitive domain only. In making this change, DSM-5
each represents a move away from the Alzheimerization of has created a category much broader than that of NIA-AA’s
dementia as an entity. dementia, including patients who previously would have
The NIA-AA work groups, who updated the National been diagnosed with DSM-IV’s amnestic disorder and
Institute of Neurological Disorders and Stroke–Alzheimer those with severe aphasia. DSM-5 cites that this evidence
Disease and Related Disorders (NINCDS–ADRDA) criteria of decline in a single domain must be accompanied by ‘con-
in 2011 (McKhann et al., 2011), have retained in their cri- cerns’, rather than complaints, on the part of the patient,
teria the nomenclature and basic framework of its prede- informant or clinician that there has been a significant
cessors; cognitive or behavioural symptoms must interfere decline in cognitive performance; this reflects that actual
with the ability to function at work or at usual activities, impairment may not have been reported, rather elicited by
representing a decline from previous levels of function- questioning. This may be viewed as a purely academic point,
ing, and not be explained by delirium or major psychiatric given the unlikelihood that assessment will occur in the
disorder. The NIA-AA self-consciously tried to maintain absence of expressed concerns by at least one of the patient,
continuity with its previous criteria, while DSM-5 intro- informant or clinician.
duced not only changes to the diagnostic criteria but also DSM-5’s decision to specify a single cognitive domain,
changes to the nomenclature itself, coining the term ‘major rather that multiple domains, represents a greater concep-
neurocognitive disorder’ (major NCD) in place of ‘demen- tual change than the new nomenclature itself; perhaps the
tia’. The latter was considered not only to be pejorative and most significant since the abandonment of OBS and is not
stigmatizing but also believed to be synonymous with AD without significant controversy. The DSM work group cite
in the eyes of many patients, policymakers and clinicians. this change as a conscious effort to classify ‘neurocognitive
It also represents an explicit intent of the part of its authors disorder’ as an entity spanning a broad range of age groups
(the Neurocognitive Disorders Work Group) to ‘reopen the and aetiologies, many of which have in recent years moved
global debate on the terminology and conceptualization of away from the use of ‘dementia’, such as frontotemporal
36 Dementia

lobar degeneration or cognitive impairment due to trau- in everyday life from a diagnosis, and therefore categorizes
matic brain injury (TBI) or HIV infection (Ganguli et al., functioning on a complementary, but separate, scale to its
2011). While the group is at pains to point out the distinc- diagnostic ICD-10 manual; the International Classification
tion between major NCD and NIA-AA’s more conventional of Functioning, Disability and Health (ICF) conceptualizes
conception of dementia (Ganguli et al., 2011), the DSM’s functioning as a dynamic interaction between an individu-
significant role in the history of psychiatric nosology, al’s medical condition, their environment and personal fac-
and the public and media spotlight shone on the manual, tors. Understandable concerns regarding both discontinuity
prompts one to wonder whether the two overlapping defi- with preceding criteria and a potential for an overemphasis
nitions could become confused outside the specialist envi- on cognitive measures, precluded removal of functioning
ronment. In the opinion of many clinicians and researchers, from the DSM-5 criteria (Ganguli et al., 2011), but its role as
dementia is an inherently broad construct with the under- a key criterion of dementia diagnosis is likely to remain the
lying pathologies necessarily involving multiple cognitive subject of debate; ICD-11, scheduled for publication in 2015,
domains. By broadening the scope of the criteria to be more aims to harmonize ICF with all diagnostic criteria while
inclusive of rarer and earlier forms of cognitive damage, continuing to recognize functioning as separate conceptual
DSM-5 has muddied the waters, grouping those with static entity from disease (Escorpizo et al., 2013).
single-domain deficits together with the major degenerative DSM-5 and NIA-AA nonetheless retain functional
causes of dementia. The majority population of people with impairment as an important component of their diag-
dementia are those who have degenerative and widespread nostic criteria, recognizing the varied complex functional
vascular brain disease, a different patient group from typi- tasks that often comprise patients’ and carers’ presenting
cally younger TBI and HIV patients. complaints, and the significance of these features in demar-
Both classifications go further than previous proposals in cating entities like MCI and dementia in the cognitive
detailing parameters of cognitive impairment, in an effort impairment spectrum. At first glance, DSM-5’s requirement
to gain consensus in a poorly defined area. DSM-5 defines of impairment ‘sufficient to interfere with independence’
and illustrates six domains (complex attention, executive for a diagnosis of major NCD might not appear to repre-
ability, learning and memory, language, visuoconstruc- sent a significant change from that of DSM-IV’s ‘sufficient to
tional and perceptual motor ability, social cognition), over interfere with social or occupational functioning’, but this
three pages of text, while NIA-AA specify five (impaired is broadly comparable with NIA-AA’s description of suffi-
ability to acquire and remember new information; impaired cient functional impairment (significant interference in the
reasoning and handling of complex tasks; poor judgement; ability to function at work or in usual daily activities) and
impaired visuospatial abilities; impaired language function helps to offer a clearer description of the boundary between
and changes in personality, behaviour or comportment). dementia and MCI. DSM-5 and NIA-AA’s respective criteria
Although these categories are broadly comparable, NIA-AA for Mild NCD and MCI recognize that a degree of impair-
has no domain analogous to DSM-5’s ‘complex attention’, ment of complex functional tasks, such as paying bills or
characterized by ‘sustained attention, divided attention, shopping, may exist without the impairment necessarily
selective attention and processing speed’. As well as defin- meeting the functional threshold for dementia diagnosis.
ing the domains, DSM-5 attempts to quantify the degree of A patient’s performance of a complex task may, for exam-
impairment necessary for diagnosis, specifying a threshold ple, involve a greater number of errors, or a longer period
for cognitive performance as two standard deviations below of time than that previously required, yet remain within
appropriate norms (or below the third percentile). the boundaries of MCI or normal ageing; the patient may
use tools like pill boxes or calendars as aids in maintain-
4.4.2 FUNCTIONAL IMPAIRMENT ing tasks like medication management. In such cases, while
the patient’s functioning has been impaired, independence
Despite the changes intended to clarify and sharpen the has been maintained. Only once assistance is required (e.g.
cognitive components of dementia diagnosis, both catego- supervision of medicine use or financial management), is
rization systems remain necessarily vague regarding the independence compromised and functional impairment
functional aspect of the dementia assessment process, and deemed sufficient for a diagnosis of dementia.
are broadly comparable with recent predecessors. Critically,
however, functional impairment remains a crucial part of 4.4.3 SUBCATEGORIZATION
the boundary separating MCI from dementia and MCI’s
DSM-5 counterpart (mild NCD) from major NCD. It should Having outlined the criteria of a mild or major NCD,
be noted that this boundary is not without controversy; the DSM-5 then further subcategorizes by aetiology, the pres-
World Health Organization (WHO) recommend that the ence or absence of behavioural disturbance, and disease
classification of functioning should be kept entirely separate severity (mild/moderate/severe). The aetiology subcategory
from diagnosis, with impaired functioning and disability represents a considerable expansion on pathologies listed
being considered as a consequence of disease, rather than a in DSM-IV, including criteria for AD, cerebrovascular dis-
diagnostic feature (World Health Organization, 2001). The ease, FTD, DLB, PDD, Huntington’s disease, TBI, prion
organization suggests that one cannot infer participation disease and substance use-associated disease. These criteria
What is dementia, and how do you assess it? Definitions, diagnostic criteria and assessment 37

are deliberately consistent with those of respective expert These requirements, while not representing diagnostic
groups on each subtype, which is important, given devel- criteria for these other diseases, rather instances in which a
opments in AD, DLB and FTD criteria since the last edi- diagnosis of probable AD is not applicable, set a high thresh-
tion. Biomarkers supportive of each aetiology, although old that continues to recognize AD as the default entity to
listed alongside respective criteria, are generally omitted be considered following dementia diagnosis. For NIA-AA,
from the criteria themselves, having been eschewed by the only if there is very good reason to think of a non-AD cause,
DSM-5 working group. A notable exception to this trend is should another aetiology be considered. This position
the inclusion of evidence of a causative AD genetic mutation would appear to be contrary to evidence suggesting that
from family history or genetic testing. It is hoped that reflec- DLB and FTD are often unrecognized in clinical practice
tion on the increasing body of evidence indicating the pres- in favour of AD (Knopman et al., 2004; Knapp et al., 2007),
ence of pathology prior to presentation will lead to inclusion and can be contrasted with DSM-5’s explicit intention to
of relevant biomarkers in future DSM criteria, something move away from the Alzheimerization of the dementia con-
which is likely to improve early identification and diagnosis cept. In contrast, DSM-5’s criteria for NCD due to probable
of dementia subtypes. The behavioural disturbance subcat- AD, requires decline in at least two domains, one of which
egory reflects the prevalence of depression, psychosis and must be memory and learning, a steadily progressive course
agitation in neurocognitive disorders, and their frequent without extended plateaus, and no evidence of mixed aetiol-
role as the primary cause for presentation to services. ogy, representing a higher threshold for probable AD, and a
DSM-5’s categorization of aetiology after identification much lower one for consideration of other pathologies.
of major NCD is outside of the remit of NIA-AA criteria, In summary, the major conceptual difference between
which after defining the dementia syndrome, goes on to list the NIA-AA and DSM-5 criteria is in the latter’s widen-
AD groups, stratified by likelihood of AD diagnosis and ing of the threshold of cognitive impairment from multi-
evidence of the AD pathological process. Examination of ple domains to that of a single domain, creating a broader
NIA-AA’s dementia and probable AD categories provides category, major NCD. However, both NIA-AA and DSM-5
an insight into how the working group perceives the rela- criteria abandon the requirement of amnesia and so rep-
tionship between the two entities, especially when con- resent a conceptual move away from the AD blueprint of
trasted with DSM-5’s description of the syndromes. Of the dementia evident in their predecessors. Despite NIA-AA
five cognitive domains listed by NIA-AA, at least two of retaining the broader traditional characteristics of demen-
which are required for a diagnosis of ‘all-cause dementia’, tia, it has a different approach to categorization of demen-
four correspond to probable AD dementia subtypes: amnes- tia subtypes, creating a high threshold for consideration of
tic presentation, the most common, and three non-amnestic non-AD pathologies, in contrast to DSM-5, which pushes
presentations (language, visuospatial and executive dys- in the opposite direction in encouraging consideration of
function presentations). Only cognitive impairment involv- other potential causes of the dementia syndrome.
ing changes in personality, behaviour or comportment, in
addition to impairment in at least one other domain, direct
the clinician away from an AD diagnosis once NIA-AA
4.5 COMPARING CLASSIFICATION
dementia criteria are met, broadening the range of clini-
cal syndromes recognized as AD, perhaps consistent with
SYSTEMS
the provenance of these criteria. This is compounded by
well-defined exclusion criteria for probable AD in demen- At first glance, differences between classification systems
tia patients that create a high threshold for consideration of and their successive revisions might appear modest, retain-
non-AD pathological causes of dementia: only ­substantial ing Kraepelin’s core concepts throughout and reflecting
concomitant cerebrovascular disease with multiple or gradual changes in the language employed in discussing
extensive infarcts or severe white matter hyperintensity bur- thresholds of functional and cognitive impairment. Closer
den is deemed sufficient to exclude patients with features of consideration of these criteria, however, particularly in the
vascular dementia (VaD), thus including all-cause demen- research setting, demonstrates that they reflect significant
tia patients with, for example, single infarcts and insidious differences in the understanding of dementia, with impor-
onset. While DLB consensus criteria require only one core tant implications for practice. This has been demonstrated
feature of fluctuating cognition, recurrent visual hallucina- with previous criteria. Erkinjuntti et al. (1997) found that
tions and spontaneous features of parkinsonism, for a diag- the prevalence of dementia in a large Canadian population
nosis of possible DLB, and two for probable DLB (McKeith cohort varied dramatically – almost tenfold – depending
et al., 2005), NIA-AA criteria require no fewer than two on which of the diagnostic criteria were applied; DSM-III
core features to exclude a diagnosis of probable AD, and so (29.1%), DSM-III-R (17.3%), DSM-IV (13.7%), ICD-9 (5.0%),
the same person can meet possible consensus DLB criteria the Cambridge Mental Disorders of the Elderly Examination
and probable NIA-AA AD criteria. Similar thresholds are (CAMDEX; 4.9%) and ICD-10 (3.1%). Furthermore, the six
illustrated by requirements for prominent features of behav- classification systems agreed a dementia diagnosis in only
ioural variant FTD or prominent features of primary pro- 20 of over 450 patients, suggesting not that some criteria
gressive aphasia. are more restrictive than others, but that different systems
38 Dementia

reflect different meanings of the dementia syndrome itself. individual cases. Dementia has moved from a broad concept
While the criteria probably have clinical utility when applied to a tightly defined cognitive syndrome due to one of sev-
flexibly, their application in a research setting requires more eral specific aetiologies, each with increasingly explicit and
standardization. In particular, alleged objective assessment detailed criteria that now often include biomarkers or com-
of functional impairment actually requires considerable plex investigations. Invariably, clinical practice will include
subjectivity on the part of the assessor, leaving broad scope patients who fail to fulfil these criteria, or indeed, those who
for differences in practice. Functioning is subject to a num- fulfil more than one set of criteria. Multiple disease contri-
ber of social and cultural factors, as well as patients’ and butions to dementia are increasingly likely with ageing, with
carers’ circumstances and expectations; an individual, for the oldest patients highly unlikely to have pure single dis-
example, living with a high degree of social support and low ease causes (Savva et al., 2009; Jellinger and Attems, 2010).
level of expectation will only meet functional criteria at a Mixed aetiologies, sensory or motor functional impairment
much more advanced level of impairment. Consideration and psychiatric or physical comorbidities further cloud the
of the multifaceted nature of factors like functional impair- diagnostic process, particularly when applied to an ageing
ments, and their significance in diagnosis and other clini- population.
cal decisions, are entirely acceptable in clinical practice, Diagnostic criteria like DSM-5 and NIA-AA have sought
but are likely to contribute towards the significant variation to include a degree of judgement of the skilled clinician
observed in prevalence studies. translating research findings to the assessment. The term
‘impairment’, used with respect to cognitive and functional
abilities, is necessarily vague, taking into account the broad
range of patients that professionals engage with in clinical
4.6 TRANSLATING DIAGNOSTIC
practice; indeed NIA-AA criteria explicitly state that ‘sig-
CRITERIA INTO THE CLINICAL
nificant interference’ in functioning is inherently a ‘clini-
ENVIRONMENT cal judgment made by a skilled clinician on the basis of the
individual circumstances of the patient’.
A number of factors make effective transfer of the concep- Current diagnostic criteria neither suggest suitable cog-
tual and biological knowledge underpinning the dementia nitive rating scales, nor ‘cut-off’ scores for diagnosis. While
criteria into the clinical environment difficult. Research DSM-5 does cite cognitive scores of less than 1.5 or 2 stan-
studies, including those contributing to diagnostic criteria, dard deviations below the mean, this figure must represent
maintain internal validity through application of exclusion ability when compared against populations of a similar age,
criteria and strict standardization of assessment, sometimes sex, education and background. It is implicit in this caveat
conducted by lay interviewers using structured interviews. that normative data will not always be available, particu-
However, this reduces their generalizability to typical clini- larly for those at either very low or very high levels of intel-
cal settings. Patients with dementia may be excluded from lectual ability, and that when assessing a patient who falls
studies due to comorbidities or diagnostic uncertainty, as outside ‘normal’ values for such parameters, clinicians must
might patients with very mild or very severe symptoms. employ their clinical judgment in consideration of cogni-
Such exclusion criteria are seldom applicable in clinical tive impairment. Clearly, where previously collected data
practice, with clinicians expected to diagnose a wider, older are available for comparison, decline can be demonstrated,
and more heterogeneous population, often with acute pre- but accurate information may not be at the clinician’s dis-
sentations of behavioural, medical or psychiatric symptoms. posal, even with the availability of a collateral history.
Thus, the role of the clinician may extend far beyond that Furthermore, in many parts of the world, everyday clinical
of diagnostician alone, and he or she will be aware of the assessments will not be able to utilize such standardized cri-
expectations and concerns of both patient and carer. The teria and will always depend on clinician judgement. While
focus of the clinical interaction is usually much more than formal neuropsychological testing is preferable in demon-
making a diagnosis, and involves engaging the patient and strating cognitive deficits, the availability of such services,
fostering a rapport with a view to further assessment and even in developed countries, can be limited. Accordingly,
investigation. In the case of primary care assessment, the NIA-AA is more realistic in stating that neuropsychologi-
relationship between the practitioner and patient and carer cal testing need only be employed when bedside testing and
may extend back several decades, providing an invaluable mental state examination is unable to provide a confident
insight into premorbid functioning and cognitive ability diagnosis.
seldom available to researchers, but may also come with Judgement of a decline in functional ability presents cli-
prejudice and perceived collusion with family members or nicians with similar scope for judgement. While research
carers in some cases. studies usually describe functional decline through scores
Subcategorization itself can introduce problems into in Bristol Activities of Daily Living Scale (Bucks et al., 1996)
clinical diagnosis. While biological findings have acted as or the Instrumental Activities of Daily Living Scale (Lawton
the main driver for the evolution of dementia-diagnostic and Brody, 1969), neither of these scales might reflect decline
criteria recently, a relative lack of scrutiny has been applied in more complex tasks perceived by patients and their car-
to the qualitative and environmental factors that influence ers. Similarly, use of these scales might fail to take into
What is dementia, and how do you assess it? Definitions, diagnostic criteria and assessment 39

account sociocultural nuances, like gender-assigned house- of interrelated issues, each requiring characterization of
hold tasks; it is common for tasks like cooking and cleaning its relationship with the clinical presentation. These may
to have been performed by a patient’s spouse throughout his include non-cognitive symptoms that, while not contrib-
adult life, and an inability to perform such tasks could sim- uting to dementia diagnosis itself, may help point toward
ply reflect a longstanding pattern rather than a manifesta- subtype and serve as a focus for symptomatic treatment.
tion of cognitive decline. Some activities may be impaired Nevertheless, the central diagnostic tenets of cognitive
due to physical disability, like visual impairment in the use decline associated with functional impairment are crucial
of telephone or electronic equipment. Where a patient no aspects of the interview, not only in relation to dementia
longer performs activities that were previously executed diagnosis but also in areas of monitoring and intervention.
with ease, external factors must also be considered; is non- Determining the course of symptoms can be invaluable
compliance with medication related to cognitive decline or in making subtype diagnosis but the insidious pattern of
unreported side effects? Might concerns over safety be influ- amnesia associated with AD can also be shared by DLB and
encing the use of public transportation? Such instances may subcortical VaD. The classical VaD presentation of sudden
require exploration of subtle decline in other functional onset dementia with stepwise deterioration is unusual in old
parameters, such as the patient’s ability to pursue hobbies age psychiatry services, as such patients more often present
and interests to a similar standard as before; the voracious to acute stroke services. A more rapid onset, over days or
reader no longer completing books at the same rate, the weeks, is consistent with delirium or depression and should
amateur musician who struggles to learn a new piece, or prompt further questioning to identify the presence of these
the casual gambler unable to complete their betting coupon syndromes. The presence of cognitive symptoms, although
may all be demonstrating evidence of functional impair- likely to be determined by cognitive testing, should also
ment that assessment scales fail to reflect. be inquired into. It is important to ask questions about
the behavioural and psychological symptoms of dementia
because these frequently cause greater distress and more
problems than cognitive symptoms. Furthermore, symp-
toms like visual hallucinations and sleep changes may be
4.7 CLINICAL ASSESSMENT OF A
suggestive of DLB (McKeith et al., 2005), or changes in
PATIENT PRESENTING WITH
social interactions and eating behaviour indicative of FTD
POSSIBLE DEMENTIA (Rascovsky et al., 2011), in turn indicating different man-
agement strategies.
The clinician conducting the assessment aims to achieve Identifying the presence of a decline in instrumental and
much more than a diagnosis; the assessment serves as a basic functioning that accompanies cognitive impairment
means of engaging the patient and their family to facilitate is not only an important component of diagnosis but also
further investigation, treatment and monitoring. The impor- a means of recognizing areas for further multidisciplinary
tance of establishing a rapport with both the patient and assessment and intervention. Structured assessment tools
carer is further underlined by the clinician’s role in discuss- such as Bristol Activities of Daily Living (Bucks et al., 1996)
ing the sensitive topic of dementia diagnosis, prognosis and can be helpful here, but assessment can be complicated by
management. Discussions will persist for years to come over varying levels of assistance, expectation and premorbid
the topic of dementia as a conceptual entity, but the demen- functioning and it is wise to consider these factors when
tia diagnosis is unquestionably important in clinical prac- appraising functioning.
tice; not only does diagnosis lead to access to treatments and A detailed account of both medical and psychiatric his-
services but also it encourages individuals to make impor- tory should be compiled, including recent operations and
tant decisions and plans at a stage in the disease process at other procedures. Particular attention should be paid to
which this is still feasible (National Institute for Clinical illnesses occurring shortly before, and since, the apparent
Excellence, 2006), and the clinician’s ability to impart such onset of symptoms suggestive of dementia. Evidence or risk
information is assisted by a warm rapport with the parties of vascular disease (stroke, transient ischaemic attacks, dia-
involved (Bamford et al., 2004). If the ongoing evolution betes, hypertension, falls) should be explored. It is wise to
of the dementia concept has taught us anything, it is that corroborate any list of medical or psychiatric conditions
exceptions to criteria exist, and that the application of core provided in the referral with the patient and/or their infor-
concepts to individual patients is a nuanced clinical skill mant. Similarly, reconciling the medication list provided
best conducted by an experienced practitioner, rather than a with the patient’s reported prescriptions can help determine
series of items on a checklist or administration of diagnostic compliance, the presence of non-prescribed and alternative
criteria. Thus, an accurate and detailed clinical history and treatments, and recent medication changes; in particular,
mental state examination, made by an experienced special- practitioners should seek to identify the presence of recently
ist, clarified by physical examination and special investiga- commenced medications that might precipitate or exacer-
tions, remains the cornerstone of the diagnostic process. bate cognitive impairment, such as anticholinergic agents.
Although one main problem may have been cited as the Recording aspects of patients’ personal history at the
reason for referral, assessment may uncover a constellation beginning of the interview can be helpful, as cognitively
40 Dementia

impaired patients often talk happily about their earlier life history in younger patients, such as the issue of substance
without being distressed by difficulties remembering recent misuse, can be administered. The subject of driving motor
events. A detailed personal history can be of considerable vehicles has become increasingly prominent in recent years;
use in determining the presence and severity of amnesia it is necessary to ask carers if concerns regarding driving
without the aid of cognitive testing, and identifying difficul- have arisen or if incidents have occurred, and the clinician
ties such as remembering the names of children and grand- may need to consider asking the patient to stop altogether
children can give an approximate indication of the point if concerns exist (see Chapter 31). Questions regarding the
at which the their memory began to fragment. Crucially, it patient’s property and financial arrangements are impor-
also helps understanding of the patient as a person. tant in establishing his or her risk of exploitation and in
In the absence of major childhood events, an account identifying future choices that they might face.
of patients’ happiness and friendships at school, and con-
tentedness at home, provide sufficient insight into early life.
It is inadvisable to infer intelligence from the age at which
an older patient left full-time education, given how socially 4.8 MENTAL STATE EXAMINATION
determined this was historically; indeed, it still is in many
places. An account of their further training and occupa- It is important to remember that symptoms reported in
tional history, focusing on positions maintained for the history may not be reflected in the patient’s current men-
longest duration, or holding particular significance for the tal state. The mental state assessment begins on arrival and
patient, can more accurately reflect premorbid intellectual continues throughout the consultation, adopting the same
ability. Clarifying the degree of responsibility or autonomy structure as that used in examination of younger adults.
held by the patient in these jobs may provide additional While cognitive assessment is the most essential component
insight. Marital history, focusing on the nature of the rela- of the mental state examination in the context of dementia
tionship with both the spouse, children and grandchildren, diagnosis, other clinical features both help refine differen-
as well as any other marriages and sexual relationships, can tial diagnosis and point toward aetiological subtype.
both give an insight into an individual’s personality as well
as the potential degree of familial support that the patient 4.8.1 APPEARANCE AND BEHAVIOUR
and carer may be able to access. Similarly, discussing the
patient’s response to stressors such as bereavements, illness Observation of the patient’s appearance can provide insight
or role transitions, allow information to be gathered about into the level of self-care and hygiene, general health, nutri-
the past that may help anticipate and address future chal- tional status and alertness. The clinician also should be alert
lenges, such as placement in residential care. While many of to the range of the behavioural signs, such as suspicious-
these stressors are often managed very well, recognized by ness, paranoia and poor eye contact. Psychomotor retar-
many as a part of growing old, they can precipitate affective dation, especially prominent in depressed older people,
illness and impact on other areas of the patient’s life; diffi- should also be considered in the context of the bradykine-
cultly adapting to retirement, for example, can create stress sia characteristic of Parkinson’s disease. The significance
at home and consequent marital disharmony in addition of parkinsonian symptoms in DLB diagnosis necessitates
to being a role transition and a life milestone. While direct careful consideration of these features throughout history
questioning regarding personality traits is unlikely to be and examination. It is prudent for every new patient to be
of significant use, the information derived throughout the assessed for bradykinesia, rigidity and gait changes, as well
history-taking process provides an insight into a patient’s as for the presence of focal neurological signs suggestive of
enduring pattern of relationships, activity and behaviours. stroke disease.
This may be difficult to distinguish from the effects of ill- Apathy is prevalent in even early dementia (Mega et al.,
ness in patients with chronic disease, but in those with a 1996), and is difficult but important to distinguish from
more recent diagnosis, premorbid personality may mould depression (Levy et al., 1998). The apathetic patient, though
the presentation of the illness; thus, the fiercely independent seemingly listless and disengaged when being discussed
patient may find adaptation at a ward or residential home with an informant, will ‘warm up’ when addressed directly,
more challenging than one more accustomed to seeking and in contrast to the depressed patient. When reporting a move
receiving assistance. away from previously enjoyed activities, apathetic patients
The patient’s social history should represent a detailed may describe this as a consequence of diminishing drive
account of their pattern of everyday life, activities and rela- to pursue affairs rather than a lack of enjoyment of them.
tionships, with the quality and quantity of care provided by As well as apathy, disinhibition, such as overfamiliarity, is
relatives, friends and formal carers explored in detail. This a feature of frontal dementias, and can be somewhat diffi-
may be supplemented by a written care plan giving insight cult to distinguish from the normal range of social interac-
into day-to-day issues as well as the programme of care, tion; watching the patient’s relatives’ reactions to his or her
which can be invaluable in identifying issues such as apa- behaviour, together with their collateral history can provide
thy or resistiveness. Having identified the pattern of living insight into whether such interactions represent a patholog-
and support, the more direct questions that comprise social ical change, or the patient’s normal behaviour.
What is dementia, and how do you assess it? Definitions, diagnostic criteria and assessment 41

4.8.2 SPEECH resulting from poor eyesight; however, since such problems
in visual processing commonly occur in DLB, the diagnosis
Dysphasia, a key feature of early dementing illness, can should be strongly considered, even when frank hallucina-
be a helpful clinical sign in distinguishing dementia from tions cannot be identified. Where a patient’s complex visual
depression, particularly as poverty of speech can be char- hallucinations are present as the sole phenomenological fea-
acteristic of both syndromes. However, receptive dyspha- ture, with no apparent cognitive impairment or alteration
sia can be difficult to discriminate from deafness when an in consciousness, a diagnosis of Charles Bonnet syndrome
apparent difficulty in understanding is reported or observed. may be appropriate. Although many such patients will
Similarly, the subtle difficulties in speech structure, such as decline cognitively, developing dementia and other features
in name-finding ability, while often present in mild demen- of DLB, not all do so. The similar phenomenon of auditory
tia, can be challenging to determine due to patients’ use of hallucinosis in the absence of cognitive decline, sometimes
circumlocutions to disguise them. called auditory Charles Bonnet syndrome, is often associ-
ated with sensorineural deafness.
4.8.3 MOOD
4.8.6 COGNITION
The clinician should be vigilant for affective symptoms.
Spontaneous, short-lived episodes of weeping or elevated Cognitive assessment is discussed in Chapters 5 and 6. A
mood should alert the practitioner to the possibility of the structured assessment tool can provide both an estimation
emotional lability associated with cerebrovascular disease of the severity of cognitive impairment and enable monitor-
(House et al., 1989). Elevated mood can be indicative of ing of progression or treatment response. However, effective
frontal dementias, and where present may be suggestive of cognitive assessment goes beyond assessment tools alone,
a more severe illness. Depressive illness, often accompanied considering inconsistencies and gaps in the patient’s history
by more anxiety than is observed in younger adults, should as possible evidence of amnesia, and detecting deficits in
be considered both amongst the differential diagnoses for orientation, cognition, spatial or executive dysfunction in
the cognitively impaired patient, and as a comorbid feature reported symptoms and observed signs.
of dementia.
4.8.7 INSIGHT
4.8.4 THOUGHT
Little formal insight is retained in moderate-to-severe
Paranoid delusions, particularly those of theft, and sev- dementia, but awareness that something is not right, and
eral variants of misidentification delusions are common that help is required, may be present; however, patients may
in dementia; Capgras syndrome (where a close relative have a greater awareness of their symptoms and illness than
is believed to have been replaced by an exact double), the they are capable of verbalizing. While greater insight may
‘mirror sign’ (where patients fail to recognize their image be retained in mild dementia, the degree to which this is
in a mirror, engaging with or becoming angry at the other acknowledged varies greatly. Insight into amnesia appears
party), the ‘TV sign’ (where the patient believes people seen greater than that into the impact of both functioning, and
on television are present in the room) and phantom boarder the impact of the disease on others.
syndrome (in which the patient believes extra people to be
living in the house), are all related to failure to recognize
or correctly identify an image. Incoherence of thought is
common in dementing illnesses, particularly in their later 4.9 PHYSICAL INVESTIGATIONS
stages, but formal thought disorder is much rarer than in
younger patients with mental illnesses. The characteristic physical signs in dementia aetiologies such
as DLB and FTD, together with those identified in reversible
4.8.5 PERCEPTION dementias, underline the importance of the physical exami-
nation in the diagnostic process. Brief observation itself may
The organic nature of illnesses that present to old age psy- identify the presence of tremors, choreoathetoid movements
chiatry services can result in prominent and persistent hal- or focal neurological deficits, as well as skin signs sugges-
lucinations in other sensory modalities to the auditory and tive of anaemia or jaundice. Cardiovascular and respiratory
olfactory hallucinations associated with schizophrenia, examination may ascertain the presence of pathology or
such as the visual and tactile hallucinations frequently seen risk factors for pathology such as hypertension, murmurs,
in patients with delirium. Complex and enduring visual rhythm disturbances, heart failure or orthostatic hypoten-
hallucinations, characteristically of people and animals, are sion. Abdominal examination can determine the presence
a core feature of DLB (McKeith et al., 2005), but delirium of liver stigmata or aortic aneurysm, while discomfort on
must also be considered. Effort should be made to distin- bladder palpation may identify pathology frequently associ-
guish whether the visual hallucinations occur in the absence ated with delirium, such as faecal loading, often precipitated
of a real stimulus or whether they represent misperception by anticholinergic or opiate agents.
42 Dementia

Abnormalities in posture, gait and movement should characteristic patterns for the different pathological
also be formally assessed, keeping in mind cerebellar ataxia, causes of dementia and molecular imaging of amyloid and
the shuffling gait classically associated with parkinsonism tau is likely to become increasingly used (see Chapters 10
and the broad-based gait of normal pressure hydrocephalus. through 12).
Tremors should be examined and characterized in an effort
to determine aetiology, considering the intention and static REFERENCES
tremors present in cerebellar disorders, the pill-rolling
tremor characteristic of parkinsonism and the action trem- Alzheimer, A. (1907). Uber eine eigenartige Erkrankung
ors exaggerated in thyrotoxicosis, uraemia and liver disease. der Hirnrinde. Allgemeine Zeitschrife Psychiatrie, 64:
The presence of facial weakness, abnormal eye movements, 146–148.
nystagmus, and visual field defects should be scrutinized as American Psychiatric Association (1952). Diagnostic and
a part of routine cranial nerve examination, as should oph- Statistical Manual of Mental Disorders. Washington,
thalmoscopy, which may identify the evidence of systemic DC: American Psychiatric Association.
disease like diabetes, hypertensive retinopathy or papilloe- American Psychiatric Association (1968). Diagnostic and
dema secondary to raised intracranial pressure. Statistical Manual of Mental Disorders, Second
Abnormalities in the pyramidal and the extra-pyramidal Edition. Washington, DC: American Psychiatric
motor systems may help confirm a diagnosis of VaD, DLB, Association.
Parkinson’s disease or amyotrophic lateral sclerosis; myoc- American Psychiatric Association (1980). Diagnostic and
lonus may be associated with epilepsy or Creutzfeldt–Jakob Statistical Manual of Mental Disorders, Third Edition.
disease. Sensory examination, though often difficult in Washington, DC: American Psychiatric Association.
patients with cognitive impairment, may reveal abnormali- American Psychiatric Association (1994). Diagnostic and
ties suggestive of cerebrovascular lesions, peripheral dia- Statistical Manual of Mental Disorders, Fourth Edition.
betic neuropathy or vitamin B12 deficiency, as well as the Washington, DC: American Psychiatric Association.
rarer, subacute combined degeneration of the spinal cord. American Psychiatric Association (2013). Diagnostic and
Reflexes can be similarly challenging to assess but the char- Statistical Manual of Mental Disorders, Fifth Edition.
acteristic acquisition of the primitive reflexes (grasp, suck- Washington, DC: American Psychiatric Association.
ing and snout reflexes) in frontal dementias can be a helpful Bamford, C., Lamont, S., Eccles, M. et al. (2004). Disclosing
diagnostic sign, otherwise occurring in advanced dementia. a diagnosis of dementia: A systematic review.
The choice of physical investigations will be guided by International Journal of Geriatric Psychiatry, 19 (2):
differential diagnosis, resource availability, patient and 151–169.
carer preference and tolerability and local and national Boller, F. and Forbes, M.M. (1998). History of dementia
guidelines. Blood tests, including full blood count, erythro- and dementia in history: An overview. Journal of the
cyte sedimentation rate (ESR) or C-reactive protein (CRP), Neurological Sciences, 158 (2): 125–133.
renal function, thyroid function, calcium, glucose, vitamin Bucks, R.S., Ashworth, D.L., Wilcock, G.K. and Siegfried, K.
B12, serum folate and liver function tests are often con- (1996). Assessment of activities of daily living in
ducted prior to secondary referral as a means of excluding dementia: Development of the Bristol Activities of
systemic illnesses that might cause, or exacerbate, cogni- Daily Living Scale. Age and Ageing, 25 (2): 113–120.
tive impairment. Other tests, such as fasting lipids, syphilis Corsellis, J.A.N. and Evans, P.H. (1965). The relation of
serology, HIV testing, mid-stream urine, chest X-ray and stenosis of the extracranial cerebral arteries to mental
electrocardiography are used if indicated by clinical find- disorder and cerebral degeneration in old age. In
ings, rather than forming part of an initial screen. Luhy, F. and Bischoff, A. (eds.), Proceedings of the
Structural imaging, using computed tomography (CT) Vth International Congress of Neuropathology.
or magnetic resonance imaging (MRI), is recommended Amsterdam, the Netherlands: Excerpta Medica.
by many authorities (National Institute for Clinical Dubois, B., Feldman, H.H., Jacaova, C. et al. (2007).
Excellence, 2006), partly for exclusion of intracranial Research criteria for the diagnosis of Alzheimer’s
pathology like tumours, normal pressure hydrocephalus disease: Revising the NINCDS–ADRDA criteria. The
and subdural haematomas. MRI provides good views of Lancet Neurology, 6 (8): 734–746.
the hippocampi, allowing the identification of the atrophy Erkinjuntti, T., Ostbye, T., Steenhuis, R. and Hachinski, V.
characteristic of AD. Modern CT scanners can provide (1997). The effect of different diagnostic criteria on
coronal reconstructions of the hippocampi to help in the the prevalence of dementia. New England Journal of
same way. MRI can also be helpful in detecting more sub- Medicine, 337 (23): 1667–1674.
tle changes, like frontal lobe atrophy, suggestive of FTD, Escorpizo, R., Kostanjsek, N., Kennedy, C. et al. (2013).
and especially of cerebrovascular disease in the white mat- Harmonizing WHO’s International Classification of
ter (lacunar infarcts and ischaemic white matter disease). Diseases (ICD) and International Classification of
Functional perfusion imaging with single photon emis- Functioning, Disability and Health (ICF): Importance
sion computed tomography (SPECT) and positron emis- and methods to link disease and functioning. BMC
sion tomography (PET), where available, can highlight Public Health, 13 (1): 1–5.
What is dementia, and how do you assess it? Definitions, diagnostic criteria and assessment 43

Fox, H.A. (1983). The DSM-III concept of organic brain McKeith, I.G., Dickson, D.W., Lowe, J. et al. (2005).
syndrome. The British Journal of Psychiatry, 142 (4): Diagnosis and management of dementia with
419–421. Lewy bodies third report of the DLB consortium.
Ganguli, M., Blacker, D.G., Blazer, D.G. et al. (2011). Neurology, 65 (12): 1863–1872.
Classification of neurocognitive disorders in DSM-5: McKhann, G.M., Knopman, D.S., Chertkow, H. et al.
A work in progress. The American Journal of Geriatric (2011). The diagnosis of dementia due to
Psychiatry, 19 (3): 205–210. Alzheimer’s ­disease: Recommendations from
George, D.R., Whitehouse, P.J. and Ballenger, J. the National Institute on Aging-Alzheimer’s
(2011). The evolving classification of dementia: Association ­workgroups on diagnostic guidelines
Placing the DSM-V in a meaningful historical for Alzheimer’s disease. Alzheimer’s & Dementia,
and cultural context and pondering the future of 7 (3): 263–269.
“Alzheimer’s”. Culture, Medicine, and Psychiatry, 35 Mega, M.S., Cummings, J.L., Fiorello, T. and Gornbein, J.
(3): 417–435. (1996). The spectrum of behavioral changes in
House, A., Dennis, M., Molyneux, A. et al. (1989). Alzheimer’s disease. Neurology, 46 (1): 130–135.
Emotionalism after stroke. BMJ: British Medical National Institute for Clinical Excellence (2006). Dementia:
Journal, 298 (6679): 991. Supporting People with Dementia and Their Carers in
Jellinger, K.A. and Attems, J. (2010). Prevalence of demen- Health and Social Care CG42. London: NICE.
tia disorders in the oldest-old: An autopsy study. Acta Rascovsky, K., Hodges, J.R., Knopman, D. et al. (2011).
Neuropathologica, 119(4): 421–433. Sensitivity of revised diagnostic criteria for the behav-
Knapp, M., Guerchet, M., McCrone, P. et al. (2007). ioural variant of frontotemporal dementia. Brain, 134
Dementia UK: A Report to the Alzheimer’s Society (9): 2456–2477.
on the Prevalence and Economic Cost of Dementia Reisberg, B. (2006). Diagnostic criteria in dementia: A com-
in the UK Produced by King’s College London and parison of current criteria, research challenges, and
London School of Economics. London: Alzheimer’s implications for DSM-V. Journal of Geriatric Psychiatry
Society. and Neurology, 19 (3): 137–146.
Knopman, D.S., Petersen, R.C., Edland, S.D. et al. (2004). Savva, G.M., Wharton, S. B., Ince, P. G. et al. (2009). Age,
The incidence of frontotemporal lobar degenera- neuropathology, and dementia. New England Journal
tion in Rochester, Minnesota, 1990 through 1994. of Medicine, 360 (22): 2302–2309.
Neurology, 62 (3): 506–508. World Health Organization (1974). Glossary of Mental
Kraepelin, E. (1899). Psychiatrie. Ein Lehrbuch für Disorders and Guide to Their Classification: For Use
Studirende und Ärzte, 6. Auflage, Band II: Klinische in Conjunction With the International Classification
Psychiatrie. Leipzig,Germany: Barth. of Diseases, 8th Revision. Geneva: World Health
Kraepelin, E. (1910). Lehrbuch der Psychiatrie, 8 Auflage. Organization.
Leipzig,Germany: Barth. World Health Organization (1978). Mental Disorders:
Kurz, A.F. and Lautenschlager, N.T. (2013). A brief history of Glossary and Guide to Their Classification in
the concept of dementia. In T. Dening and A. Thomas Accordance Wwith the 9th Revision of the
(eds.), Oxford Textbook of Old Age Psychiatry. International Classification of Diseases. Geneva:
Oxford: Oxford University Press. World Health Organization.
Lawton, M.P. and Brody, E.M. (1969). Assessment of older World Health Organization (1992). The ICD-10
people: Self-maintaining and instrumental activities of Classification of Mental and Behavioural Disorders:
daily living. The Gerontologist, 9 (3): 179. Clinical Descriptions and Diagnostic Guidelines.
Levy, M.L., Cummings, J.L., Fairbanks, L.A. et al. Geneva: World Health Organization.
(1998). Apathy is not depression. The Journal of World Health Organization (2001). International
Neuropsychiatry and Clinical Neurosciences, 10 (3): Classification of Functioning, Disability and Health
314–319. (ICF). Geneva: World Health Organization.
5
Screening and assessment instruments for
the detection and measurement of cognitive
impairment

LEON FLICKER

5.1 INTRODUCTION 5.2 DESCRIPTION OF TESTS

A frequently used method in the ascertainment of demen- The short cognitive tests have been used in case finding for
tia in older people is the performance of a short cognitive over 50 years. A minor change that has occurred in the more
test to determine the need to perform further testing to recently developed tests is the use of lengthier delayed recall
establish the diagnosis. The case for detection of milder tasks, both cued and non-cued. One of the earliest instru-
forms of cognitive impairment has not been established, ments was the Mental Status Questionnaire (MSQ) (Kahn
and therapeutic strategies targeted for patients with condi- et al., 1960). In 1968, Blessed, Tomlinson and Roth described
tions such as mild cognitive impairment (MCI) (Petersen the Information Memory Concentration (IMC) test, which
et al., 2001) have not been developed. Another use for was validated against the criterion of neuropathology.
these tests is to detect changes in cognitive impairments Hodkinson (1972) described a shortened version of the IMC
over time. Thus, the focus of this chapter remains on the test and called this the Abbreviated Mental Test (AMT)
use of short cognitive tests to aid in the detection of older score. In North America, Pfeiffer (1975) described the Short
people with dementia, and to monitor the progression Portable Mental Status Questionnaire (SPMSQ) and Folstein
of cognitive impairment in people with dementia. These et al. (1975) devised the Mini-Mental Status Examination
short cognitive tests are commonly described as ‘screening (MMSE), which has undergone standardization (Molloy
instruments’, but this is in fact a misnomer. Screening has et al., 1991). Subsequently, additional questions were added
been defined as ‘An organized attempt to detect, among to the MMSE and a modified scoring system was introduced
apparently healthy people in the community, disorders to produce a test called the 3MS (Teng and Chui, 1987).
or risk factors of which they are unaware’ (Cadman et al., Other short tests that have been developed include Kokmen’s
1984). The U.S. Preventive Services Task Force (Moyer Short Test of Mental Status (STMS) (Kokmen et al., 1991), the
et al., 2014) has concluded yet again that the evidence is Mini-Cog (Borson et al., 2000) and the General Practitioner
insufficient to recommend for, or against, routine screen- Assessment of Cognition (GPCOG) (Brodaty et al., 2002)
ing for cognitive impairment in older adults. However, all of which include the clock drawing tests. A short test has
these short cognitive tests are used as a means of case been developed, which does not utilize orientation or other
finding in certain clinical situations for which there is a questions prone to cultural bias, the Rowland Universal
high prior probability of finding individuals with cogni- Dementia Assessment Scale (RUDAS). All these instruments
tive impairment, usually associated with dementia or in have in common their brevity, taking 10 minutes or less to
those individuals who are medically unwell, delirium. An administer with relatively little training.
example of this situation is acute hospital admission for One of the main problems with these cognitive tests
people over the age of 65 years (Ames and Tuckwell, 1994). is the fact that they exhibit both floor and ceiling effects.

44
Screening and assessment instruments for the detection and measurement of cognitive impairment 45

There have been attempts to develop tests that would be these tests applied, and possibly all patients over the age
more suitable for testing impaired patients (Plutchik et al., of 65 years who present acutely to hospital (Australian
1971; Albert and Cohen, 1992). Longer tests and word lists Commission on Safety and Quality in Health Care, 2015).
have the advantage that they are less prone to ceiling effects. Even before the arrival of specific symptomatic treat-
Word list instruments generally require some training. ments for the symptoms of Alzheimer’s disease, the pres-
These include the 7-minute screen (Solomon et al., 1998), ence or absence of significant cognitive impairment had
which is a test that comprises a cued recall task, verbal flu- major implications for the management of patients. In par-
ency, an orientation test and a clock drawing task. Similarly, ticular, older individuals with multiple medical problems
the Syndrom Kurztest (SKT) (Lehfeld and Erzigkeit, 1997) requiring medications could not be managed appropriately
has nine subtests, which include naming, memory, atten- without an assessment for the presence of cognitive impair-
tion, cued recall and visuospatial functioning. A very short ment and some idea of its severity. Also, there is some evi-
version of delayed free and cued recall tests has been devel- dence that the earlier identification of people with dementia
oped called the Memory Impairment Screen (Buschke et al., and appropriate referral to support services and counselling
1999), which consists of just four items. Another test, the can alleviate some of the stresses associated with caring for
Montreal Cognitive Assessment (MoCA) (Nasreddine et al., people with dementia and delay institutionalization (Green
2005), was recently developed for the purpose of detecting and Brodaty, 2002).
MCI. It incorporates an adjustment for education level, with These short cognitive tests have limited domains of
those people scoring less than 26 classified as abnormal. It measurement. Table 5.1 refers to the domains of measure-
also has included the clock drawing test. It has also been ment of the common assessment tools. Orientation figures
used for the detection of people with dementia but at the prominently in most of these tools and is a reflection of
usual cut-off seems more sensitive but less specific (Larner, recent memory acquisition. In a longitudinal study of cog-
2012). There have been relatively few validity studies at this nitive function in people with Alzheimer’s disease, it was
stage. demonstrated that dementing processes particularly affect
Several other survey instruments have been devel- remote memory, immediate memory and language function
oped, which have incorporated both cognitive screen- (Flicker et al., 1993). Since memory impairment is a sine qua
ing instruments and diagnostic schedules. These include non in the categorization of dementia in both the Diagnostic
the Cambridge Diagnostic Examination for the Elderly and Statistical Manual of Mental Disorders, Fourth Edition
(Roth et al., 1986), Consortium to Establish a Registry (DSM-IV) and ICD-10 criteria it is hardly surprising that the
for Alzheimer’s Disease (Morris et al., 1989), Geriatric short cognitive tests focus on this domain. More recently,
Mental State schedule (Copeland et al., 1976) and a Diagnostic and Statistical Manual of Mental Disorders, Fifth
Structured Interview for the Diagnoses of Dementia of Edition (DSM-V) no longer specifies the requirement for
the Alzheimer’s type multi-infarct dementia (SIDAM) memory impairment for the classification of major neuro-
and dementias of other aetiology according to the cognitive disorder but rather a decline in one or more cog-
International Classification of Diseases, 10th Edition nitive domains (American Psychiatric Association, 2013).
(ICD-10) and Diagnostic and Statistical Manual of Exactly how this will be operationalized clinically is still
Mental Disorders, Third Edition, Revised (DSM-III-R) uncertain.
(Zaudig et al., 1991). The cognitive components of the It is clear that these tests are highly correlated with each
Cambridge Diagnostic Examination for the Elderly (Roth other. For example, Stuss et al. (1996) observed that the pro-
et al., 1986), the Alzheimer’s Disease Assessment Scale- portion of variance accounted for by a single common com-
Cognition (ADAS-Cog) (Rosen et al., 1984) and the ponent was in excess of 0.80 for the Mattis Dementia Rating
Mental Deterioration Battery (Carlesimo et al., 1996) take Scale, an abbreviated six-item version of the Orientation
longer to administer and are not usually performed as ini- Memory Concentration test adapted from Blessed et al.
tial brief tests, although the Organic Brain Scale (OBS) (1968) a Mental State Questionnaire and an Ottawa mental
from the Geriatric Mental State has been used as a stand- state examination. Also in this study there was no added
alone test (Ames et al., 1992). benefit from longer tests and the shorter cognitive tests
performed as well as longer tests with multiple domains. In
another study, the correlation between the AMT score and
MMSE was greater than 0.85 (Flicker et al., 1997). Similarly,
5.3 USE FOR THESE TESTS correlations between the MMSE and the ADAS-Cog have
been found to be strong, –0.9 (Burch and Andrews, 1987),
The need for these ‘screening’ instruments has arisen although the correlation between the MMSE and the STMS
because there is good evidence that clinicians will com- was only 0.74 (Kokmen et al., 1991). In a comparison of the
monly miss dementia in their routine practice without the MMSE and 3MS, both the modified scoring system and
assistance of formal cognitive assessment (Williamson the additional questions of the 3MS increased its discrimi-
et al., 1964; Mant et al., 1988; Valcour et al., 2000). It natory ability, but at the expense of increased burden of
remains a clearly defined guideline that for those individu- administration time and training requirements (McDowell
als suspected of a cognitive disorder should have one of et al., 1997).
46 Dementia

Table 5.1 Cognitive domains in commonly used short cognitive tests

Visuospatial
Personal Short-Term Long-Term Visuo-
Test Information Orientation Memory Memory Attention Naming Construction Other
MSQ ♦ ♦ ♦
IMC ♦ ♦ ♦ ♦ ♦ ♦
AMT ♦ ♦ ♦ ♦ ♦ ♦
MMSE ♦ ♦ ♦ ♦ ♦ ♦
3MS ♦ ♦ ♦ ♦ ♦ ♦ ♦
SPMSQ ♦ ♦ ♦ ♦
OBS ♦ ♦ ♦ ♦ ♦
STMS ♦ ♦ ♦ ♦ ♦ ♦
GPCOG ♦ ♦ ♦
TICS ♦ ♦ ♦ ♦ ♦ ♦ ♦
MINI-COG ♦ ♦
RUDAS ♦ ♦ ♦
MoCA ♦ ♦ ♦ ♦ ♦ ♦
Abbreviations: MSQ, Mental Status Questionnaire; IMC, Information Memory Concentration; AMT, Abbreviated Mental Test; MMSE, Mini-
Mental Status Examination; 3MS, modified Mini-Mental Status Examination; SPMSQ, Short Portable Mental Status Questionnaire;
OBS, Organic Brain Scale; STMS, Short Test of Mental Status; GPCOG, General Practitioner Assessment of Cognition; TICS, Telephone
Interview for Cognitive Status; MINI-COG; RUDAS, Rowland Universal Dementia Assessment Scale; MoCA, Montreal Cognitive
Assessment.

One of the major advances in the last 5 years has been The use of self-administered tests has been comprehensively
the standardization of the methodology for reporting of reviewed (Cherbuin et al., 2008) with some promising results
the validity of these tests (Noel-Storr et al., 2014). This will for the informant-based tests, but otherwise this method of
allow the systematic review of the validity of these cogni- administration was not recommended. There is evidence that
tive tests over many settings. The Cochrane Group has the place of testing can cause significant changes in the test
recently committed to reviews of Diagnostic Test Accuracy score, with testing in a patient’s own residence being associ-
for many of these cognitive tests (Davis et al., 2013), and ated with a higher score (Ward et al., 1990).
other tests including biomarkers, which will allow compari- These short cognitive screens obviously have their limi-
son of clinical utility across differing approaches. Using this tations, the most obvious being that they test only limited
standardized methodology, the MMSE did not seem to per- domains of cognition. Besides the use of full neuropsycho-
form well for detecting those patients who converted from logical assessment, attempts have been made to develop
MCI to dementia with sensitivities ranging from 23% to other short tests of specific functions. An example of this is
76% and specificities from 40% to 94% (Arevalo-Rodriguez the executive interview (Royall et al., 1992), which attempts
et al., 2015). On the other hand, the Mini-Cog when used for to assess executive cognitive function. Also, the ability to
the diagnosis of dementia within a community setting had draw a clock has been extensively studied as a short test,
sensitivities in the individual studies reported as 0.99, 0.76 and which examines other cognitive domains. It has been used
0.99 with specificities of 0.93, 0.89 and 0.83 (Fage et al., 2015). by itself or in conjunction with other cognitive tests such
These short cognitive scales appear robust in many set- as the SPMSQ or MMSE, or as part of the 7-minute screen,
tings. The IMC test has been validated on telephone inter- GPCOG or STMS. Clock drawing may miss a quarter of
view (Kawas et al., 1995) as has the Telephone Cognitive cognitively impaired individuals (Gruber et al., 1997).
Assessment Battery (Debanne et al., 1997), and the telephone Clock drawing may be able to detect cognitive alterations
version of the MMSE (Roccaforte et al., 1992). The Telephone not related to delirium or dementia, with abnormal clock
Interview for Cognitive Status (TICS) (Brandt et al., 1988) is drawing being associated with other psychiatric disorders
also a widely used instrument and because the word list is (Gruber et al., 1997). Also it may provide additional infor-
capable of delineating subjects who may have a memory dis- mation to the MMSE as suggested by Ferrucci et al. (1996),
order, which does not fulfil criteria for dementia, is able to where the clock drawing test predicted cognitive decline in
identify conditions such as amnestic MCI. The instruments older people independently of the MMSE.
have been used across national boundaries and have wide Short cognitive tests are known to be sensitive to bias
applicability, e.g. the OBS has been found to have good validity associated with education, but adjustment for this effect
in three separate countries (Ames et al., 1992). Some attempt does not necessarily improve test performance (Belle et al.,
has been made to develop self-administered tests, such as the 1996). For this reason, the RUDAS (Storey et al., 2004) was
Early Assessment Self Inventory (EASI) (Horn et al., 1989). developed, which has attempted to be less culturally biased
Screening and assessment instruments for the detection and measurement of cognitive impairment 47

and uninfluenced by educational attainment. Special tests in the choice of informants as much of the sample described
may also be required in specific situations such as with by Ritchie and Fuhrer (1992) were recruited through France
indigenous groups (LoGiudice et al., 2006). Alzheimer and may have represented carers that were well
Another consideration is that these tests may not be sen- attuned to the problems associated with dementia, while in
sitive to all stages of the disease. Stern et al. (1994) dem- the other study (Flicker et al., 1997) informants were drawn
onstrated that there was a quadratic relationship with from attendees at a memory clinic.
dementia severity showing that the ADAS-Cog and IMC There have been three Cochrane reviews of the IQCODE
test show greater deterioration for patients with moderate over diverse settings. For community dwelling populations,
dementia and that the deterioration is slower for mildly and at a threshold of 3.3, the sensitivity was 0.80 (95% confidence
severely demented patients. This may represent the relative interval [CI] 0.75–0.85) and specificity was 0.84 (95% CI
insensitivity of the tests for patients with the least and great- 0.78–0.90) (Quinn et al., 2014). For the primary care setting,
est impairment, or may reveal true patterns of progressive at a threshold of 3.2, sensitivity was 100% and specificity
cognitive decline at different parts of the disease process. was 76%, at a threshold of 3.7 sensitivity was 75%, and speci-
Attempts have also been made to shorten these tests even ficity was 98% (Harrison et al., 2014). For a secondary care
further – for example, the 10-item AMT has been further setting at a threshold of 3.3, the sensitivity was 0.91 (95%
shortened to four items (Swain and Nightingale, 1997), but it CI 0.86–0.94) and specificity was 0.66 (95% CI 0.56–0.75)
does seem to lose some predictive efficiency at this length. The (Harrison et al., 2015).
potential problem of inadequate specificity of short cognitive A meta-analysis (Jorm, 1997) suggests that informant
tests for population screening has been revealed in a commu- tests may perform better than short cognitive tests, but it is
nity study: the Canadian Study of Health in Ageing (Graham important to emphasize that there was great heterogeneity
et al., 1996). In this study, the 3MS was used as a community within this meta-analysis and clearly the test results were
screen before a diagnostic battery. Cognitive impairment, not dependent on the subject samples and the samples of infor-
dementia, was found to be more common than dementing mants. A subsequent meta-analysis confirmed relative inde-
processes, with the prevalence of cognitive impairment not pendence of education and premorbid ability but also that
dementia found to be 16.8%, whereas the prevalence of all these type of tests may be biased by depression and anxiety
types of dementia combined was 8% (Graham et al., 1997). in the informant and the quality of the relationship between
The conditions identified within this category of cognitive the informant and the subject (Jorm, 2004). Harwood et al.
impairment included delirium, alcohol use, drug intoxica- (1997) also demonstrated comparable discriminatory abil-
tion, depression, psychiatric disorders, memory impairment ity of the AMT and the IQCODE in a sample of medical
associated with the ageing process and intellectual disability. inpatients over the age of 65 admitted to a general medical
unit. In that study, the 16 item version of the IQCODE was
utilized with good effect. The IQCODE has been validated
by longitudinal changes of cognitive tests (Jorm et al., 1996)
5.4 INFORMANT-BASED TESTS and also has been validated retrospectively against post-
mortem diagnosis (Thomas et al., 1994).
Perhaps the most exciting development in the detection One of the important considerations for the use of com-
of cognitive impairment in older people in recent times binations of tests or additional items is that the correla-
has been the refinement and validation of structured tests tion between individual tests should not be too great. The
administered to informants. Examples of this type of test importance of asymmetry of associations between tests to
include the DECO test – Deterioration Cognitive Observée increase the positive predictive value of combinations of
(Ritchie and Fuhrer, 1992) and the Informant Questionnaire tests has been highlighted (Marshall, 1989). Hooijer et al.
for Cognitive Decline in the Elderly (IQCODE) (Jorm and (1993) found some improvement in diagnosis in using pairs
Korten, 1988). Important advantages of these tests are that of short screening tests. This was not the case in work by
they correlate relatively poorly with premorbid function Little et al. (1987), where a short AMT score was found to
and are relatively insensitive to the effects of education. have better predictive value than longer combined tests.
The other consideration about informant questionnaires To date, most work has focused on using either infor-
is that they have shown lower correlations with the cogni- mant tests or short cognitive tests separately as a screen for
tive tests: somewhere in the order of 0.6 (Flicker et al., 1997). further investigations and management, but clearly the tests
This is an important consideration in that their relatively low can be combined. This was raised by Flicker et al. (1997),
correlation with cognitive testing per se implies they provide where the lower correlations and asymmetry of the test
additional independent information, which may assist in the properties within different populations could be utilized so
discrimination of people with functionally important cog- that the combined tests’ sensitivity and specificity would be
nitive impairment. The work by Ritchie and Fuhrer (1992) improved. This is quite different to the use of the alteration
would suggest that they seem to be better able to discrimi- of cut-points, where a trade-off occurs between sensitiv-
nate mild dementia from normality as opposed to cognitive ity and specificity. The judicious use of the combination of
screens such as the MMSE, although this was not replicated informant and short cognitive tests potentially may result in
by others (Flicker et al., 1997). This may represent a difference an increase in both sensitivity and specificity. The tests can
48 Dementia

be applied so that for the combination to be positive both


tests are required to be positive, which is called ‘in series’. 5.6 CONCLUSIONS
Alternatively, the combination of tests could be judged posi-
tive if either test is positive, which is called in ‘parallel’. There is now available a plethora of short cognitive tests, which
This has been demonstrated by Gallo and Breitner (1995) appear useful in helping clinicians to determine whether
where a telephone administered cognitive status was used as individuals require further assessment for the presence of a
a first screen followed by an informant test. The informant cognitive disorder. The tests seem to perform in a similar fash-
test was falsely described as being more specific, but in fact ion. The characteristics of the population in which the tests
it was the combination of using the two tests in series that are used, seem to be at least as important to performance as
resulted in the increased specificity needed for this two- the test themselves. The addition of informant test informa-
stage screen. Another example of this approach being used tion appears more valuable rather than increasing the length
in challenging population research is the work of Hall et al. and complexity of the cognitive screen. It is difficult to predict
(1993, 1996). In these studies, a combination of short cogni- the performance of these tests, either singly, or in combina-
tive tests and informant testing was used in cross-cultural tion with informant tests, from one population to the next.
studies of African American, Cree Indian and Nigerian An improvement in the efficacy of cognitive assessments can
populations, and the relative insensitivity of the informant be most easily achieved by combining informant and short
tests to the effects of culture and education improved the cognitive tests to a single summative score, or using infor-
performance of these combinations. The combination of the mant and cognitive tests with specific cut-points and rules of
MMSE and IQCODE has now been evaluated extensively. combination. The most important limiting factor in clinical
In three separate studies (Mackinnon and Mulligan, 1998; practice is encouraging clinicians to use any one of these tests.
Knafelc et al., 2003; Mackinnon et al., 2003), the total evi- If clinical suspicion remains the combination of both, a short
dence suggested that the combination of tests performed cognitive test with an informant test is recommended.
better than either test alone, and that the tests could be
combined usefully by either a requirement for both or either
test to be positive (in parallel) or calculating a new score REFERENCES
using a weighted sum rule.
Besides the characteristics of the population being Albert, M. and Cohen, C. (1992). The test for severe
assessed, it is important to realize that the absolute preva- impairment: An instrument for the assessment of
lence of the condition will also cause a need for an adjust- patients with severe cognitive dysfunction. Journal of
ment in the cut-points in both the cognitive and informant the American Geriatrics Society, 40: 449–453.
tests. Very low prevalence will increase the need for speci- American Psychiatric Association (2013). Desk Reference
ficity; otherwise, there will be a large number of patients to for the Diagnostic Criteria from DSM5. Arlington, VA:
further investigate. This is necessitated in both research and American Psychiatric Association.
clinical practice. It is important to emphasize that the cut- Ames, D., Ashby, D., Flicker, L. et al. (1992). Jim who?
points used in the combination of these tests in either series Recall of national leaders by elderly people in
or parallel, may be quite different to the ones normally three countries. International Journal of Geriatric
employed by the tests in isolation. Psychiatry, 7: 437–442.
Ames, D. and Tuckwell, V. (1994). Psychiatric disorders
among elderly patients in a general hospital. Medical
Journal of Australia, 160: 671–674.
5.5 COSTS OF THE TESTS Arevalo-Rodriguez, I., Smailagic, N., Roqué, I. et al. (2015).
Mini-Mental State Examination (MMSE) for the
Until relatively recently, these tests have generally been detection of Alzheimer’s disease and other demen-
administered freely without charge. The major expense has tias in people with mild cognitive impairment (MCI).
been that of the clinicians’ time, both in training and admin- The Cochrane Database Systematic Reviews, 3:
istration. Two commonly used instruments, the MMSE and CD010783.
TICS are now licensed for use by Psychological Assessment Australian Commission on Safety and Quality in Health Care.
Resources (Inc.). The MMSE may be administered free of (2015). A better way to care: Safe and high-quality care
charge from memory or from an original paper (Folstein for patients with cognitive impairment (dementia and
et al., 1975) or from a source authorized by Psychological delirium) in hospital. Available at http://www.safetyan-
Assessment Resources. Otherwise, there is a charge for using dquality.gov.au/our-work/­cognitive-impairment/better-
the score sheets obtained from Psychological Assessment way-to-care. Accessed 18 May, 2015.
Resources. Similar charges apply to the TICS. It would seem Belle, S.H., Seaberg, E.C., Ganguli, M. et al. (1996). Effect
a pity if these proprietary interests inhibit the use of these of education and gender adjustment on the sensitiv-
short tests as often the major obstacle to the initial diagnosis ity and specificity of a cognitive screening battery
of dementia is applying one of these short tests when there for dementia: Results from the MoVIES project.
is clinical concern. Neuroepidemiology, 15: 321–329.
Screening and assessment instruments for the detection and measurement of cognitive impairment 49

Blessed, G., Tomlinson, B.E. and Roth, M. (1968). The Ferrucci, L., Cecchi, F., Guralnik, J.M. et al. (1996). Does
association between quantitative measures of the clock drawing test predict cognitive decline in
dementia and of senile change in the cerebral older persons independent of the Mini-Mental State
grey matter of elderly subjects. British Journal of Examination? The FINE study group, Finland, Italy,
Psychiatry, 114: 797–811. The Netherlands Elderly. Journal of the American
Borson, S., Scanlan, J., Brush, M. et al. (2000). The Mini- Geriatrics Society, 44: 1326–1331.
Cog: A cognitive ‘vital signs’ measure for dementia Flicker, C., Ferris, S.H. and Reisberg, B. (1993). A two year
screening in multi-lingual elderly. International Journal longitudinal study of cognitive function in normal
of Geriatric Psychiatry, 15: 1021–1027. ageing and Alzheimer’s disease. Journal of Geriatric
Brandt, J., Spencer, M. and Folstein, M. (1988). Psychiatry and Neurology, 6: 84–96.
The telephone interview for cognitive status. Flicker, L., Logiudice, D., Carlin, J.B. and Ames, D. (1997).
Neuropsychiatry Neuropsychology Behavioral The predictive value of dementia screening instru-
Neurology, 1: 111–117. ments in clinical populations. International Journal of
Brodaty, H., Pond, D., Kemp, N.M. et al. (2002). The Geriatric Psychiatry, 12: 203–209.
GPCOG: A new screening test for dementia designed Folstein, M.F., Folstein, S.E. and McHugh, P.R. (1975).
for general practice. Journal of the American Mini-mental state: A practical method for grading the
Geriatrics Society, 50: 530–534. cognitive state of patients for the clinician. Journal of
Burch, E.A. Jr. and Andrews, S.R. (1987). Comparison of Psychiatric Research, 12: 189–198.
two cognitive rating scales in medically ill patients. Gallo, J.J. and Breitner, J.C. (1995). Alzheimer’s disease
International Journal of Psychiatry in Medicine, 17: in the NAS-NRC Registry of aging twin veterans, IV.
193–200. Performance characteristics of a two-stage telephone
Buschke, H., Kuslansky, G., Katz, M. et al. (1999). Screening screening procedure for Alzheimer’s dementia.
for dementia with the Memory Impairment Screen. Psychological Medicine, 25: 1211–1219.
Neurology, 52: 231–238. Graham, J.E., Rockwood, K., Beattie, B.L. et al. (1996).
Cadman, D., Chambers, L., Feldman, W. and Sackett, D. Standardization of the diagnosis of demen-
(1984). Assessing the effectiveness of community tia in the Canadian study of health and ageing.
screening programs. Journal of the American Medical Neuroepidemiology, 15: 246–256.
Association, 251: 1580–1585. Graham, J.E., Rockwood, K., Beattie, B.L. et al. (1997).
Carlesimo, A., Caltagirone, C., Gainotti, G. et al. (1996). Prevalence and severity of cognitive impairment
The mental deterioration battery: Normative data, with and without dementia in an elderly population.
diagnostic reliability and qualitative analyses of Lancet, 349: 1793–1796.
­cognitive impairment. European Neurology, 36: Green, A. and Brodaty, H. (2002). Care-giver interven-
378–384. tions. In N. Qizilbash (ed.), Evidence Based Dementia
Cherbuin, N., Anstey, K.J. and Lipnicki, D.M. (2008). Practice. Oxford, United Kingdom: Blackwell Science,
Screening for dementia: A review of self- and pp. 764–794.
informant-assessment instruments. International Gruber, N.P., Varner, R.V., Chen, Y.W. and Lesser,
Psychogeriatrics, 20: 431–458. J.M. (1997). A comparison of the clock drawing
Copeland, J.R.M., Kelleher, M.J., Kellett, J.M. et al. (1976). test and the Pfeiffer Short Portable Mental
A semi-structured interview for the assessment Status Questionnaire in a geropsychiatry clinic.
of diagnosis and mental state in the elderly: The International Journal of Geriatric Psychiatry, 12:
Geriatric Mental State 1. Development and reliability. 526–532.
Psychological Medicine, 6: 439–449. Hall, K.S., Hugh, C., Hendrie, H.C. et al. (1993). The devel-
Davis, D., Creavin, S.T., Noel-Storr, A. et al. (2013). opment of a dementia screening interview in two
Neuropsychological tests for the diagnosis of distinct languages. International Journal of Methods
Alzheimer’s disease dementia and other dementia: in Psychiatric Research, 3: 1–28.
A generic protocol for cross-sectional and delayed- Hall, K.S., Ogunniyi, A.O., Hendrie, H.C. et al. (1996). A
verification studies. The Cochrane Database of cross-cultural community based study of dementias:
Systematic Reviews, 3: CD010460. Methods and performance of the survey instrument
Debanne, S.M., Patterson, M.B., Dick, R. et al. (1997). Indianapolis, U.S.A. and Ibadan, Nigeria. International
Validation of a telephone cognitive assessment bat- Journal of Methods in Psychiatric Research, 6:
tery. Journal of the American Geriatrics Society, 45: 129–142.
1352–1359. Harrison, J.K., Fearon, P., Noel-Storr, A.H. et al. (2014).
Fage, B.A., Chan, C.C., Gill, S.S. et al. (2015). Mini-Cog Informant Questionnaire on Cognitive Decline in
for the diagnosis of Alzheimer’s disease dementia the Elderly (IQCODE) for the diagnosis of demen-
and other dementias within a community setting. tia within a general practice (primary care) setting.
The Cochrane Database Systematic Reviews, 2: The Cochrane Database Systematic Reviews, 7:
CD010860. CD010771.
50 Dementia

Harrison, J.K., Fearon, P., Noel-Storr, A.H. et al. (2015). Lehfeld, H. and Erzigkeit, H. (1997). The SKT ‒ A short
Informant Questionnaire on Cognitive Decline in cognitive performance test for assessing deficits of
the Elderly (IQCODE) for the diagnosis of demen- memory and attention. International Psychogeriatrics,
tia within a secondary care setting. The Cochrane 9 (Suppl. 1): 115–121.
Database of Systematic Reviews, 3: CD010772. Little, A., Hemsley, D., Bergmann, K. et al. (1987).
Harwood, D.M., Hope, T. and Jacoby, R. (1997). Cognitive Comparison of the sensitivity of three instruments for
impairment in medical inpatients. 1: Screening for the detection of cognitive decline in elderly living at
dementia ‒ Is history better than mental state? Age home. British Journal of Psychiatry, 150: 808–814.
and Ageing, 26: 31–35. LoGiudice, D., Smith, K., Thomas, J. et al. (2006).
Hodkinson, H.M. (1972). Evaluation of a mental test score Kimberley Indigenous Cognitive Assessment tool
for the assessment of mental impairment in the (KICA): Development of a cognitive assessment
elderly. Age and Ageing, 1: 233–238. tool for older indigenous Australians. International
Hooijer, C., Jonker, C. and Lindeboom, J. (1993). Cases of Psychogeriatrics, 18: 269–280.
mild dementia in the community: Improving efficacy Mackinnon, A. and Mulligan, R. (1998). Combining cogni-
of case finding by concurrent use of pairs of screen- tive testing and informant report to increase accu-
ing tests. International Journal of Geriatric Psychiatry, racy in screening for dementia. American Journal of
8: 561–564. Psychiatry, 155: 1529–1535.
Horn, L., Cohen, C.I. and Teresi, J. (1989). The EASI: A self- Mackinnon, A., Khalilian, A., Jorm, A.F. et al. (2003).
administered screening test for cognitive impairment Improving screening accuracy for dementia in a com-
in the elderly. Journal of the American Geriatrics munity sample by augmenting cognitive testing with
Society, 37: 848–855. informant report. Journal of Clinical Epidemiology,
Jorm, A.F. (1997). Methods of screening for dementia: 56: 358–366.
A meta-analysis of studies comparing an informant Mant, A., Eyland, E.A., Pond, D.C. et al. (1988).
questionnaire with a brief cognitive test. Alzheimer’s Recognition of dementia in general practice:
Disease and Associated Disorders, 11: 158–162. Comparison of general practitioners’ opinions with
Jorm, A.F. (2004). The Informant Questionnaire on cog- assessments using the Mini-Mental State Examination
nitive decline in the elderly (IQCODE): A review. and the Blessed Dementia Rating Scale. Family
International Psychogeriatrics, 16: 275–293. Practice, 5: 184–188.
Jorm, A.F., Christiansen, H., Henderson, A.S. et al. (1996). Marshall, R.J. (1989). The predictive value of simple rules
Informant ratings of cognitive decline of elderly for combining two diagnostic tests. Biometrics, 45:
people: Relationship to longitudinal change on cogni- 1213–1222.
tive tests. Age and Ageing, 25: 125–129. McDowell, I., Kristjansson, B., Hill, G.B. and Hébert, R.
Jorm, A.F. and Korten, A.E. (1988). Assessment of cognitive (1997). Community screening for dementia: The
decline in the elderly by informant interview. British Mini Mental State Exam (MMSE) and Modified
Journal of Psychiatry, 152: 209–213. Mini-Mental State Exam (3MS) compared. Journal of
Kahn, R., Goldfarb, A., Pollack, M. and Peck, A. (1960). Clinical Epidemiology, 50: 377–383.
Brief objective measures of mental status in the aged. Molloy, D.W., Alemayehu, E. and Roberts, R. (1991).
American Journal of Psychiatry, 117: 326–328. Reliability of a standardised Mini-Mental State
Kawas, C., Karagiozis, H., Resau, L. et al. (1995). Reliability Examination compared with the traditional Mini-
of the Blessed telephone Information-Memory- Mental State Examination. American Journal of
Concentration test. Journal of Geriatric Psychiatry Psychiatry, 148: 102–105.
and Neurology, 8: 238–242. Morris, J.C., Heymann, A., Mohs, R.C. et al. (1989). The
Knafelc, R., LoGiudice, D., Harrigan, S. et al. (2003). The consortium to establish a registry for Alzheimer’s dis-
combination of cognitive testing and an informant ease (CERAD). Part 1. Clinical and neuropsychological
questionnaire in screening for dementia. Age and assessment of Alzheimer’s disease. Neurology, 39:
Ageing, 32: 541–547. 1159–1165.
Kokmen, E., Smith, G.E., Petersen, R.C. et al. (1991). Moyer, V.A. (2014). Screening for cognitive impairment in
The short test of mental status. Correlations with older adults: U.S. Preventive Services Task Force rec-
standardized psychometric testing. Archives of ommendation statement. Annals of Internal Medicine,
Neurology, 48: 725–728. 160: 791–797.
Larner, A.J. (2012). Screening utility of the Montreal Nasreddine, Z.S., Phillips, N.A., Bedirian, V. et al. (2005).
Cognitive Assessment (MoCA): In place of ‒ or as well The Montreal Cognitive Assessment, MoCA: A brief
as ‒ the MMSE? International Psychogeriatrics, 24: screening tool for mild cognitive impairment. Journal
391–396. of the American Geriatrics Society, 53: 695–699.
Screening and assessment instruments for the detection and measurement of cognitive impairment 51

Noel-Storr, A.H., McCleery, J.M., Richard, E. et al. (2014). Stern, R.G., Mohs, R.C., Davidson, M. et al. (1994).
Reporting standards for studies of diagnostic test A longitudinal study of Alzheimer’s disease:
accuracy in dementia: The STARDdem Initiative. Measurement, rate and predictors of cognitive
Neurology, 83: 364–373. deterioration. American Journal of Psychiatry, 151:
Petersen, R.C., Doody, R., Kurz, A. et al. (2001). Current 390–396.
concepts in mild cognitive impairment. Archives of Storey, J., Rowland, J., Basic, D. et al. (2004). The Rowland
Neurology, 58: 1985–1992. Universal Dementia Assessment Scale (RUDAS): A
Pfeiffer, E. (1975). A short portable mental status question- multicultural cognitive assessment scale. International
naire for the assessment of organic brain deficiency Psychogeriatrics, 16: 13–31.
in elderly patients. Journal of the American Geriatrics Stuss, D.T., Meiran, N., Guzman, D.A. et al. (1996).
Society, 23: 433–441. Do long tests yield a more accurate diagnosis of
Plutchik, R., Conte, H. and Lieberman, M. (1971). dementia than short tests? A comparison of 5 neu-
Development of a scale (GIES) for assessment of ropsychological tests. Archives of Neurology, 53:
cognitive and perceptual functioning in geriatric 1033–1039.
patients. Journal of the American Geriatrics Society, Swain, D.G. and Nightingale, P.G. (1997). Evaluation of a
19: 614–623. shortened version of the abbreviated mental test in a
Quinn, T.J., Fearon, P., Noel-Storr, A.H. et al. (2014). series of elderly patients. Clinical Rehabilitation, 11:
Informant Questionnaire on Cognitive Decline in 243–248.
the Elderly (IQCODE) for the diagnosis of dementia Teng, E.L. and Chui, H.C. (1987). The Modified Mini-
within community dwelling populations. The Cochrane Mental State (3MS) examination. Journal of Clinical
Database Systematic Reviews, 4: CD010079. Psychiatry, 48: 314–318.
Ritchie, K. and Fuhrer, R. (1992). A comparative study of Thomas, L.D., Gonzales, M.F., Chamberlain A. et al. (1994).
the performance of screening tests for senile demen- Comparison of clinical state, retrospective informant
tia using receiver operating characteristics analysis. interview and the neuropathologic diagnosis of
Journal of Clinical Epidemiology, 45: 627–637. Alzheimer’s disease. International Journal of Geriatric
Roccaforte, W.H., Burke, W.J., Bayer, B.L. and Wengel, S.P. Psychiatry, 9: 233–236.
(1992). Validation of a telephone version of the Mini- Valcour, V.G., Masaki, K.H., Curb, J.D. and Blanchette, P.L.
Mental State Examination. Journal of the American (2000). The detection of dementia in the primary
Geriatrics Society, 40: 697–702. care setting. Archives of Internal Medicine, 160:
Rosen, W.G., Mohs, R.C. and Davis, K.L. (1984). A new rat- 2964–2968.
ing scale for Alzheimer’s disease. American Journal of Ward, H.W., Ramsdell, J.W., Jackson, J.E. et al. (1990).
Psychiatry, 141: 1356–1364. Cognitive function testing in comprehensive geriatric
Roth, M., Tym, E., Mountjoy, C.Q. et al. (1986). CAMDEX: assessment: A comparison of cognitive test perfor-
A standardised instrument for the diagnosis of mance in residential and clinic settings. Journal of the
mental disorder in the elderly with special reference American Geriatrics Society, 38: 1088–1092.
to the early detection of dementia. British Journal of Williamson, J., Stokoe, I.H., Gray, S. et al. (1964). Old
Psychiatry, 149: 698–709. people at home: Their unreported needs. Lancet, 1
Royall, D.R., Mahurin, R.K. and Gray, K.F. (1992). Bedside (7343): 1117–1120.
assessment of executive cognitive impairment: The Zaudig, M., Mitelhammer, J., Pauls, A. et al. (1991).
executive interview. Journal of the American Geriatric SIDAM ‒ A structured interview for the diagno-
Society, 40: 1221–1226. ses of dementia of the Alzheimer’s type, multi
Solomon, P.R., Hirschoff, A., Kelly, B. et al. (1998). A 7 min- infarct dementia and dementias of other aetiology
ute neurocognitive screening battery highly sensitive according to ICD-10 and DSM III R. Psychological
to Alzheimer’s disease. Archives of Neurology, 55: Medicine, 21: 225–236.
349–355.
6
Neuropsychological assessment of dementia

GREG SAVAGE

which is due to Alzheimer’s disease (AD), but extends to


6.1 INTRODUCTION other diseases and conditions associated with cognitive
decline. Many of these present late in the lifespan and are
Dementia is a fundamentally neuropsychological concept, contenders in differential diagnosis of the underlying dis-
describing the unravelling of the mind due to a brain dis- ease. Accordingly, there is discussion of typical test find-
order: neuro plus psychological. The disabling cognitive and ings in cases of dementia with Lewy bodies (DLB), vascular
behavioural impacts of dementia are typically preceded by dementia (VaD), three kinds of frontotemporal dementia
gradually emerging signs of cognitive change, providing the (FTD) (the behavioural variant of FTD [bvFTD] and the
first inkling that something is not quite right. A brief cogni- focal-onset variants known as semantic dementia [SD] and
tive screening test by a geriatrician or neurologist may be progressive non-fluent aphasia [PNFA]) and also cognitive
enlightening, but if the signs are subtle a referral for com- impairment associated with cases of major depressive disor-
prehensive neuropsychological testing should be made. The der (MDD), sometimes called depressive pseudodementia.
emergence of clinical neuropsychology as a mainstream Other conditions where dementia is not a primary feature
specialty discipline in hospital outpatient clinics (e.g. so- will not feature here. Thus, while dementia develops in neu-
called ‘memory clinics’) has allowed for the quantitative rodegenerative movement disorders such as Parkinson’s
evaluation of cognitive decline with the kind of metric disease and Huntington’s disease, the early diagnosis and
resolution which healthcare professionals might reasonably overriding clinical concerns relate to morbidity due to the
expect of an empirical investigation. primary movement disturbance. Dementia due to infec-
This chapter outlines the role of neuropsychological tious diseases, toxic exposure, hydrocephalus or chronic
assessment in helping to make a diagnosis of the underlying severe alcoholism is less common and prominent signs in
cause of dementia. It characterizes the neuropsychological the clinical history may serve to argue against a more com-
domains affected in various kinds of dementia, the tests of mon dementia diagnosis, and to orient a neuropsychologi-
those domains used to inform neuropsychological opinion cal referral to specialist services.
and perspectives commonly taken by neuropsychologists in Dementia is usually defined in terms of deteriora-
forming an opinion. Other chapters in this volume provide tion in two areas: cognition and instrumental activities
expert and comprehensive reviews of the clinical and cog- of daily living (IADL). Historically, classification systems
nitive features of conditions associated with dementia ‒ so have required cognitive decline involving memory and at
this chapter will focus in a ‘hands-on’ manner on the process least one other cognitive domain but recent clinical and
followed by the neuropsychologist in responding to referral research consensus guidelines allow different combina-
questions. The chapter is intended to serve two audiences: tions of two domains (McKhann et al., 2011). When there
neuropsychologists fielding dementia-related referrals, and is measurable cognitive decline but the person is still cop-
other health professionals who make such referrals (in par- ing adequately with daily activities, the term mild cogni-
ticular, those who do not use neuropsychological services tive impairment (MCI) is typically used. Many people are
routinely, but might do so given a better understanding of referred to a neuropsychologist with this quasi-diagnosis
the assessment process). and turn out to have had prodromal AD when followed up,
The chapter adopts a broad definition of dementia, con- reflecting an annual overall rate of transition from MCI
sistent with the overall perspective of this volume. Coverage to ‘AD dementia’ estimated at around 10% (Bruscoli and
focuses primarily on the most common form of dementia, Lovestone, 2004).

52
Neuropsychological assessment of dementia 53

edition (WMS-IV; Wechsler, 2009a). Their intelligence quo-


6.2 PERSPECTIVES ON tient (IQ) or Index scores have a normative mean of 100 and
NEUROPSYCHOLOGICAL a standard deviation of 15, and partitioning of the normal
ASSESSMENT distribution of IQs/Indexes into ranges provides descriptors
such as average (with IQs between 90 and 110), low aver-
Neuropsychological assessment of dementia cannot be age (80–90), high average (110–120) and so on. These scores
purely psychometric. It is placed squarely in the context translate directly to Scaled Scores when subtests of the
provided by a comprehensive interview with the patient and Wechsler tests are discussed (mean = 10, standard devia-
an informant. This interview must leave the clinician with tion = 3). Some tests provide percentile ranks, and others
a clear understanding of the time course of the emergence use T-scores (mean = 50, standard deviation = 10). Z scores
of signs/symptoms, and the patterning of what seem to be (i.e. quantifying departure from average performance in
preserved versus declining abilities. A perspective on edu- standard deviation units for a particular test) are frequently
cational and vocational achievements will inform expecta- used as a common currency in comparing performances
tions about premorbid cognitive status, and the interview across a range of tests using different summary parameters.
can provide important information about lifetime exposure
to brain injuries or substances, which could account for 6.2.1 COMPONENTS OF A BATTERY FOR
cognitive decline. DEMENTIA ASSESSMENT
The assessment process outlined in this chapter is based
on a cognitive neuropsychological approach to clinical test- A typical test battery used when dementia is suspected takes
ing (Mapou and Spector, 1995), which structures the test- between one and two hours to administer and covers orien-
ing session and interprets test data according to processing tation, ability to attend and manipulate information in mind,
models developed in cognitive psychology. The process is speed of information processing (usually tested via psycho-
flexible and hypothesis driven in contrast to fixed-battery motor tasks), constructional praxis, executive functioning,
approaches, and conclusions about higher cognitive func- language and new learning and retention (usually for both
tions are parsimonious, taking into account the influences verbal and nonverbal material). A context for interpreting
of ‘foundation-level’ abilities such as attention and speed performances in these domains is established by estimating
of processing. Outcomes of the assessment are framed in premorbid intellectual level. Neuropsychologists also try to
cognitive terms (e.g. memory problems being amenable to characterize recent mood state, usually by the administra-
cueing strategies or not) as opposed to performance-level tion of a formal self-report inventory (although such mea-
terms (e.g. verbal memory being placed in the low average sures are invalid when memory impairment is prominent).
range), as these tend to be understandable by those mak- The following section describes commonly used tests
ing referrals, as well as those who care for the patient. For and their utility in differentiating between varieties of
elaboration on this cognitive neuropsychological formulation potential diagnoses: AD, DLB, VaD, bvFTD, SD, PNFA and
perspective see Savage (2016). depression. Table 6.1 outlines likely patterns of preserved
Neuropsychologists vary in terms of their utiliza- and impaired cognition in these disorders over the domains
tion of psychometric properties of tests. Those taking a covered in this section.
hypothesis-testing approach may administer subtests of
­
larger batteries, admixing with other ‘single-focus’ tests, and 6.2.1.1 Tests of orientation and personal
might refer to a variety of normative databases without any and general information
formal statistical comparison. Aggregation of subtest scores
into summary indexes can be seen as a confound, poten- Neuropsychologists usually begin testing with non-con-
tially obscuring clinically meaningful differences across fronting questions, which are administered by most health
subtests. In common with the fixed-battery approach, the professionals in some form; these are not psychometrically
hypothesis-testing approach prescribes that tests should sophisticated, but answers here can provide significant
possess robust psychometric properties in terms of reliabil- insights into what to expect on later testing (e.g. mistak-
ity and gracefully graded sensitivity to variation in perfor- ing current age by years, or the year by a decade, or citing
mance, even if the full potential for statistical analysis is not past political leaders as current). Such strikingly aberrant
used. Both approaches judge cognitive performance relative responses are common in early AD; occasional incorrect
to age-appropriate norms, and sometimes sex and educa- responses may represent the ‘patchy dysmnesia’ often seen in
tional level are taken into account. depressive illness. The Information and Orientation subtest
Measured levels of performance, as well as differences of the WMS-III has 14 items and tests orientation more thor-
between performances for different domains, or over time, oughly than its WMS-IV counterpart (the Brief Cognitive
are characterized using a range of parameters. The flag- Status Examination, with only six items); the first 10 items
ship tests of the ubiquitous Wechsler suite are the Wechsler of the Mini-Mental State Examination (MMSE; Folstein
Adult Intelligence Scale ‒ Fourth edition (WAIS-IV; et al., 1975) or the Addenbrooke’s Cognitive Examination-
Wechsler, 2008) and Wechsler Memory Scale ‒ Fourth III (ACE-III; Hsieh et al., 2013) may be used instead.
54 Dementia

Table 6.1 Typical patterns of cognition for conditions associated with dementia
Expected performance in various conditions
Cognitive Typical
domain tests AD DLB VaD bvFTD SD PNFA MDD
Orientation I/Oa Impaired Intact Impaired? Intact Intact Intact Variable
MMSEb
ACE-IIIc
Attention/ DSd Intact early on Impaired Impaired? Impaired Intact May be Impaired
working impaired
memory
Processing CDe Intact Intact? Impaired? Intact Intact Intact Impaired
speed
Reasoning Simf Intact early on Intact? Impaired? Impaired Impaired due Intact Intact
PCg to semantic
loss?
Language BNTh Impaired Intact Impaired? Intact Impaired Largely intact Intact
GNTi naming;
CFj effortful
speech
affects fluency
Executive LFk Mildly Impaired Impaired Impaired Intact early on Intact early on; Impaired
functioning CFj impaired? effortful
H&Bl speech may
CWITm affect speed
on language-
based tasks
Construction RCFTn Mildly Impaired Impaired? Mildly impaired Intact Intact Mildly
BDo impaired due to poor impaired
planning? due to poor
planning?
Memory LMp Impaired Impaired Impaired recall Impaired recall, May be Intact Impaired
VRq recall and recall, intact and intact impaired recall, intact
RAVLT  r recognition recognition? recognition? recognition? due to recognition?
CVLT-IIs semantic
HVLT-Rt loss?
BVMT-Ru
LLTv
RCFTn
Abbreviations: AD, Alzheimer’s disease; DLB, dementia with Lewy bodies; VaD, vascular dementia; bvFTD, behavioural variant frontotemporal
­dementia; SD, semantic dementia; PNFA, progressive non-fluent aphasia; MDD, major depressive disorder.
a Wechsler Memory Scale – Third edition (WMS-III) Information and orientation

b Mini-Mental State Examination

c Addenbrooke’s Cognitive Examination-III

d Wechsler Adult Intelligence Scale – Fourth edition (WAIS-IV) Digit Span

e WAIS-IV Coding

f WAIS-IV Similarities

g WAIS-IV Picture Completion

h Boston Naming Test

i Graded Naming Test (McKenna and Warrington, 1983)

j Category fluency

k Letter fluency

l Hayling and Brixton Tests

m Delis–Kaplan Executive Function System (D-KEFS) Color–Word Interference Test

n Rey Complex Figure Test

o WAIS-IV Block Design

p WMS-IV Logical Memory

q WMS-IV Visual Reproduction

r Rey Auditory Verbal Learning Test

s California Verbal Learning Test – Second edition

t Hopkins Verbal Learning Test – Revised

u Brief Visuospatial Memory Test – Revised

v Location Learning Test


Neuropsychological assessment of dementia 55

6.2.1.2 Estimation of premorbid intellectual 6.2.1.4 Tests of processing speed


level
Speed of mentation is hard to measure without introduc-
Inferences about cognitive decline are necessarily made ing confounds related to the task employed. Some think-
with respect to expected levels of performance in the ing speed tasks use motoric responses to visual stimuli and
absence of brain illness. Typically there are no ‘intact’ base- care must be taken to ensure a putative speed measure is
line data available for comparison, and so broadly placed not overly reflecting the influence of other variables (e.g.
estimates are made (e.g. average, high average, etc.). One difficulty seeing or fixating visual stimuli, or motoric dif-
method takes account of educational background, voca- ficulty/slowness). A widely used test of processing speed is
tional achievements and/or demographic variables (either the Coding subtest of the WAIS-IV, which measures speed
informally or via input to normatively derived regression of transcription of symbols into a long series of numbered
equations). Another approach is to measure current abil- boxes, guided by a number–symbol matching key. The
ity in a domain which is usually resistant to even moderate total number of symbols correctly transcribed within two
dementia, such as word knowledge: correct reading aloud of minutes indicates speed of the translation from number to
so-called irregular words whose pronunciations do not fol- symbol. However, it can index other factors, such as visual
low predictable spelling–sound rules suggests intact word scanning of the arrays, motoric slowness, working memory,
knowledge and tests incorporating graded difficulty afford a associative learning of matches and sustained attention.
psychometric estimate of likely premorbid intellectual level. Reduced speed of processing (and the confounded influ-
Typically used tests of word knowledge include the National ence of working memory and sustained attention) is not
Adult Reading Test (NART; Nelson, 1982) and a recent vari- usually compromised in mild or preclinical AD or in SD,
ant, the Test of Premorbid Functioning (TOPF; Wechsler, but can be seen in bvFTD, DLB, VaD and depression.
2009b), co-normed with the WAIS-IV and WMS-IV. Care Against a background established in terms of both likely
should be taken if SD is a differential under consideration: premorbid capabilities and current foundation-level skills,
while word knowledge is typically resilient against the testing of higher mental functions can then proceed to be
impact of neurodegeneration, this is by definition not the interpreted.
case with SD.
A cognitively framed assessment then proceeds to assess 6.2.1.5 Tests of language functioning
foundation-level domains such as concentration and speed
of thinking, as weaknesses here can undermine (and mis- A typical neuropsychological battery used in dementia-
represent) higher-level functioning. related referrals tends not to assess language comprehensively,
but ability to name objects is routinely assessed. Naming
reflects semantic access (and/or representation) as well as
6.2.1.3 Tests of concentration and working phonological processing, and commonly used tests present
memory line drawings of objects graded by frequency of encounter.
The Boston Naming Test-2 (BNT-2; Kaplan et al., 2001) com-
Ability to engage in routine sequencing (concentration prises 60 items, with a default entry point at item 30, and
or focused attention) and to hold information in mind allows for phonological cueing if the patient cannot name the
while working on a problem (so-called working memory) item spontaneously (assessing a so-called ‘tip-of-the-tongue’
is assessed for two purposes. A general purpose serves to state). It is often administered in either a 15- or 30-item short
qualify interpretation of performances on more complex form. Care should be taken to recognize that certain items
or focal cognitive abilities, and an efficient strategy on the are culture-bound (e.g. beaver is intended to be an easy item,
part of the neuropsychologist is to establish how well the but probably this is true only for North American residents).
patient can attend quite early in the course of the assess- Dysnomia is a prominent sign in mild AD and focal
ment. Poor moment-to-moment concentration might ‘reca- dementias such as SD and PNFA; degraded semantic
librate’ expectations in the examiner and suggest use of knowledge is thought to be the problem in AD and SD, and
simplified versions of tests, where floor-level performances a phonological account is proposed in PNFA. Integrity of
are less likely. semantic knowledge can also be assessed by testing vocabu-
A specific purpose for assessing concentration and work- lary and general knowledge (e.g. using the Vocabulary and
ing memory is to provide differential diagnostic informa- Information subtests of the WAIS-IV). Syntactic aspects
tion. Poor performance here can reflect dysfunction in of language are usually preserved in all but severe cases of
frontal systems and their connections to other brain regions: dementia, and formal testing is rarely undertaken.
Patients with depressive and/or anxiety disorders, bvFTD
or DLB often perform poorly on tests such as the Digit Span 6.2.1.6 Tests of construction
and/or Letter–Number Sequencing subtests of the WAIS-IV
and Mental Control from the WMS-III (Wechsler, 1997). Visuoconstructional ability is always tested in some form.
Cases with mild AD, however, do not tend to have promi- The Rey Complex Figure Test (RCFT; Meyers and Meyers,
nent difficulty in this cognitive domain. 1995) affords good insights into constructional praxis, and
56 Dementia

evaluating the organizational approach taken in copying responses in dementias associated with hypofrontality, such
the figure informs an opinion about executive function- as bvFTD, DLB and often VaD.
ing. Dementia affecting posterior functions (e.g. in mild- Perhaps the most widely used test of executive func-
to-moderate AD and in DLB) can produce distortion in tioning, included in most batteries, involves word genera-
reproduction of spatial relationships among elements of the tion under time pressure and with compliance to a set of
figure, often culminating in a bizarrely misshapen copy. In stated rules. Letter fluency (sometimes incorrectly called
contrast, distortion is seldom seen in dementias with ante- phonemic fluency) requires production of as many words as
rior degeneration (e.g. bvFTD, SD), although misalignment possible which begin with a prescribed letter in a 1-minute
may occur due to a poorly planned approach. Matching of period. The D-KEFS version requires observance of three
blocks to specified patterns is another frequently adminis- rules: no proper nouns, no numbers and no variations on
tered test (e.g. the Block Design subtest of the WAIS-IV). the same word. Trials are undertaken for three letters (usu-
This test can be failed at easy levels in mild AD, and while ally F, A and S), and the total score indexes flexible thinking
patients with bvFTD (and perhaps DLB) may have trouble, in a non-routine generation task requiring self-monitoring,
they can benefit from provision of structure: once the test is rule observance, mindful exploitation of lexical similari-
finished, using a Perspex overlay to segment the component ties (e.g. flip suggesting flap and flop in quick succession)
blocks may assist initiation of a response. and adaptability in the face of ‘thought blocking’. A vari-
ant, which is more structured (and hence normally easier),
6.2.1.7 Tests of executive functioning simply requires generation of words on a given theme. This
category (or semantic) fluency test should also be given with
In an assessment tailored to be time efficient and mini- multiple trials for robust measurement: in the D-KEFS ver-
mally frustrating, many neuropsychologists avoid lengthy sion, two trials are given with different topics; an additional
concept-formation tests such as the Wisconsin Card Sorting trial requires switching between two topics (which adds
Test (WCST), and prefer brief tests of the same construct extra executive load).
such as the Brixton test (Burgess and Shallice, 1997). Here, Fluency tests are very informative in dementia assess-
the patient is required to anticipate the next location in a ments, as the normal pattern of better performance on cat-
spatial sequence; the patient is warned at the outset that egory fluency than letter fluency tends to be reversed in even
the sequential rule will change from time to time, and they mild or prodromal AD: letter fluency can be normal, but
will need to pick up on the new sequence. Ability to inhibit category fluency fails badly due to deteriorating semantic
prepotent responses is often tested using a variant of the processing. This pattern may be seen in SD, but letter flu-
Stroop test (e.g. the Color–Word Interference Test from the ency can also be impaired. In depression, bvFTD, DLB and
Delis–Kaplan Executive Function System; D-KEFS; Delis VaD letter fluency is typically impaired, with category flu-
et al., 2001). Here, a series of colour names is printed using ency following suit or remaining largely intact.
incongruently coloured ink (e.g. the word red is printed in
blue ink); speeded naming of ink colour is normally difficult 6.2.1.8 Tests of memory
due to a natural tendency to read the word itself, and failure
to inhibit this tendency (evident as slowness or error-prone Formal testing of learning and retention of new informa-
performance) indicates poor executive control. In a simi- tion assesses integrity of episodic memory functioning and
lar vein, the Hayling test (packaged with the Brixton) mea- is intended to characterize the forgetfulness in everyday life,
sures response latency and error rate in completing sentence which is such a frequent complaint – either by the patient
fragments with intentionally nonsensical final words (e.g. or more often by significant others. Learning is sometimes
London is a very busy: banana); executive failures lead to tested with only one exposure to material, but preferably
either frankly predictable completions (e.g. city), or margin- with repeated trials of exposure and testing, otherwise brief
ally less predictable ones (e.g. location) or abnormally long attentional lapses can confound interpretation. Testing of
latencies to produce an acceptable response. Tests of strat- learning is performed immediately, but the most reveal-
egy formation and response inhibition are useful in charac- ing data are usually provided after a delay of about 20–30
terizing frontal systems dysfunction in cases with bvFTD, ­minutes. Testing might require free recall of information,
and also in DLB and VaD. Cases with AD are less likely to cued recall or recognition of the information when pre-
fail these tests because of flawed strategies or poor inhibi- sented among potentially confusable information. This gra-
tion; rather, they may fail because they cannot understand dient between recall versus recognition failure maps onto
the nature of the task itself. cognitive constructs of retrieval versus encoding/consolida-
Ability to engage in abstract reasoning is an aspect of tion problems, and this distinction is thought to have diag-
executive functioning, which tends to decline gradually nostic value in discriminating among dementia types. In
in AD, and is impaired earlier and more prominently in brief, retrieval problems are characterized by poor recall but
conditions which compromise frontal systems function- better recognition performances, due to failure of frontally
ing. The Similarities and Picture Completion subtests from mediated initiation of access to a stored memory trace; this
the WAIS-IV test reasoning based on verbal and pictorial pattern is typically observed in depression, DLB, VaD and
materials, respectively, and can elicit concrete and literal bvFTD. In contrast, an encoding/consolidation problem
Neuropsychological assessment of dementia 57

is thought to represent temporal lobe dysfunction, as seen of Visual Reproduction by testing at the end of a series of 24
in AD and perhaps in SD to the extent that the informa- items; delayed recognition tests retention. Finally, the Location
tion source itself may be degraded. In these conditions, an Learning Test (LLT; Bucks et al., 2000) measures correct place-
adequate memory trace is never formed and so patients per- ment of drawings of 10 common objects in a 5 × 5 grid over
form poorly on both recall and recognition testing. multiple trials. It has good metric properties, which capture
Assessment of unilateral memory disorders (e.g. in cases of the degree of error in misremembering location.
stroke, tumour or epilepsy) has resulted in a tradition of test- In all these tests, both verbal and nonverbal, patients
ing memory using both verbal materials (usually word list, with AD tend to have difficulty with both recall and rec-
word pairings and/or prose passages) and nonverbal materi- ognition modes of testing, and patients with bvFTD, DLB,
als (usually drawn diagrams, faces and/or object locations). VaD and depression tend to have less difficulty on recogni-
Free recall of prose passages (e.g. WMS-IV Logical Memory) tion testing.
is often tested, although performance here is not useful in
distinguishing between a temporal lobe encoding/consolida-
tion problem and a more frontally mediated retrieval-based
impairment. In addition, large amounts of information are 6.3 COMMUNICATING A
presented without multiple repeated trials, and attentional NEUROPSYCHOLOGICAL OPINION
problems can confound interpretation. Performance on free
recall word lists over repeated trials, combined with delayed
The outcome of the assessment is typically conveyed in a
recall and subsequent recognition testing addresses both these
formal report to the referrer. Neuropsychological reports
issues, and the Rey Auditory Verbal Learning Test (RAVLT)
are usually comprehensive documents, which combine
(Rey, 1958; Strauss et al., 2006) is widely used in dementia-
appraisal of the clinical history, qualitative aspects of
related assessments. A similar test, the semantically clustered
behaviour on interview and throughout testing, and inter-
California Verbal Learning Test ‒ Second edition (CVLT-II;
pretation of test scores, Here, the operative word is inter-
Delis et al., 2000) is popular in North America but inclusion
pretation: the score itself may be qualified by the manner in
of a few culturally bound items may confound its use in other
which it was achieved (e.g. normatively average repetition
English-speaking countries (e.g. where subway refers to a
of digits might represent graceful arrival at a threshold of
road underpass and thereby does not fall into the semantic
difficulty, or a chaotic pattern of early failures on easy items,
cluster modes of transport). These tests are particularly use-
which persists through to extremely difficult items; the total
ful in the early detection of dementia, but the list length (15
score does not distinguish between these clinically quite
and 16 items respectively) may prove overwhelming in mild-
different performances). Overall, the assessment process
to-moderate cases, resulting in floor effects, which preclude
and the resulting opinion is fundamentally neuropsycho-
measurement of deterioration over time. Easier semanti-
logical, not neuropsychometric. This should be reflected in
cally clustered versions such as the 12-item Hopkins Verbal
the conclusions reached: an opinion should be conveyed in
Learning Test ‒ Revised (HVLT-R; Benedict et al., 1998)
plain language with minimal recourse to statistics, and the
or 9-item CVLT-II Short Form are extremely useful when
logic underlying the interpretation of the findings should
screening of memory reveals strikingly impaired ability.
be transparent. If the referrer has primary responsibility
In the nonverbal domain, memory for designs is frequently
for clinical care of the patient, they will usually discuss the
tested but relies on intact constructional ability. The Visual
findings and opinion in a review consultation, possibly in
Reproduction (VR) subtest of the WMS-IV measures recall for
a synthesis with conclusions from other investigations. It
a series of designs with increasing complexity after a 10-second
is very useful to supplement this meeting with face-to-face
exposure; such immediate recall reflects working memory,
feedback from the neuropsychologist to the patient, in the
but subsequent recall after 30 minutes does reflect retention.
company of a carer; this provides the opportunity for clari-
Incidental testing of recall for the Rey Complex Figure (i.e.
fication and for discussing potential management strategies.
without informing the patient ahead of time) indexes reten-
tion, but can only be interpreted validly when the initial
copy was reasonably accurate and completed in an organized REFERENCES
manner. The same caveats applied above to prose passages
regarding attentional lapses and retrieval versus encoding/ Benedict, R.H.B. (1997). Brief Visuospatial Memory
consolidation interpretations apply to these visually based Test ‒ Revised. Odessa, FL: Psychological Assessment
free recall tests with only single exposure learning phases. Resources.
The Brief Visuospatial Memory Test – Revised (BVMT-R) Benedict, R.H.B., Schretlen, D., Groninger, L. and Brandt, J.
(Benedict, 1997) is a nonverbal analogue to the HVLT-R, and (1998). Hopkins Verbal Learning Test ‒ Revised:
uses repeated trials of free recall of six simple designs viewed Normative data and analysis of inter-form and test-
at once for 10 seconds; subsequent recognition testing after retest reliability. Clinical Neuropsychologist, 12: 43–55.
delayed recall quantifies retrieval difficulty. The Faces sub- Bruscoli, M. and Lovestone, S. (2004). Is MCI really just
test of the WMS-III tests immediate recognition memory for early dementia? A systematic review of conversion
photographs of faces, avoiding the working memory confound studies. International Psychogeriatrics, 16: 129–140.
58 Dementia

Bucks, R.S., Willison, J.R. and Byrne, L.M.T. (2000). McKhann, G.M., Knopman, D.S., Chertkow, H. et al. (2011).
Location Learning Test: Manual. Bury St. Edmunds, UK: The diagnosis of dementia due to Alzheimer’s disease:
Thames Valley Test Company. Recommendations from the National Institute on
Burgess, P.W. and Shallice, T. (1997). The Hayling and Aging-Alzheimer‘s Association workgroups on diag-
Brixton Tests. Thurston, UK: Thames Valley Test nostic guidelines for Alzheimer’s disease. Alzheimer’s
Company. & Dementia, 7: 263–269.
Delis, D.C., Kaplan, E. and Kramer, J.H. (2001). The Delis- Meyers, J.E. and Meyers, K.R. (1995). Rey Complex Figure
Kaplan Executive Function System (D-KEFS). San Test and Recognition Trial. Odessa, FL: Psychological
Antonio, TX: The Psychological Corporation. Assessment Resources.
Delis, D.C., Kramer, J. H., Kaplan, E. and Ober, B.A. Nelson, H.E. (1982). The National Adult Reading Test
(2000). California Verbal Learning Test, Second Edition, (NART). Windsor, England: NFER-Nelson.
Adult Version. San Antonio, TX: The Psychological Rey, A. (1958). L’examen Clinique en Psychologie. Paris,
Corporation. France: Presse Universitaire de France.
Folstein, M.F., Folstein, S.E. and McHugh, P.R. (1975). Savage, G. (2016). Cognitive neuropsychological formu-
“Mini-mental state”: A practical method for grading lation. In J.A.B. Macniven (ed.), Neuropsychological
the cognitive state of patients for the clinician. Journal Formulation: A Clinical Casebook. New York, NY:
of Psychiatric Research, 12: 189–198. Springer pp. 221–239.
Hsieh, S., Schubert, S., Hoon, C., Mioshi, E. and Hodges, Strauss, E., Sherman, E.M.S. and Spreen, O. (2006).
J.R. (2013). Validation of the Addenbrooke’s Cognitive A Compendium of Neuropsychological Tests:
Examination III in frontotemporal dementia and Administration, Norms, and Commentary, Third
Alzheimer’s disease. Dementia and Geriatric Cognitive Edition. New York, NY: Oxford University Press.
Disorders, 36: 242–250. Wechsler, D. (1997). Wechsler Memory Scale, Third
Kaplan, E.F., Goodglass, H. and Weintraub, S. (2001). The Edition. San Antonio, TX: The Psychological
Boston Naming Test, Second Edition. Philadelphia, PA: Corporation.
Lippincott Williams & Wilkins. Wechsler, D. (2008). Wechsler Adult Intelligence Scale,
Mapou, R.L. and Spector, J. (1995). Clinical Fourth Edition. San Antonio, TX: Pearson.
Neuropsychological Assessment: A Cognitive Wechsler, D. (2009a). Wechsler Memory Scale, Fourth
Approach. New York, NY: Plenum Press. Edition. San Antonio, TX: Pearson.
McKenna, P. and Warrington, E. (1983). Graded Naming Wechsler, D. (2009b). Test of Premorbid Functioning. San
Test. Windsor, England: NFER-Nelson. Antonio, TX: The Psychological Corporation.
7
Neuropsychiatric aspects of dementia

NILIKA PERERA, MEHRAN JAVEED, CONSTANTINE G. LYKETSOS AND IRACEMA LEROI

7.1 INTRODUCTION 7.2 IMPACT OF NEUROPSYCHIATRIC


SYMPTOMS
‘Neuropsychiatric symptoms’ is an umbrella term that
describes the non-cognitive symptoms and behaviours that The negative impact of neuropsychiatric complications on
often occur in people with dementia. These symptoms include people with dementia, caregivers and the community is
changes in mood and personality, perceptual abnormalities, significant, and far greater than the impact of dementia-
psychomotor disturbances (such as apathy, agitation, aggres- related functional limitations or cognitive decline (Banerjee
sion, wandering and purposeless behaviour) and neurovegeta- et al., 2006). According to the Alzheimer Europe Dementia
tive changes (such as sleep, appetite and energy disturbances). Carers’ Survey, 50% of caregivers report such symptoms
Neuropsychiatric symptoms have been variously termed as the most problematic aspect of caring for someone with
‘behavioural and psychological symptoms of dementia’ dementia.
(BPSDs), ‘non-cognitive changes’ and ‘challenging behav- Apathy, irritability and agitation are noted to be the most
iours’. The importance and ubiquity of these symptoms are prevalent neuropsychiatric symptoms in dementia in gen-
highlighted by Alois Alzheimer’s first clinical description of eral (Bergh and Selbæk, 2012), whilst agitation, aggression
the condition which later became known as Alzheimer dis- and personality change are rated as the most troublesome
ease (AD). In this first case study, Alzheimer wrote: dementia-related symptoms for at least 16% of caregivers
(Georges et al., 2008).
The first noticeable symptoms of illness shown Identifying neuropsychiatric symptoms in people with
by this 51-year old woman was suspiciousness of dementia can aid in anticipating further cognitive deteriora-
her husband. Soon, a rapidly increasing memory tion and guiding clinical decision making (Javeed and Leroi,
impairment became evident; she could no lon- 2014). Neuropsychiatric complications are a significant risk
ger orient herself in her own dwelling, dragged factor for institutionalization (O’Donnell et al., 1992), and
objects here and there and hid them, and at are associated with a worse prognosis and a more rapid rate
times, believing that people were out to murder of illness progression (Paulsen et al., 2000). Furthermore,
her, started to scream loudly (Alzheimer, trans- the cost of caring for people with dementia with concurrent
lated by Jarvik and Greenson, 1987). neuropsychiatric complications is significantly greater than
the cost of caring for someone with dementia without such
Neuropsychiatric symptoms are a common first presen- complications (Murman et al., 2002).
tation of most dementia syndromes and often prompt refer- Neuropsychiatric complications in dementia can mark-
ral for assessment. In some cases, changes in personality or edly increase the risk of injury to the person with demen-
behaviour in later life, namely a putative diagnosis of ‘mild tia and their caregiver(s). Caregivers also have an increased
behavioural disturbance’ may be a harbinger of later demen- risk of depression, anxiety and social isolation (Gonzalez-
tia. These changes are most likely secondary to frontotem- Salvador et al., 1999; Brodaty and Donkin, 2009), although
poral degeneration, in an analogous way that a proportion of this may depend on their own personal vulnerabilities, social
people with ‘mild cognitive impairment (MCI)’ frequently circumstances as well as the nature of the behavioural dis-
go on to develop later significant cognitive impairment war- turbances. For example, in dementia related to Parkinson’s
ranting a diagnosis of dementia (Taragano et al., 2009). disease (PD), those with emotional blunting related to apathy

59
60 Dementia

have significantly worse caregiver outcomes compared to of cognitive deterioration and should therefore be identified
those with PD and apathy without emotional blunting (Leroi and managed early (Leroi et al., 2012; Mauri et al., 2012).
et al., 2014). Moreover, in care homes or nursing facilities,
the risk of injury and insult to other residents and staff can
lead to a high turnover of staff leading to a less positive work
environment. This can precipitate further behavioural dis- 7.5 NEUROPSYCHIATRIC SYMPTOMS
turbances and a reduction in the quality of life in residents. IN MILD COGNITIVE IMPAIRMENT
Finally, if severe, the presence of neuropsychiatric symptoms
raises the risk of the person with dementia being managed Neuropsychiatric symptoms affect between 40% and 75% of
with restraints and antipsychotic medications which can people with MCI (Lyketsos et al., 2002; Hwang et al., 2004).
increase risk of mortality and morbidity and more rapidly In the Cardiovascular Health Study (CHS), 43% of the 320
deteriorating cognition (Bianchetti et al., 1997). individuals who were identified as having MCI exhibited
neuropsychiatric symptoms, with depression, apathy and
irritability being the most common (Lyketsos et al., 2002).

7.3 EPIDEMIOLOGY AND SPECTRUM


OF NEUROPSYCHIATRIC
SYMPTOMS OF DEMENTIA 7.6 NEUROPSYCHIATRIC SYMPTOMS IN
DEMENTIA IN COMMUNITY AND
Neuropsychiatric symptoms are extremely common in CLINICAL SETTINGS
the dementias, despite variations in settings and cultures.
However, unlike the predictable decline seen in the cogni- The Medical Research Council Cognitive Function and
tive and functional abilities of dementia sufferers, neuro- Ageing Study (MRC CFAS) in United Kingdom (Brayne
psychiatric symptoms may fluctuate in their presence and et al., 2006) and the Cache County Study of Memory in
intensity and could be eliminated through appropriate Aging (CCSMA) in Utah, United States (Breitner et al.,
management. This, in turn, could improve quality of life for 1999) are landmark population-based studies of neuropsy-
the person with dementia and their caregiver. chiatric symptoms. The MRC CFAS prospectively followed
13,004 individuals over the age of 64 for 10 years assessing
the participants’ cognition and neuropsychiatric symptoms
using the Mini-Mental State Exam (MMSE) and an adapted
7.4 NEUROPSYCHIATRIC SYMPTOMS Geriatric Mental State (GMS). Of the 587 individuals with
AS PROGNOSTIC MARKERS dementia, irritability was more common in those with less
severe cognitive dysfunction (MMSE score of ≥22), whereas
Neuropsychiatric symptoms can precede or follow the wandering, elation, apathy and psychosis were more com-
emergence of cognitive and functional impairment and mon in those with more severe cognitive dysfunction
may be predictive of more rapid decline in these domains. (MMSE <17) (Savva et al., 2009). Depression and anxiety
For example, the risk of developing AD, the most common were more common in younger people with dementia, as
type of dementia, increases with each episode of later-life well as in those with vascular risk factors. In the CCSMA,
depression over the age of 65, if the depression is associated 61% of all the participants with dementia had one or more
with cognitive symptoms (Wilson et al., 2002). Moreover, neuropsychiatric symptom in the month prior to interview.
the Multi-institutional Research in Alzheimer’s Genetic Specifically, among those with a diagnosis of AD, 23% (n =
Epidemiology (MIRAGE) study found that depression 49) had delusions and 13% (n = 28) had hallucinations. In
occurring many years before the onset of cognitive decline contrast, in those with a diagnosis of vascular dementia
may be a significant risk factor for the future development of (VaD), point prevalences were 8% (n = 5) and 13% (n = 8)
AD (Green et al., 2003). Social withdrawal may occur almost for these symptoms respectively (Leroi et al., 2003).
3 years prior to the onset of the full dementia syndrome, Psychotic symptoms are relatively persistent and are
and anxiety and psychotic symptoms may also be portents often seen in those with moderate or severe cognitive
of later cognitive decline (Taragano et al, 2009). Finally, impairment due to dementia (Savva et al., 2009). They most
apathy, characterized by loss of motivation and interest and commonly take the form of delusions of theft or jealously,
emotional blunting, is a frequent marker of conversion to the phantom boarder syndrome or misidentification phe-
dementia in those with early or MCI. For example, in PD, nomena (Harvey, 1996). In AD, the most common hallu-
the only neuropsychiatric symptom that differs in fre- cinations are visual (80%), which a notably more common
quency and magnitude between those with intact compared than auditory hallucinations (20%) (Leroi et al., 2003).
to those with impaired cognition is the syndrome of apathy, The prevalence of AD-related neuropsychiatric symp-
which continues to worsen as cognition declines. This sug- toms in clinic-based populations can be ascertained from the
gests that the emergence of apathy is a proxy for the onset European Alzheimer Disease Consortium’s dataset, which is
Neuropsychiatric aspects of dementia 61

the largest pooled set of data relating to people with AD. These stay, and that these and other neuropsychiatric symptoms
cross-sectional data, which are derived from 12 treatment cen- were associated with the risk of further adverse events and
tres across Europe, include Neuropsychiatric Inventory (NPI) higher mortality rates.
scores (Cummings et al., 1997) from 2,354 clinic attendees Aside from the specific setting, the prevalence of neu-
with a diagnosis of AD. They reveal that apathy is the most ropsychiatric symptoms may also vary with the specific
common symptom, appearing in 55% of clinic patients fol- dementia subtype, although the rates and profiles of these
lowed by anxiety and depression in about 37% (Aalten et al., symptoms are most often not specific enough to reliably dis-
2007). Delusions and hallucinations have a similar prevalence tinguish between the diagnostic sub-types. For example, in a
to the population-based findings from the United States. study population of people with AD and VaD, the rates with
Residential and nursing home prevalence figures differ which different NPI domains were endorsed did not signifi-
slightly from the findings of population- or clinic-based cantly differ (Leroi et al., 2003). In contrast, the neuropsychi-
cohorts of people with dementia, with over 91% of those in atric manifestations of some of the ‘frontal lobe dementias’
nursing home facilities having at least one behavioural prob- are embedded within the diagnostic characteristics and can
lem (Tariot et al., 1993) with up to 60% having aggression be used to distinguish between the subtypes, as in Table 7.1.
and non-aggressive agitation (Margallo-Lana et al., 2001). In the Lewy body diseases, such as dementia with Lewy
This high proportion may be due, in part, to the higher rate bodies (DLB) and Parkinson’s disease dementia (PDD),
of detection, as well as the more advanced stage of dementia apathy, depression and visual hallucinations are particu-
of people in 24-hour care. larly prominent. Interestingly, in PD without dementia, the
The Maryland Assisted Living Study (MALS) examined same neuropsychiatric presentation is evident, but with a
a stratified random sample of residents in assisted living lower frequency, suggesting that neuropsychiatric symp-
(AL) facilities and established that 70% has clinically signif- toms are a precursor to subsequent cognitive decline (Leroi
icant neuropsychiatric symptoms (Rosenblatt et al., 2004). et al., 2012; Javeed and Leroi, 2014). Dopamine has a role in
Residents in AL tend to be less cognitively and function- reward, motivation, emotion regulation and motor function
ally impaired than those in nursing and care home facilities. and the above findings suggests that a spectrum of similar
Interestingly, MALS revealed the magnitude and frequency but increasing pathology exists across the three disorders
of symptoms to be twice as high in smaller AL facilities depending on the distribution of Lewy bodies and subse-
(<15 beds) compared to larger AL facilities, likely reflecting quent damage to dopaminergic and cholinergic pathways
differences in care practices and even resident characteris- (Leroi et al., 2012).
tics (Leroi and Burns, 2007). This suggests that the specific
features of different residential types need to be consid-
ered when determining the prevalence of neuropsychiatric
7.7 RISK FACTORS FOR
symptoms in populations with dementia.
THE DEVELOPMENT OF
Within acute hospital settings, neuropsychiatric symp-
toms are also prevalent and need to be recognized and
NEUROPSYCHIATRIC SYMPTOMS
managed. Sampson et al. (2014) reviewed 230 patients
admitted to an acute medical unit in the United Kingdom Genetic factors may play a role in the emergence of neuro-
and found moderately or severely troubling neuropsychia- psychiatric symptoms in dementia. The CCSMA established
try symptoms in over 40% of patients. The most common an association between a positive apolipoprotein E gene
symptoms were aggression (57%) and ‘activity distur- (ApoE 4) status and aberrant motor behaviour (Steinberg
bances’ (44%). The same study also discovered that over et al., 2006). This study also showed women to have an
a third of patients reported sleep disturbances, depres- increased risk of developing anxiety and delusions in AD.
sion, phobia or anxiety at some point during their hospital Genetic variations in dopamine- and serotonin-related

Table 7.1 Diagnostic characteristics of frontal lobe dementia


Type of frontal lobe
dementia Diagnostic characteristics
Frontotemporal • Insidious onset
dementia (FTD) • Gradual decline in social functioning and regulation of personal conduct, accompanied by
(Mychack et al., 2001) emotional blunting and behaviours that may be stereotypic, perseverative or stimulus bound
• Tends to have a predominance of right-sided temporal dysfunction
• Tends to present with aberrant social behaviour, hyperorality, hypersexuality and changes in
religious and ideological belief systems
Semantic dementia (SD) • Predominance of left-sided temporal lobe pathology leads to compulsive behaviours
Progressive non-fluent • Language dysfunction is a core presenting feature
aphasia (PNA) • Behavioural symptoms are not prominent
62 Dementia

genes, such as the catechol-O-methyltransferase (COMT) The argument supporting a ‘split’ in symptoms is based
or 5-HTT gene-linked promoter region (5-HTTLPR), and on findings of distinct underlying neuropathology of dif-
neurotrophins, such as brain-derived neurotrophic factor ferent individual symptoms. For example, delusions in
(BDNF) are related to developing neuropsychiatric symp- AD have been associated with the presence of neurofibril-
toms in AD (Borroni et al., 2010). lary tangles (NFTs), and may be associated with an ‘affec-
Ropacki and Jeste (2005) reviewed 55 studies published tive syndrome’ and ‘psychotic’ syndrome. Hallucinations
on the epidemiology of psychosis in AD between 1990 and appear to be more closely associated with degeneration of
2003. This review revealed that black ethnicity, low education cholinergic pathways (Perry et al., 1990; Lyketsos et al.,
level, female gender and family history of dementia or psychi- 2000). Hence, it may be misleading to ‘lump’ these different
atric disorder to be weakly associated with increased risk for symptoms under the rubric of ‘psychosis of AD’.
psychosis in AD. However, the overall evidence for this risk
remains inconclusive except for psychosis, namely hallucina-
tions, being significantly associated with cognitive decline. 7.9 ASSESSMENT, RATING
Finally, premorbid personality traits may predispose to AND DIAGNOSIS OF THE
the later development of neuropsychiatric symptoms in AD.
NEUROPSYCHIATRIC SYMPTOMS
Specifically, lower premorbid agreeableness is associated with
agitation and irritability symptoms in AD and also predicts
OF DEMENTIA
an ‘agitation/apathy’ syndrome. The relationship between pre-
morbid neuroticism and neuropsychiatric symptoms are less The assessment of neuropsychiatric symptoms should be
straightforward, and premorbid neuroticism does not appear considered an essential part of a complete assessment of a
to be associated with depression in AD (Archer et al., 2007). patient with dementia. Several assessment tools and rating
scales have been validated and can aid in the detection and
diagnosis of the symptoms, as well as their quantification.
Quantifying the frequency and severity of the symptoms
7.8 CLASSIFICATION OF SYMPTOMS can be important, particularly as they may be the key focus
of a clinical management plan. It will also enable a clear
Various classification schemes have been proposed to cap- determination of the outcome of the therapeutic interven-
ture the range of behavioural and psychiatric presentations tion and aid in monitoring the patient’s progress. The most
in dementia. The symptoms could be ‘lumped’ or ‘split’ into commonly used and best-validated tools for assessing the
specific and definable syndromes or considered as mono- full range of symptoms include the informant-rated NPI
symptomatic phenomena. (Cummings et al., 1997), which is considered the gold stan-
As per the ‘lumping’ approach, multiple statistical meth- dard assessment tool, and is often used as an efficacy out-
ods have been employed to examine neuropsychiatric symp- come measure in clinical trials. A clinician-rated version,
toms according to ‘syndrome’. One of the largest analyses NPI-C is available in several languages (De Medeiros et al.,
was based on the European Alzheimer Disease Consortium’s 2010). There is also NPI-NH, a nursing home version, which
cross-sectional study of 2,808 patients with dementia from could be used in such institutions (Wood et al., 2000).
12 different European clinical centres (Aalten et al., 2008). Another commonly used full spectrum tool is the
Using principle component analyses, four neuropsychiatric behavioural pathology in Alzheimer disease rating scale
syndromes were identified across various dementia types and (BEHAVE-AD) (Reisberg et al., 1987). Other rating scales
included: ‘hyperactivity’ (agitation, aggression, euphoria, dis- focus on specific behavioural syndromes such as the Cornell
inhibition, irritability, aberrant motor behaviour), ‘psychosis’ Scale for Depression in Dementia (CSDD) (Alexopoulos et al.,
(hallucinations and delusions), ‘affective symptoms’ (depres- 1988), which is an informant-rated depression scale; the
sion and anxiety) and ‘apathy’ (apathy and eating behaviours). Cohen-Mansfield agitation inventory (Cohen-Mansfield,
In contrast to the factor analytic approaches, latent class 1986) for assessment of agitation and aggression; and the
analysis is a method that examines whether individuals tend apathy evaluation scale (AES) (Marin et al., 1991) for the
to cluster into groups (classes) based on their individual assessment of apathy and loss of motivation.
clinical profile rather than examining the groupings of clin- The clinical assessment of neuropsychiatric symptoms
ical characteristics across individuals, as in the former case. can be enhanced through the use of diagnostic criteria
The latter method has revealed that three groups or classes specific for particular syndromes. Such diagnostic criteria,
of AD patients typically manifest: Those with no neuropsy- although not widely used in the clinical setting, have come
chiatric symptoms (40%) or with a monosymptomatic dis- about due to attempts to operationalize the examination
turbance (19%). A second class (28%) has a predominantly of separate behavioural phenomena. The symptoms have
affective syndrome, while a third class (13%) presents with been classified into distinct sets of unique, validated crite-
a psychotic syndrome (Lyketsos et al., 2000). A similar pat- ria, similar to the approach to categorization of symptoms
tern using latent class analysis emerged from the analysis by used by the American Psychiatric Association’s Diagnostic
Moran et al. (2004), however, aggression, rather than psy- and Statistical Manual of Mental Disorders. From a research
chosis, appeared more distinctly as a separate grouping. perspective, this approach fosters reliability and enables
Neuropsychiatric aspects of dementia 63

the development of a sound evidence-base for management neuropathological and neurochemical studies have gen-
strategies. Examples of such diagnostic criteria include: erally considered hallucinations and delusions under the
‘Psychosis of AD’ (Jeste and Finkel, 2000); ‘depression of common rubric of ‘psychosis’ and suggest that those with
AD’ (Olin et al., 2002); ‘sleep disorder and AD’ and ‘agita- psychosis and AD have more extensive degenerative changes
tion and AD’ (Sunderland et al., 2008). These criteria have in frontal and temporal areas, compared with those AD
not, however, been adopted into standard clinical practice. sufferers with no psychosis (Cummings, 2000a). However,
there is growing support for considering hallucinations and
delusions separately when considering the pathophysiology
7.10 PATHOPHYSIOLOGY OF of ‘psychosis’. In particular, the association between lower
levels of choline acetyltransferase levels in parietal and tem-
NEUROPSYCHIATRIC SYMPTOMS
poral lobes of dementia sufferers with hallucinations (Perry
OF DEMENTIA et al., 1990) and the relative efficacy of cholinesterase inhibi-
tors in treating hallucinations in dementia suggests a cho-
Some of the most significant recent advances in our under- linergic basis for hallucinations (Cummings, 2000b).
standing of neuropsychiatric symptoms of dementia have Depression in dementia is common and over 30% of
arisen from new findings of the underlying pathophysiol- people may experience depression in the month prior to
ogy of these symptoms. A thorough understanding of such assessment for the presence of dementia (Lyketsos et al.,
pathophysiological processes will aid validation of the clus- 2002), suggesting that depression may be part of the pro-
tering of the neuropsychiatric symptoms and syndromes drome of dementia. Prevalence estimates of depression in
and further clarify whether mechanisms distinct from dementia vary due to multiple and complex factors, includ-
those underlying cognitive symptoms exist. By understand- ing frequent comorbidity with apathy as well as the inabil-
ing the neurobiological basis of these symptoms, appropri- ity of the depressed person with dementia to communicate
ate interventions can be employed to improve quality of life their distress and report their symptoms accurately (Lee
and reduce carer burden in AD sufferers. and Lyketsos, 2003). A family or past personal history of
Agitation and aggression occur frequently in AD and depression may also be contributory. A recent UK study,
cause a great deal of distress to both AD suffers and their studying polymorphisms in dopamine gene pathways in
caregivers. In AD, agitation and aggression in isolation those with probable AD found an association between the
may be significantly associated with impairment in activi- dopamine receptor 4 (DRD4) 2R allele and depression sug-
ties of daily living (ADL) (D’Onofrio et al., 2012), although gesting a dopaminergic system in depression (Proitsi et al.,
a proportion of those with agitation will have underlying 2010). Those developing depression after the onset of AD
delusions driving the behavioural problem. Considered will often have had a positive family history of mood dis-
an ‘enigmatic syndrome’ in AD, the underlying neurobi- order or have suffered depression in early or midlife. The
ology of agitation and aggression is still not entirely clear subsequent presentation may therefore be a recurrence of
(Cummings, 2000a). The degree of agitation has been found former symptoms. In contrast, for some, depression may
to be associated with the degree of grey matter loss in the represent an emotional reaction to cognitive deficits, cere-
anterior cingulate bilaterally and the left insula (Bruen et al., brovascular damage or a direct consequence of the neuro-
2008). There is also some evidence for a link between these degenerative process (Lee and Lyketsos, 2003). The latter
symptoms and the glutamatergic system, which may be dis- process is most likely the same as that underlying ‘late-
rupted due to the formation of NFT in frontal and cingulate life depression’, which is commonly prodromal to a later
cortices (Tekin et al., 2001). This pathology may result in dementia syndrome. Neuropathology studies have found
an exaggerated response to triggers and other symptoms serotonergic deficits in the raphe nuclei (Chen et al., 2000)
such as depression, anxiety and psychosis, through the low- and an excessive loss of noradrenergic neurones have also
ering of the threshold that triggers aggressive behaviour both been implicated (Hermann et al., 2004), allowing these
(Senanarong et al., 2004). Although some have not been mechanisms to be targets for pharmacological intervention.
able to show correlation, other studies have demonstrated Apathy, or loss of motivation or indifference, is consid-
an association between agitation and delusions with the ered the most common neuropsychiatric manifestation in
presence of ApoE-ε4 (van der Flier et al., 2007). dementia and in the EADC study was found to occur in
Psychosis is one of the most common presenting neu- almost 65% of AD patients (Aalten et al., 2007). Apathy
ropsychiatric features in dementia, particularly in AD as syndrome appears to be stable across several forms
and DLB, where visual hallucinations may be the present- of dementia, including VaD, DLB and frontotemporal
ing symptoms. Delusions occur frequently, particularly in dementia (FTD) (Aalten et al., 2008). As with depression,
the earlier stages of dementia and in those who are older apathy may be a prodrome of a dementia syndrome and in
(Leroi et al., 2003). Positron emission tomography (PET) MCI, comorbidity with the ‘lack of interest’ component of
has shown reduced glucose metabolism in right frontal the apathy syndrome predicted a higher rate of conversion
regions of the brain in those suffering from delusions, with to AD (Onyike et al., 2007; Robert et al., 2008). Apathy is
severity linked to hypometabolism in prefrontal and ante- very often under-recognized yet it has significant conse-
rior cingulate regions (Lyketsos et al., 2011). Post-mortem quences with greater levels of impairment in ADL, more
64 Dementia

associated with cognitive impairments and greater carer Banerjee, S., Smith, S.C., Lamping, D.L. et al. (2006).
burden. The most likely pathophysiology underlying apa- Quality of life in dementia: More than just cogni-
thy syndromes is that which involves a lesion at any point tion. An analysis of associations with quality of life in
along the distributed frontal-subcortical network. The dementia. Journal of Neurology, Neurosurgery and
location of the lesions may determine the particular mani- Psychiatry, 77: 146–148.
festation of the apathy syndrome. For example, lesions Benoit, M., Koulibaly, P.M., Migneco, O. et al. (2002). Brain
involving the mesiofrontal circuit, of which the anterior perfusion in Alzheimer’s disease with and without
cingulate gyrus forms a part of and which mediates goal- apathy: A SPECT study with statistical parametric map-
directed and motivated behaviour, is likely to manifest in ping analysis. Psychiatric Research Neuroimaging, 114:
lack of drive or action. Lesions involving the dorsolateral 103–111.
frontal cortex are more likely to disrupt the cognitive and Bergh, S. and Selbæk, G. (2012). The prevalence and the
executive functioning abilities required to generate goal- course of neuropsychiatric symptoms in patients with
directed behaviours (Landes et al., 2001). The importance dementia. Norsk Epidemiologi, 22 (2): 225–232.
of these regions to the apathy syndrome is supported by Bianchetti, A., Benvenuti, P., Ghisla, K.M. et al. (1997). An
various studies, including the post-mortem findings of Italian model of dementia special care units: Results of
greater NFT burden in the anterior cingulate (Marshall a preliminary study. Alzheimer Disease and Associated
et al, 2006), and the neuroimaging findings suggesting Disorders, 11: 53–56.
that apathy is associated with decreased perfusion and Borroni, B., Costanzi, C. and Padovani, A. (2010). Genetic
metabolism of fronto-subcortical brain areas and the ante- susceptibility to behavioural and psychological
rior cingulate (Benoit et al., 2002). symptoms in Alzheimer disease. Current Alzheimer
Research, 7 (2): 158–164.
Brayne, C., McCracken, C. and Matthews, F.E. (2006).
Cohort profile: The Medical Research Council
7.11 CONCLUSIONS Cognitive Function and Ageing Study (CFAS).
International Journal of Epidemiology, 35: 1140–1145.
The neuropsychiatric symptoms of the various dementia Breitner, J.C.S., Wyse, B.W., Anthony, J.C. et al. (1999).
syndromes are common and nearly universal. It is now evi- APOE epsilon-4 count predicts age when prevalence
dent that certain neuropsychiatric phenotypes are linked of Alzheimer’s disease increases then declines: The
with particular underlying pathophysiological processes. Cache County Study. Neurology, 53: 321–331.
These symptoms, which may appear even before the cog- Brodaty, H. and Donkin, M. (2009). Family caregiv-
nitive and functional decline has become apparent, have ers of people with dementia. Dialogues in Clinical
significant consequences for both the patient and their Neuroscience, 11 (2): 217.
caregiver. Bruen, P.D., McGeown, W.J., Shanks, M.F. et al. (2008).
On an optimistic note, while therapeutic approaches to Neuroanatomical correlates of neuropsychiatric symp-
the cognitive and functional decline remain limited, the toms in Alzheimer’s disease. Brain, 131: 2455–2463.
neuropsychiatric symptoms remain the most treatment Chen, C.P., Eastwood, S.L., Hope, T. et al. (2000).
responsive aspects of dementia. Immunocytochemical study of the dorsal and median
raphe nuclei in patients with Alzheimer’s disease
REFERENCES prospectively assessed for behavioural changes.
Neuropathology and Applied Neurobiology, 26:
Aalten, P., Verhey, F.R., Boziki, M. et al. (2007). 347–355.
Neuropsychiatric syndromes in dementia: Results from Cohen-Mansfield, J. (1986). Agitated behaviors in the
the European Alzheimer disease consortium. Dementia elderly: II. Preliminary results in the cognitively dete-
and Geriatric Cognitive Disorders, 24: 457–463. riorated. Journal of the American Geriatric Society, 34:
Aalten, P., Verhey, F.R., Boziki, M. et al. (2008). 722–727.
Consistency of neuropsychiatric syndromes across Cummings, J., Mega, M., Gray, K., Rosenberg-Thompson,
dementias: Results from the European Alzheimer S., Carusi, D. A., and Gornbein, J. (1994). The
disease consortium. Dementia and Geriatric Cognitive Neuropsychiatric Inventory: Comprehensive assess-
Disorders, 25: 1–8. ment of psychopathology in dementia. Neurology, 44,
Alexopoulos, G.S., Abrams, R.C., Young, R.C. and 2308–2314.
Shamoian, C.A. (1988). Cornell scale for depression in Cummings, J.L. (2000a). Cognitive and behavioral hetero-
dementia. Biological Psychiatry, 23: 271–284. geneity in Alzheimer’s disease: Seeking the neurobio-
Archer, N., Brown, R.G., Reeves, S.J. et al. (2007). logical basis. Neurobiology of Aging, 21: 845–861.
Premorbid personality and behavioral and psychologi- Cummings, J.L. (2000b). Cholinesterase inhibitors: A new
cal symptoms in probable Alzheimer disease. American class of psychotropic compounds. American Journal of
Journal of Geriatric Psychiatry, 15 (3): 202–213. Psychiatry, 157: 4–15.
Neuropsychiatric aspects of dementia 65

De Medeiros, K., Robert, P., Gauthier, S. et al. (2010). The Leroi, I., Perera, N., Harbeshettar, V. and Robert, P. (2014).
Neuropsychiatric Inventory-Clinician rating scale (NPI- Apathy and emotional blunting in Parkinson’s disease.
C): Reliability and validity of a revised assessment of Brain Disorders Therapy, 3 (5): 1–8.
neuropsychiatric symptoms in dementia. International Leroi, I., Voulgari, A., Breitner, J.C.S. and Lyketsos, C.G.
Psychogeriatrics, 22 (6): 984–994. (2003). The epidemiology of psychosis in dementia.
D’Onofrio, G., Sancarlo, D., Panza, F. et al. (2012). American Journal of Geriatric Psychiatry, 11: 83–91.
Neuropsychiatric symptoms and functional status in Lyketsos, C.G., Carrillo, M.C., Ryan, J.M. et al. (2011).
Alzheimer’s disease and vascular dementia patients. Neuropsychiatric symptoms in Alzheimer’s disease.
Current Alzheimer Research, 9 (6): 759. Alzheimer’s and Dementia, 7 (5): 532–539.
Georges, J., Jansen, S., Jackson, J. et al. (2008). Lyketsos, C.G., Lopez, O., Jones, B. et al. (2002).
Alzheimer’s disease in real life – The dementia carer’s Prevalence of neuropsychiatric symptoms in demen-
survey. International Journal of Geriatric Psychiatry, 23 tia and mild cognitive impairment: Results from the
(5): 546–551. cardiovascular health study. Journal of the American
Green, R.C., Cupples, L., Kurz, A. et al. (2003). Depression Medical Association, 288: 1475–1483.
as a risk factor for Alzheimer disease: The MIRAGE Lyketsos, C.G., Steinberg, M., Tschanz, J.T. et al. (2000).
study. Archives of Neurology, 60 (5): 753–759. Mental and behavioural disturbances in dementia:
Gonzalez-Salvador, T., Arango, C., Lyketsos, C.G. and Findings from the Cache County study on memory
Barba, A.C. (1999). The stress and psychological mor- aging. American Journal of Psychiatry, 157: 708–714.
bidity of the Alzheimer patient caregiver. International Margallo‐Lana, M., Swann, A., O’Brien, J. et al. (2001).
Journal of Geriatric Psychiatry, 14: 701–710. Prevalence and pharmacological management of behav-
Harvey, R.J. (1996). Review: Delusions in dementia. Age ioural and psychological symptoms amongst dementia
and Ageing, 25 (5): 405–408. sufferers living in care environments. International
Hermann, N., Lanctot, K.L. and Khan, L.R. (2004). The role Journal of Geriatric Psychiatry, 16 (1): 39–44.
of norepinephrine in the behavioural and psychologi- Marin, R.S., Biedrzycki, R.C. and Firinciogullari, S. (1991).
cal symptoms of dementia. Journal of Neuropsychiatry Reliability and validity of the apathy evaluation scale.
and Clinical Neurosciences, 16: 261–276. Psychiatry Research, 38: 143–162.
Hwang, T.J., Masterman, D.L., Ortiz, F. et al. (2004). Mild Marshall, G.A., Fairbanks, L.A., Tekin, S. et al. (2006).
cognitive impairment is associated with characteristic Neuropathologic correlates of apathy in Alzheimer’s
neuropsychiatric symptoms. Alzheimer’s Disease and disease. Dementia and Geriatric Cognitive Disorders,
Associated Disorders, 18: 17–21. 21 (3): 144–147.
Jarvik, L. and Greenson, H. (1987). Translation of ‘About Mauri, M., Sinforiani, E., Zucchella, C. et al. (2012).
a peculiar disease of the cerebral cortex’, 1907 by Progression to dementia in a population with amnestic
A. Alzheimer. Alzheimer Diseases and Associated mild cognitive impairment: Clinical variables associated
Disorders, 1: 7–8. with conversion. Functional Neurology, 27 (1): 49–54.
Javeed, M. and Leroi, I. (2014). Neuropsychiatric features Moran, M., Walsh, C., Lynch, A. et al. (2004). Syndromes
of Parkinson’s disease with cognitive impairment: An of behavioural and psychological symptoms in mild
overview. EMJ Neurology, 1: 95–102. Alzheimer’s disease. International Journal of Geriatric
Jeste, V. and Finkel, S.I. (2000). Psychosis of Alzheimer’s Psychiatry, 19: 359–364.
disease and related dementias: Diagnostic criteria for Murman, D.L., Chen, Q., Powell, M.C. et al. (2002). The
a distinct syndrome. American Journal of Geriatric incremental direct cost associated with behavioral
Psychiatry, 8: 29–34. symptoms in AD. Neurology, 59: 1721–1729.
Landes, A.M., Sperry, S.D., Strauss, M.E. and Mychack, P., Kramer, J.H., Boone, K.B. and Miller, B.L.
Geldmacher, D.S. (2001). Apathy in Alzheimer’s dis- (2001). The influence of right frontotemporal dysfunc-
ease. Journal of the American Geriatrics Society, 49: tion on social behavior in frontotemporal dementia.
1700–1707. Neurology, 56 (Suppl. 4): S11–S15.
Lee, H.B. and Lyketsos, C.G. (2003). Depression in O’Donnell, B.F., Drachman, D.A., Barned, H.J. et al. (1992).
Alzheimer’s disease: Heterogeneity and related issues. Incontinence and troublesome behaviors predict
Biological Psychiatry, 54: 353-62. institutionalisation in dementia. Journal of Geriatric
Leroi, I. and Burns, A.B. (2007). Behavioural and psy- Psychiatry and Neurology, 5: 45–52.
chological symptoms of dementia associated with Olin, J.T., Schneider, L.S., Katz, I.R. et al. (2002). Provisional
Parkinson’s disease (BPSD-PD). Journal of Neurology, diagnostic criteria for depression of Alzheimer disease.
Neurosurgery and Psychiatry, 78 (1): 2–3. American Journal of Geriatric Psychiatry, 10: 125–128.
Leroi, I., Pantula, H., McDonald, K. and Harbeshettar, Onyike, C.U., Sheppard, J.M., Tschanz, J.T. et al. (2007).
V. (2012). Neuropsychiatric symptoms in Parkinson’s Epidemiology of apathy in older adults: The Cache
disease with mild cognitive impairment and dementia. County Study. American Journal of Geriatric Psychiatry,
Parkinson’s Disease, 2012: 308097. 15 (5): 365–375.
66 Dementia

Paulsen, J.S., Salmon, D.P., Thai, L.J. et al. (2000). Incidence Senanarong, V., Cummings, J.L., Fairbanks, L. et al. (2004).
of and risk factors for hallucinations and delusions in Agitation in Alzheimer’s disease is a manifestation
patients with probable AD. Neurology, 54: 1965–1971. of frontal lobe dysfunction. Dementia and Geriatric
Perry, E.K., Marshall, E., Kerwin, J. et al. (1990). Evidence Cognitive Disorders, 17: 14–20.
of a monoaminergic-cholinergic imbalance related to Steinberg, M., Corcoran, C., Tschanz, J.T. et al. (2006).
visual hallucinations in Lewy body dementia. Journal of Risk factors for neuropsychiatric symptoms in demen-
Neurochemistry, 55: 1454–1456. tia: The Cache County Study. International Journal of
Proitsi, P., Lupton, M.K., Reeves, S.J. et al. (2010). Geriatric Psychiatry, 21 (9): 824–830.
Association of serotonin and dopamine gene path- Sunderland, T., Jeste, D.V., Baiyewu, O. et al. (2008).
ways with behavioral subphenotypes in dementia. Diagnostic issues in dementia: Advancing the research
Neurobiology Aging, 33 (4): 791–803. agenda for DSM-V. American Psychiatric Association,
Reisberg, B., Borenstein, J., Salob, S.P. et al. (1987). 78–79.
Behavioral symptoms in Alzheimer’s disease: Taragano, F., Allegri, R., Krupitzki, H. et al. (2009). Mild
Phenomenology and treatment. Journal of Clinical behavioral impairment and risk of dementia. Journal of
Psychiatry, 48 (Suppl.): 9–15. Clinical Psychiatry, 70 (4): 584–592.
Robert, P.H., Berr, C., Volteau, M. et al. (2008). Importance Tariot, P.N., Podgorski, C.A., Blazina, L. and Leibovici, A.
of lack of interest in patients with mild cognitive (1993). Mental disorders in the nursing home: Another
impairment. American Journal of geriatric Psychiatry, perspective. American Journal of Psychiatry, 150 (7):
16: 770–776. 1063–1069.
Ropacki, S.A. and Jeste, D.V. (2005). Epidemiology of Tekin, S., Mega, M.S., Masterman, D.M. et al. (2001).
and risk factors for psychosis of Alzheimer’s disease: Orbitofrontal and anterior cingulated cortex neuro-
A review of 55 studies published from 1990 to 2003. fibrillary tangle burden is associated with agitation
American Journal of Psychiatry, 162 (11): 2022–2030. in Alzheimer disease. Annals of Neurology, 49 (3):
Rosenblatt, A., Samus, Q.M., Steele, C.D. et al. (2004). 355–361.
The Maryland assisted living study: Prevalence, recog- van der Flier, W.M., Staekenborg, S., Pijnenburg, Y.A.L.
nition and treatment of dementia and other psychiatric et al. (2007). Apolipoprotein E genotype influences
disorders in the assisted living population of central presence and severity of delusions and aggressive
Maryland. Journal of the American Geriatrics Society, behavior in Alzheimer disease. Dementia and Geriatric
52 (10): 1618–1625. Cognitive Disorders, 23: 42–46.
Sampson, E.L., White, N., Leurent, B. et al. (2014). Wilson, R.S., Barnes, L.L., Mendes de Leon, C.F. et al.
Behavioural and psychiatric symptoms in people with (2002). Depressive symptoms, cognitive decline, and
dementia admitted to the acute hospital: Prospective risk of Alzheimer disease in older persons. Neurology,
cohort study. British Journal of Psychiatry, 205 (3): 59: 364–370.
189–196. Wood, S., Cummings, J.L., Hsu, M.-A. et al. (2000). The
Savva, G.M., Zaccai, J., Matthews, F.E. et al. (2009). use of the neuropsychiatric inventory in nursing
Prevalence, correlates and course of behavioural and home residents: Characterization and measurement.
psychological symptoms of dementia in the popula- American Journal of Geriatric Psychiatry, 8: 75–83.
tion. British Journal of Psychiatry, 194 (3): 212–219.
8
Measurement of behaviour disturbance,
non-cognitive symptoms and quality of life

AJIT SHAH

properties of instruments measuring BPSD are illustrated


8.1 BEHAVIOURAL AND in Table 8.2. An important characteristic of some instru-
PSYCHOLOGICAL SYMPTOMS OF ments measuring aggression is the spontaneous tendency of
DEMENTIA aggression declining during serial measurements (Nilsson
et al., 1988; Shah, 1999b).
8.1.1 DEFINITION 8.1.2.3 Comment
Behavioural and psychological symptoms of dementia Although many instruments measure aggression, not all
(BPSD) are defined as ‘a heterogeneous range of psychologi- the psychometric and other properties have been adequately
cal reactions, psychiatric symptoms and behaviours occur- evaluated. Individual instruments have been developed for
ring in people with dementia of any aetiology’ (Finkel and use in specific settings, for specific diagnostic groups and
Burns, 2000). They include disorders of behaviour, mood, by specific groups of raters and their generalizability to
thought content and perception, and personality alteration other settings, diagnostic groups and categories of raters are
(Burns et al., 1990b, 1990c, 1990d, 1990e; Shah et al., 2005). unclear. For example, the rating scale for aggressive behav-
The definition of any individual BPSD should be hierarchi- iour in the elderly (RAGE), although developed for use
cal and allow discrimination among the cognitive and other by nursing staff to measure aggression in psychogeriatric
BPSD domains, different BPSD domains and individual inpatients with dementia (Patel and Hope, 1992a), has been
symptoms within an estimated BPSD domain (Shah, 2000). used in nursing homes for all diagnostic groups (Shah et al.,
1997). Nevertheless, comparisons among individual scales
8.1.2 DISORDERS OF BEHAVIOUR are emerging (Whall et al., 2013).

8.1.2.1 Definition 8.1.3 DISORDERS OF PERSONALITY


ALTERATION
A typical example of disturbed behaviour is aggression, but
similar arguments can be rehearsed for other behaviours. 8.1.3.1 Definition
Aggression has been variably defined (Shah, 1999a), but a
useful definition is: ‘Aggressive behaviour is an overt act, Personality changes in dementia include emergence of new
involving the delivery of noxious stimuli to (but not nec- traits (Dian et al., 1990; Siegler et al., 1991; Chatterjee et al.,
essarily aimed) at another organism, object or self, which 1992; Petry et al., 1988; Lautenschlager et al., 2007) or exag-
clearly is not accidental’ (Patel and Hope, 1992a). geration of existing traits (Lautenschlager et al., 2007).

8.1.2.2 Samples, settings and data 8.1.3.2 Samples, settings and data


collection methods collection methods
Table 8.1 illustrates the samples studied and methods of Table 8.3 illustrates the data collection methods, ­samples
data collection in several settings (Shah, 1999a). Essential and the settings examining personality alterations.
67
68 Dementia

Table 8.1 Aggression: samples studied and methods of data collection


Samples
Undifferentiated dementia Haider and Shah (2004), Shah et al. (2004, 2005), Hinton et al. (2008),
Koopmans et al. (2009), Truzzi et al. (2013), Whall et al. (2013)
Alzheimer’s disease Burns et al. (1990b, 1990f, 1990a), Devanand et al. (1992a), Lyketos et al.
(2001), Shah et al. (2005), Senanarong et al. (2005)
Vascular dementia Sultzer et al. (1993), Shah et al. (2005), Pinto and Seethalakshmi (2006)
Huntington’s chorea Burns et al. (1990a)

Methods of Data Collection


Case-notes Haider and Shah (2004)
Semi-structured telephone interview Devanand et al. (1992a)
Postal questionnaire directed at staff Lukovits and McDaniel (1992)
Postal questionnaire directed at informal carers Lukovits and McDaniel (1992)
Staff completed incident forms Shah (1995)
Informal staff reports Hallberg et al. (1993)
Staff completed specially designed forms Winger et al. (1987), Palmsteirna and Wistedt (1987)
Staff completed formal rating scales Koopmans et al. (2009), Whall et al. (2013)
Informal carers completed formal rating scales Ryden (1988)
Informal carers completed informal forms Onishi et al. (2006)
Semi-structured interviews Devanand et al. (1992b), Senanarong et al. (2005), Shah et al. (2004, 2005),
Pinto and Seethalakshmi (2006), Hinton et al. (2008), Truzzi et al. (2013)
Direct observations Hallberg et al. (1993)
Voice recordings Hallberg et al. (1990)
Mechanical body movements Rindlisbacher and Hopkins (1992)

Table 8.2 Psychometric and other properties Studies included a comparison group of normal aged
of instruments measuring BPSD in dementia individuals (Rubins et al., 1987a, 1987b; Petry et al.,
1988; Cummings et al., 1990; Dian et al., 1990; Jacoub
Patient characteristics
User characteristics
and Jorm, 1996; Rankin et al., 2005; Talassi et al., 2007),
Type of instruments
vascular dementia (Sultzer et al., 1993) and frontotem-
Setting for administration
poral dementia (Miller et al., 1997; Rankin et al., 2005)
Data source
and compared Alzheimer’s disease (AD) with norms for
Output of the instrument inpatients (Meins and Dammast, 2000). Comparison
Psychometric properties with premorbid personality and normal aged individuals
Reliability: allows identification of personality changes (Siegler et al.,
Test-retest 1991; Chatterjee et al., 1992) and exclusion of personality
Inter-rater changes due to ageing (Rubins et al., 1987a, 1987b; Petry
Internal consistency et al., 1988; Cummings et al., 1990; Dian et al., 1990;
Validity: Rankin et al., 2005; Talassi et al., 2007), respectively.
Face Difficulties in measuring the type, nature and severity of
Content personality change include: reduced insight; judgement
Concurrent and memory may preclude use of self-reports (Petry et al.,
Construct 1988; Meins and Dammast, 2000; Rankin et al., 2005),
Predictive particularly, in frontotemporal dementia (Rankin et al.,
Incremental 2005); collateral account from relatives may be biased due
Sensitivity to change to emotions involved in it (Dian et al., 1990; Jacoub et al.,
Spontaneous tendency to decline over serial measurements 1994; Meins and Dammast, 2000), personality of the rela-
Time frame for symptoms tive acting as the carer and carer burden (Welleford et al.,
Training needs 1995; Meins, 2000).
Methods of administration
Qualification of users 8.1.3.3 Measurement instruments
Acceptability to rates
Costs The items irritability and disinhibition on the Neuro­
Source: Adapted from Zaudig, M, International Psychogeriatrics, psychiatric Inventory (NPI) (Cummings et al., 1994) cou-
8, 183–200, 1996. pled with case-note review examined socially disruptive
Measurement of behaviour disturbance, non-cognitive symptoms and quality of life 69

Table 8.3 Samples, settings and methods of data collection for personality alteration
Samples
Undifferentiated dementias Siegler et al. (1991), Balsis et al. (2005)
Vascular dementia Dian et al. (1990), Sultzer et al. (1993), O’Connor (2000)
Alzheimer’s disease Rubins et al. (1987a, 1987b), Petry et al. (1988, 1989), Bozzola et al. (1992),
Chatterjee et al. (1992), Sultzer et al. (1993), Jacoub and Jorm (1996), Meins
and Dammast (2000), Rankin et al. (2005), Talassi et al. (2007)
Frontotemporal dementia Miller et al. (1997), Rankin et al. (2005), Mendez et al. (2006)
Huntington’s chorea Kirkwood et al. (2002)

Settings
Community sample Nilsson (1983), Jacoub et al. (1994), O’Connor (2000)
Dementia centre Miller et al. (1997)
Memory clinic sample Meins and Dammast (2000)
Mixed group of psychogeriatric inpatients Pearson (1990, 1992)

Methods of Data Collection


Specially designed personality Petry et al. (1988, 1989), Chaterjee et al. (1992), Dian et al. (1990), Siegel et al.
questionnaires administered to (1991), Bozzola et al. (1992), Jacoub et al. (1994), Jacoub and Jorm (1996),
relatives Talassi et al. (2007), Balsis et al. (2005), Rankin et al. (2005)
As above and an open interview Rubins et al. (1988a,b)
Specially designed personality Nilsson (1983), Pearson (1990, 1992), Rankin et al. (2005)
questionnaire administered to patients

and antisocial behaviour in AD and frontotemporal demen- enthusiastic, less stable, more unreasonable, more lifeless,
tia (Miller et al., 1997). The Brooks and McKinlay (1983) more unhappy, less affectionate, less kind, more irritable
personality inventory, used originally on patients who have and less generous after the onset of dementia (Petry et al.,
undergone a brain injury, was administered to relatives 1988); similar results were observed in vascular demen-
of dementia sufferers (Petry et al., 1988, 1989; Cummings tia (Dian et al., 1990). A significant shift from positive
et al., 1990; Talassi et al., 2007). The 11 personality items on to negative characteristics was observed after the onset
the Blessed Dementia Rating Scale (Blessed et al., 1968) were of AD in 12 out of the 18 adjectives forming the Brooks
alone used (Bozzola et al., 1992; Balsis et al., 2005) combined and McKinlay Personality Inventory (Talassi et al., 2007).
with six open-ended questions (Rubins et al., 1987a, 1987b). Acquired extroversion has been reported to be associ-
In psychiatric inpatients and day patients with organic ated with frontotemporal dementia (Mendez et al., 2006).
disorders, impulsiveness correlated with extraversion but Forensic behaviours, like indecent exposure and shoplift-
psychoticism was not noticed on the Eysenck Personality ing were more common in frontotemporal dementia than
Inventory (EPI) (Pearson, 1990, 1992). Subjects with mild– AD (Miller et al., 1997). A 3-year follow-up projects emerg-
moderate dementia and neurotic disorders scored high for ing of four patterns of personality alteration in AD: (1)
neuroticism on the EPI in a community sample (Nilsson, change at onset with little change as the disease progresses,
1983). The premorbid trait of neuroticism in AD was associ- (2) ongoing change as the disease progresses, (3) no change
ated with ‘troublesome behaviour’ (Meins et al., 1998). at all and (4) regression of previously altered personality
characteristics (Petry et al., 1989). Personality changes can
8.1.3.4 Clinical features precede a clinical diagnosis of AD (Balsis et al., 2005) and
Huntington’s disease (Kirkwood et al., 2002).
Changes in personality features include: coarsening of
affect, disinhibition, increase in passivity, apathy, spon- 8.1.3.5 Comment
taneity, irritability, belligerence, demanding attention,
indifference, egocentricity, less conscientiousness, lower The relationship between personality changes and BPSD
extraversion, higher neuroticism and higher openness may be part of the same phenomena, aetiologically linked
(Seltzer and Sherwin, 1983; Ishii et al., 1986; Petry et al., or discrete entities. Several personality change features
1988, 1989; Cummings et al., 1990; Siegler et al., 1991; listed above overlap with features of frontal lobe dysfunc-
Bozzola et al., 1992; Chatterjee et al., 1992; Welleford tion (Dian et al., 1990) and depression (Sultzer et al., 1993).
et al., 1995; Jacoub and Jorm, 1996; Meins and Dammast, Paucity of data on psychometric and other properties of
2000). Patients with AD, compared with normal elderly instruments measuring personality suggests the need for
people, had greater changes on 12 personality traits by rigorous evaluation of existing instruments (Strauss and
being more out of touch, less self-reliant, less mature, less Pasupathi, 1994) and development of new instruments
70 Dementia

measuring personality features that are sensitive from the Visser, 2000), instruments have been validated against def-
premorbid to illness state and allow separation of personal- initions of depression according to Diagnostic and Statis­
ity changes due to ageing (Strauss et al., 1997); sadly, this has tical Manual of Mental Disorders, Third Edition (DSM-III)
not occurred so far. (Reifler et al., 1986; Cummings et al., 1987; MacKenzie
et al., 1989), Diagnostic and Statistical Manual of Mental
Disorders, Third Edition, Revised (DSM-IIIR) (Burns et al.,
8.1.4 DISORDERS OF MOOD 1990e; Skoog, 1993; Weiner et al., 1997), Diagnostic and
Statistical Manual of Mental Disorders, Fourth Edition
8.1.4.1 Definition (DSM-IV) (Brodaty and Luscombe, 1996), Research Dia­
gnostic Criteria (Alexopoulos et al., 1988) and Tenth
Depressed patients may have cognitive deficits and depres- Revision of the International Classification of Diseases and
sion may precede the onset of dementia either as an early Related Health Problems (ICD-10) (Burns et al., 1990c).
presentation of incipient dementia or as a risk factor (Verhey
and Visser, 2000; Jorm, 2001; Korcyzn and Halperin, 2009).
Also, depressive symptoms and illness in dementia require 8.1.4.3 Measurement instruments
careful definition due to overlap of features such as per-
sonality changes and cognitive impairment, especially Table 8.4 illustrates the self-rated and interviewer-rated
frontal lobe dysfunction (Weiner et al., 1997) and apathy scales. Data on psychometric and other properties for
(Starkstein et al., 2005). these instruments are gradually emerging (Korner et al.,
2006). They are generally less accurate as cognitive impair-
ment increases and insight decreases (Ott and Fogel,
8.1.4.2 Samples, settings and data 1992) because good attention, concentration, memory and
collection methods judgement are required for their completion (Burke et al.,
1989) and the discrepancy between depression reported by
Table 8.4 illustrates the diagnostic categories, settings and patients and carers increases in this context (Ott and Fogel,
methods of data collection. Despite an absence of ‘gold 1992; Cohen-Mansfield et al., 2013). Thus, instruments
standard’ diagnosis of depression in dementia (Verhey and using collateral sources of information (e.g. relatives or

Table 8.4 Sample, settings and methods of data collection for disorders of mood
Samples
Undifferentiated dementia Shah et al. (2004), Aalten et al. (2006)
Vascular dementia Cummings et al. (1987), Brodaty and Luscombe (1996),
Shah et al. (2005)
Alzheimer’s disease Burns et al. (1990c), Lyketsos et al. (2001), Shah et al.
(2005), Lam et al. (2006), Debruyne et al. (2009)

Settings
Community Lyketsos et al. (2001), Shah et al. (2004, 2005)
Outpatient clinic Cummings et al. (1987), Brodaty and Luscombe (1996)
Nursing homes Brodaty et al. (2001)
Mixed settings Alexopoulos et al. (1988), Burns et al. (1990c)
Acute and continuing care psychogeriatric wards Akoo and Shah (1998), Ellanchenny and Shah (2001)

Methods of Data Collection


Self-rating scales Ott and Fogel (1992), Korner et al. (2006), Debruyne et al.
(2009)
Interviewer-rated scales directed at patients Cummings et al. (1987), Ott and Fogel (1992), Brodaty and
Luscombe (1996)
Interviewer-rated scales directed at carers Brodaty and Luscombe (1996), Shah et al. (2004, 2005),
Aalten et al. (2006), Lam et al. (2006), Debruyne et al.
(2009)
Combination of interviewer and observer rating Sunderland et al. (1988), Ott and Fogel (1992)
Combination of interview and collateral sources Katona and Aldridge (1985), Alexopoulos et al. (1988),
Korner et al. (2006)
Pure observer scales Shah and Gray (1997), Akoo and Shah (1998), Ellanchenny
and Shah (2001)
Measurement of behaviour disturbance, non-cognitive symptoms and quality of life 71

nurses) to supplement clinical examination were developed: 8.1.5.3 Comment


Depressive Signs Scale (Katona and Aldridge, 1985), the
Cornell Scale (Alexopoulous et al., 1988) and the Depression The prevalence of delusions (Cummings et al., 1987; Burns
in Dementia Mood Scale (Sunderland et al., 1988). The et al., 1990d; Selbeck et al., 2014), hallucinations (Burns
Cornell Scale has been used most widely (Shah et al., 2004, et al., 1990e; Ballard et al., 1995) in AD and vascular
2005; Aalten et al., 2006; Korner et al., 2006; Debruyne dementia range from 20% to 50%, 17% to 36%; 11% to 34%,
et al., 2009). The Geriatric Depression Scale (GDS) has also respectively. The boundaries between hallucinations, delu-
been proved to have good psychometric properties (Korner sions and misidentification syndromes are often blurred
et al., 2006). (Whitehouse et al., 1996) and delusions are difficult to dif-
ferentiate from confabulations (Cummings et al., 1987). The
definitions and descriptions of the psychopathology require
8.1.4.4 Comment refinement and clarity (Trabucchi and Bianchetti, 1996).
For example, professional caregivers and family caregiv-
The prevalence of depressive symptoms and illness in AD
ers may give different information to complete the whole
was 0%–87% (median 41%) and 0%–86% (median 19%),
picture of delusions and hallucinations (Cohen-Mansfeild
respectively (Wragg and Jeste, 1989; Verhey and Visser,
et al., 2013) and symptoms keep changing with the pas-
2000; Starkstein et al., 2005). This variability may be due
sage of time (Selbaek et al., 2014). A systematized checklist
to: measurement instruments being insensitive in severe
containing 17 categories of psychotic symptoms is the only
dementia or those with severe dementia may not experi-
pure instrument designed to measure psychotic features,
ence complex emotions like depression (Burns et al., 1990c);
but poorly evaluated (Ballard et al., 1995). Psychometric
depression in dementia may lack traditional clinical fea-
and other properties of instruments measuring psychotic
tures (Verhey and Visser, 2000) and depressive symptoms
symptoms have been poorly studied and require rigorous
may be transient (Ballard et al., 2001).
evaluation. Clinicians will generally treat psychotic symp-
toms in dementia only if the patient is distressed or if there
is risk of harm to others or self, but none of the extant
8.1.5 DISORDERS OF THOUGHT
instruments measure these facets and such instruments
CONTENT AND PERCEPTION need to be developed.
8.1.5.1 Definition and classification
8.1.6 A WAY FORWARD
The definitions of hallucinations and delusions in dementia
are the same as for other psychiatric disorders (Cummings Research should be directed at comparing different instru-
et al., 1987). Presence of at least 7 days is an added require- ments measuring the same BPSD domain with each other,
ment to exclude delirium. Auditory (Cummings et al., 1987; generating data on all of their properties, refining these
Burns et al., 1990e), visual (Cummings et al., 1987; Burns instruments to improve their properties, adapting them
et al., 1990e) and olfactory (Rubins and Kinscherf, 1989) for use in different settings, different dementia diagnos-
hallucinations have been studied. Delusions were then tics groups, different severities of cognitive impairment
divided in four groups: (1) simple persecutory, (2) complex and by introducing different types of raters to enable data
persecutory, (3) grandiose and (4) those associated with spe- collection in a comparable common currency, as well as
cific neurological deficits (Cummings, 1985). Another clas- develop guidelines and protocols for their use in research
sification for delusions was delusions of theft, delusions of and clinical practice (Shah and Allen, 1999); this would be
suspicion and systematized delusions (Burns et al., 1990d). the beginning of a revolution which is yet to happen (Whall
There are four types of misidentifications: (1) people in the et al., 2013). This, in turn, would facilitate examination of
house, (2) misidentification of mirror images, (3) misiden- the interrelationship between individual BPSD symptoms
tification of television and (4) misidentification of people within and across different BPSD domains (Van Der Linde
outside the house (Burns et al., 1990e). et al., 2014). This is also beginning to happen with uni-
variate analysis (Brodaty et al., 2001), latent class analysis
(Lyketsos et al., 2001; Lam et al., 2006) and factor analysis
8.1.5.2 Samples, settings, data collection (Hope et al., 1997; Dechamps et al., 2008; Youn et al., 2008;
methods and measurement Savva et al., 2009; Truzzi et al., 2013). Ultimately, such anal-
instruments ysis will allow identification of the nosological validity of
the empirically derived five BPSD domains by establishing
Table 8.5 illustrates the samples, settings, methods of data symptom and syndrome clusters (Lyketsos et al., 2001; Lam
collection and instruments used to measure psychotic et al., 2006) and, in turn, facilitate a greater understanding
symptoms. Several semi-structured interviews and spe- of the demographic, clinical, genetic, biochemical, neuro-
cially designed forms measure psychotic symptoms along physiological and neuroanatomical correlates of BPSD and
with several other BPSD, thus producing diluted data on their longitudinal course and prognosis; this too is in the
pure psychotic features (Ballard et al., 1995). beginning stages (Trzepacz et al., 2013).
72 Dementia

Table 8.5 Samples, settings, methods of data collection and instruments used to measure psychotic symptoms
Setting
All nursing home residents Brodaty et al. (2001)
Undifferentiated dementia Haider and Shah (2004), Shah et al. (2004), Aalten et al. (2006), Onishi et al.
(2006), Hinton et al. (2008), Dechamps et al. (2008), Koopmans et al. (2009)
Vascular dementia Cummings et al. (1987), Shah et al. (2005), Hsieh et al. (2009)
Lewy body dementia Ballard et al. (1995)
Alzheimer’s disease Cummings et al. (1987), Burns et al. (1990d, 1990e), Lyketsos et al. (2001), Shah
et al. (2005), Lam et al. (2006), Pinto and Seethalaskmi (2006), Hancock and
Lavner (2008), Hsieh et al. (2009)
Fronto-temporal dementia Hancock and Lavner (2008)

Setting
Community Skoog (1993), Lyketsos et al. (2001), Aalten et al. (2006), Hinton et al. (2008)
Memory clinic Ballard et al. (1995), Hancock and Lavner (2008)
Outpatient clinic Cummings et al. (1987), Lam et al. (2006)
Referrals to psychogeriatric service Ballard et al. (1995), Shah et al. (2004, 2005)
Day hospital Haider and Shah (2004)
Inpatients Trabucchi and Bianchette (1996)
Nursing homes Brodaty et al. (2001), Onishi et al. (2006), Hinton et al. (2008), Dechamps et al.
(2008), Koopmans et al. (2009)
Continuing care Onishi et al. (2006)
Group homes Onishi et al. (2006)
Mixed Burns et al. (1990d, 1990e), Whitehouse et al. (1996), Pinto and Seethalaskmi (2006)

Methods of Data Collection


Patient interview Cummings et al. (1987)
Interview with caregiver Shah et al. (2004, 2005), Aalten et al. (2006), Lam et al. (2006), Pinto and
Seethalaskmi (2006), Hinton et al. (2008), Dechamps et al. (2008), Koopmans
et al. (2009)
Formal scales completed by caregivers Hancock and Lavner (2008)
Unvalidated scales completed by Onisihi et al. (2006)
caregivers
Case-notes Cummings et al. (1987), Haider and Shah (2004)

More recently, a narrower concept, health-related quality of


8.2 QUALITY OF LIFE life (HRQoL) has emerged. HRQoL has been defined as ‘the
individual’s subjective perception of the impact of a health
8.2.1 DEFINITION OF QUALITY OF LIFE condition on life’ (Smith et al., 2003; Banerjee et al., 2002).

There are several definitions of quality of life (QoL) and 8.2.2 MEASUREMENT OF QOL AND
sometimes it is not defined (Lawton, 1997). The best defini- MEASUREMENT INSTRUMENTS
tion is that from the World Health Organization (WHO):
‘individuals perception of their position in life in the con- The measurement of QoL in dementia poses unique chal-
text of the culture and value system in which they live, and lenges. Impairments in memory, language and judgement
in relation to their goals, expectations, standards and con- can interfere with the self-evaluation of dementia, partic-
cerns’ (WHOQOL Group, 1995). ularly in the later advanced stages of the disease. The lack
QoL is a multidimensional construct that includes the of a universally accepted definition of QoL or a ‘gold stan-
subjective experience of the patient in addition to objective dard’ measure of QoL raises important questions about the
criteria (Lawton, 1994; Ettema et al., 2005). Lawton (1991) validity of available measures (Shah et al., 2005). Concerns,
described QoL as a multidimensional construct that should such as these, have led to questions such as, whether instru-
include objective (observable) indices of well-being, judged ments to measure QoL in dementia should be self-rating,
against socio-normative criteria, in addition to the individ- proxy-rating or a combination of both (Banerjee et al., 2002;
ual’s own subjective perception of his or her position in life. Smith et al., 2003; Davies et al., 2014; Moyle et al., 2014).
Measurement of behaviour disturbance, non-cognitive symptoms and quality of life 73

Proxy measures by informal or professional carers may be symptoms, physical morbidity and temporary fluctuations
necessary in severe dementia when patients have poor com- make the subjective assessment of QoL difficult. However,
prehension and communication skills (Smith et al., 2007; the measurement of QoL is beneficial in the screening and
Davies et al., 2014; Moyle et al., 2014). monitoring of psychosocial problems in individual patients,
Generic and dementia-specific instruments have been population surveys of perceived health problems, clinical
used to measure QoL in dementia. Generic measures allow audit, measuring outcome in health service and evaluation
for comparisons with other diseases, but the use of demen- research, clinical trials and in cost-utility analysis (CUA).
tia-specific measures of QoL have been favoured because Future research efforts should focus on: developing an
the symptoms of dementia are very different from those of agreed definition of QoL in dementia with operational crite-
other disorders (Rabins and Kasper, 1997; Selai et al., 2001). ria; developing (existing) instruments that have robust psy-
Selected instruments for measuring QoL in dementia are chometric data that are valid, reliable, sensitive to change
illustrated in Table 8.6. and can be used across various care settings and with differ-
ent levels of severity in dementia. This is beginning to hap-
8.2.3 COMMENT pen including: impact of grief (Shuter et al., 2013) and other
features of dementia (Bosboom and Almeida, 2014), refine-
The measurement of QoL in dementia poses unique chal- ment of psychometric properties (Cooke and Chaudhury,
lenges. Impaired judgement, reduced insight, cognitive 2013; Mjorud et al., 2014; Papastavrou et al., 2014), exami-
impairment, personality changes, affective and psychotic nation of the QoL towards the end-of-life in the palliative

Table 8.6 Quality of life scales in dementia


Scale Severity Respondent
Alzheimer’s Disease-Related Quality of Life (ADQRL) Mild to severe Caregivers
Rabins et al. (1999)
Bath Assessment of Subjective Quality of Life in Dementia Mild to moderate Patients
(BASQID)
Trigg et al. (2007)
Cornell-Brown Scale for Quality of Life in dementia (CBS-QoLD) Mild to moderate Clinician with carer and
Ready et al. (2002) patient input

Cognitively Impaired Life Quality Scale (CILQ) DeLetter et al. Severe Nursing caregivers
(1995)
Dementia Care Mapping (DCM) Kitwood and Bredin (1994) Moderate to severe Trained rater

Dementia Quality of Life (DQoL) instrument Mild to moderate Patients


Brod et al. (1999)
Developing a method to measurethe quality of life in family Mild to moderate Proxy and patient
carers of people with dementia (DEMQOL) (Smith et al., 2007)
The Pleasant Events Schedule ‒ AD (PES-AD) (Logsdon and Teri, Mild Proxy and patient
1997)
Progressive Deterioration Scale (PDS) (De Jong et al., 1989) Mild to severe Proxy

Psychological Well-being in Cognitively Impaired Persons Severe Proxy


(PWB-CIP) (Burgener and Twigg, 2002)
Quality of Life Assessment Schedule (QoLAS) (Selai et al., 2001) Mild to moderate Patient and proxy

Quality of Life in Late-Stage Dementia (QUALID) (Weiner et al., Severe Proxy


2000)
A dementia specific QoL questionnaire rated by professionals Mild to moderate Professional caregiver
that can be applied in residential care: QUALIDEM (Ettema
et al., 2007)
Source: Adapted from Black, B.S. and Rabins, P.V., Dementia, Arnold Hodder, London, 2005.
74 Dementia

care context (Crowthers et al., 2013; Kumar and Kuriakose, Brodaty, H., Draper, B., Saab, D. et al. (2001). Psychosis
2013; Davies et al., 2014) and development of models for and behaviour disturbance in Sydney nursing home
quality of life adjusted to years based on functional impair- residents: Prevalence and predictors. International
ment (Kasai and Megura, 2013), which could be used in Journal of Geriatric Psychiatry, 16: 504–512.
cost-utility analysis. Brodaty, H. and Luscobme, G. (1996). Studies on affective
symptoms and disorders: Depression in persons with
dementia. International Psychogeriatrics, 8: 609–622.
REFERENCES Brooks, D.N. and McKinlay, W. (1983). Personality and
behavioural change after severe blunt head injury: A
Aalten, P., van Valen, E., de Vugt, M.E. et al. (2006). relative’s view. Journal of Neurology, Neurosurgery
Awareness of behavioural problems in demen- and Psychiatry, 46 (4): 336–344.
tia patients: A prospective study. International Burgener, S. and Twigg, P. (2002). Relationships among
Psychogeriatrics, 18: 3–18. caregiver factors and quality of life in care recipients
Akoo, S. and Shah, A.K. (1998). Screening for depresison with irreversible dementia. Alzheimer Disease and
by the nursing staff in an acute psychogeriatric unit. Associated Disorders, 16: 88–102.
Australasian Journal on Ageing, 17: 81–84. Burke, W.J., Houston, M.J., Boust, S.J. and Roccaforte,
Alexopoulous, G.S., Abrams, R.C., Young, R.C. and W.H. (1989). Use of geriatric depresison scale in
Shamoian, C.A. (1988). Cornell scale for depression in dementia of Alzheimer’s type. Journal of the American
dementia. Biological Psychiatry, 23 (3): 271–284. Geriatric Society, 37: 856–860.
Ballard, C.G., O’Brien, J., Swann, A.G. et al. (2001). Burns, A., Folstein, S., Brandt, J. and Folstein, M. (1990a).
The natural history of psychosis and depression in Clinical assessment of irritability, aggression and apa-
dementia with Lewy bodies and Alzheimer’s disease: thy in Huntington and Alzheimer’s disease. Journal of
Persitance and new cases over one year follow-up. Nervous and Mental Disease, 178: 20–26.
Journal of Clinical Psychiatry, 62: 46–49. Burns, A., Jacoby, R. and Levy, R. (1990b). Psychiatric
Ballard, C.G., Saad, K., Patel, A. et al. (1995). The preva- phenomena in Alzheimer’s disease. IV: Disorders of
lence and phenomenology of psychotic symptoms in behaviour. British Journal of Psychiatry, 157: 86–94.
dementia sufferers. International Journal of Geriatric Burns, A., Jacoby, R. and Levy, R. (1990c). Psychiatric phe-
Psychiatry, 10: 477–485. nomena in Alzheimer’s disease III: Disorders of mood.
Balsis, S., Carpenter, B.D. and Storandt, M. (2005). British Journal of Psychiatry, 157: 81–86.
Personality changes precedes clinical diagnosis of Burns, A., Jacoby, R. and Levy, R. (1990d). Psychiatric phe-
dementia of Alzheimer’s type. Journal of Gerontology nomena in Alzheimer’s disease I: Disorders of thought
Series B: Psychological Sciences and Social Sciences, content. British Journal of Psychiatry, 157: 72–76.
60: P98–P101. Burns, A., Jacoby, R. and Levy, R. (1990e). Psychiatric phe-
Banerjee, S., Smith, S., Murray, J. et al. (2002). nomena in Alzheimer’s disease II: Disorders of percep-
DEMQOL: A new measure of health related qual- tion. British Journal of Psychiatry, 157: 76–81.
ity of life in dementia. Neurobiology of Ageing, Burns, A., Jacoby, R. and Levy, R. (1990f). Behavioural
23 (Suppl. 1): S154. abnormalities and psychiatric symptoms in
Black, B.S. and Rabins, P.V. (2005). Quality of life in Alzheimer’s disease: Preliminary findings. International
dementia: Conceptual and practical issues. In J. O’ Psychogeriatrics, 2: 25–36.
Brien, D. Ames and A. Burns (eds.), Dementia. London: Chatterjee, A., Strauss, M.E., Smyth, K.A. and Whitehouse,
Arnold Hodder, pp. 215–228. P.J. (1992). Personality change in Alzheimer’s disease.
Blessed, G. (1968). The association between quantitative Archives of Neurology, 49: 486–491.
measures of dementia and of senile change in grey Cohen-Mansfeild, J., Golander, H. and Heinik, J. (2013).
matter of elderly subjects. British Journal of Psychiatry, Delusions and hallucinations in persons with dementia:
114: 797–811. A comparison of formal and informal caregivers. Journal
Bosboom, P.R. and Almeida, O. (2014). Do changes in of Geriatric Psychiatry and Neurology, 26: 251–258.
specific cognitive functions predict changes in health- Cooke, H.A. and Chaudhury, H. (2013). An examination of
related quality of life in people with Alzheimer’s psychometric properties and efficacy of dementia care
disease. International Journal of Geriatric Psychiatry, mapping. Dementia, 12: 790–805.
29: 694–703. Crowthers, J., Wilson, K.C., Horton, S. and Llyod-Williams,
Bozzola, F.G., Gorelick, P.B. and Freels, S. (1992). M. (2013). Compassion in healthcare ‒ Lessons learnt
Personality changes in Alzheimer’s disease. Archives of from a qualitative study of the end of life care of
Neurology, 49: 297–300. people with dementia. Journal of Royal Society of
Brod, M., Stewart, A.L., Sands, L. and Walton, P. (1999). Medicine, 106 (12): 492–497.
Conceptualization and measurement of quality of life Cummings, J.L. (1985). Organic delusions:
in dementia: The dementia quality of life instrument Phenomenology, anatomical correlations and review.
(DQoL). The Gerontologist, 39: 25–35. British Journal of Psychiatry, 146: 184–197.
Measurement of behaviour disturbance, non-cognitive symptoms and quality of life 75

Cummings, J.L., Mega, M. and Gray, K. et al. (1994). The Finkel, S.I. and Burns, A. (2000). Introduction to behav-
neuropsychiatric inventory: Comprehensive assess- ioural and psychological symptoms of dementia
ment of psychopathology in dementia. Neurology, 44: (BPSD): A clinical and research update. International
2308–2314. Psychogeriatrics, 12 (Suppl. 1): 9–12.
Cummings, J.L., Miller, B., Hill, M.A. and Neshkes, R. Haider, I. and Shah, A.K. (2004). A pilot study of behav-
(1987). Neuropsychiatric aspects of multi-infarct ioural and psychological signs and symptoms of
dementia and dementia of Alzheimer’s type. Archives dementia in patients of Indian sub-continent origin
of Neurology, 44 (4): 389–393. admitted to a dementia day hospital in the United
Cummings, J.L., Petry, S., Dian, L., Shapira, J.et al. (1990). Kingdom. International Journal of Geriatric Psychiatry,
Organic personality disorder in dementia syndromes: 19 (12): 1195–1204.
An inventory approach. Journal of Neuropsychiatry, 2: Hallberg, I.P., Edberg, A., Normark, A. et al. (1993).
261–267. Daytime vocal activity in institutionalised severely
Davies, N., Maio, L., Rait, G. and Iiliffe, S. (2014). Quality demented patients identified as vocally disruptive by
end-of-life care for dementia: What family carers nurses. International Journal of Geriatric Psychiatry, 8:
have told us so far? A narrative synthesis. Palliative 155–164.
Medicine, 28: 919–930. Hallberg, I.P., Norberg, A. and Ericckson, A. (1990).
Debruyne, H., Van Buggenhout, M., Le Bastard, N. et al. Functional impairment and behaviour disturbances in
(2009). Is the geriatric depression scale a reliable vocally disruptive patients in psychogeriatric wards
screening tool for depressive illness in elderly patients compared with controls. International Journal of
with cognitive impairment. International Journal of Geriatric Psychiatry, 5: 53–61.
Geriatric Psychiatry, 24: 556–562. Hancock, P. and Larner, A.J. (2008). Cambridge behav-
Dechamps, A., Jutand, M.A., Onifade, C. et al. (2008). ioural inventory for the diagnosis of dementia.
Co-occurance of neuropsychiatric syndromes in Progress in Neurology and Psychiatry, 12: 23–25.
demented and psychotic institutionalised elderly. Hinton, L., Tomaszewski Farias, S. and Wegelin, J. (2008).
International Journal of Geriatric Psychiatry, 23: Neuropsychiatric symptoms are associated with
1182–1190. disability in cognitively impaired Latino elderly with
De Jong, R., Osterlund, O.W. and Roy, G.W. (1989). and without dementia: Results from the Sacramento
Measurement of quality of life changes in patients Area Latino study on aging. International Journal of
with Alzheimer’s disease. Clinical Therapeutics, 11: Geriatric Psychiatry, 23 (1): 102–108.
545–554. Hope, T., Keene, J., Fairburn, C. et al. (1997). Behaviour
DeLetter, M.C., Tully, C.L., Wilson, J.F. and Rich, E. (1995). changes in dementia II: Are there behavioural syn-
Nursing staff perceptions of quality of life of cogni- dromes? International Journal of Geriatric Psychiatry,
tively impaired elders: Instrumental development. 12: 1074–1078.
Journal of Applied Gerontology, 14: 426–443. Hsieh, C.J., Chang, C.C. and Lin, C.C. (2009).
Devanand, D.P., Brockington, C.D., Moody, B.J. et al. Neuropsychiatric profiles of patients with
(1992a). Behaviour syndromes in Alzhiemer’s Disease. Alzheimer’s disease and vascular dementia in Taiwan.
International Psychogeriatrics, 4 (Suppl. 2): 161–184. International Journal of Geriatric Psychiatry, 24 (6):
Devanand, D.P., Miller, L., Richards, M. et al. (1992b). The 570–577.
Columbia University scale for psychopathology in Ishii, N., Nishihara, Y. and Imamura, T. (1986). Why do fron-
Alzheimer’s disease. Archive of Neurology, 49: 371–376. tal lobe symptoms predominate in vascular dementia
Dian, L., Cummings, J.L., Petry, S. and Hill, M.A. (1990). with lacunes? Neurology, 36 (3): 340–345.
Personality alterations in multi-infarct dementia. Jacoub, P. and Jorm, A. (1996). Personality change in
Psychosomatics, 4: 415–419. dementia of Alzheimer’s type. International Journal of
Ellanchenny, N. and Shah, A.K. (2001). Evaluation of Geriatric Psychiatry, 11: 201–207.
three nurse-administered depression rating scales Jacoub, P., Jorm, A., Christensen, H. et al. (1994).
on acute and continuing care psychogeriatric wards. Personality change in normal and cognitively impaired
International Journal of Methods in Psychiatric elderly: Informant reports in a community sample.
Research, 10: 43–51. International Journal of Geriatric Psychiatry, 9:
Ettema, T.P., Droes, R., de Lange, J. et al. (2005). The 313–320.
concept of quality of life in dementia in the different Jorm, A.F. (2001). History of depression as a risk factor
stages of the disease. International Psychogeriatrics, for dementia: An updated review. The Australian and
17: 353–370. New Zealand Journal of Psychiatry, 35 (6): 776–781.
Ettema, T.P., Droes, R., de Lange, J. et al. (2007). Kasai, M. and Meguro K. (2013). Estimated quality-
QUALIDEM: Development and evaluation of a demen- adjusted life-year associated with the degree of
tia specific quality of life instrument ‒ Validation. activities of daily living in patients with Alzheimer’s
International Journal of Geriatric Psychiatry, 22: disease. Dementia Geriatric Cognitive Disorder Extra,
424–430. 3: 482–488.
76 Dementia

Katona, C.L.E. and Aldridge, C.R. (1985). The dexametha- MacKenzie, T.B., Robiner, W.N. and Knopman, D.S. (1989).
sone test and depressive signs in dementia. Journal of Differences between patient and family assesments of
Affective Disorders, 8 (1): 83–89. depression in Alzheimer’s disease. American Journal of
Kirkwood, S.C., Siemers, E., Viken, R. et al. (2002). Psychiatry, 146: 1174–1178.
Longitudinal personality changes among pres- Meins, W. (2000). Impact of personality on behav-
ymptomatic Huntington disease gene carriers. ioural and psychological symptoms of dementia.
Neuropsychiatry, Neuropsychology and Behavioral International Psychogeriatrics, 12 (Suppl. 1): 107–109.
Neurology, 15 (3): 192–197. Meins, W. and Dammast, J. (2000). Do personality
Kitwood, T. and Bredin, K. (1994). Evaluating Dementia Care: traits predict the occurrence of Alzheimer’s disease.
The DCM Method, Sixth Edition. Bradford: University of International Journal of Geriatric Psychiatry, 15: 120–124.
Bradford, Bradford Dementia Research Group. Meins, W., Frey, A. and Thiesemann, R. (1998). Premorbid
Koopmans, R.T., van der Molen, M., Raats, M. and Ettema, personality traits in Alzheimer’s disease: Do they
T.P. (2009). Neuropsychiatric symptoms and qual- predispose to noncognitive behavioural symptoms?
ity of life in patients in the final phase of dementia. International Psychogeriatrics, 10 (4): 369–378.
International Journal of Geriatric Psychiatry, 24: 25–32. Mendez, M.F., Chen, A.K., Shapira, J.S. et al. (2006).
Korcyzn, A.D. and Halperin, L. (2009). Depression and Acquired extroversion associated with bitempo-
dementia. Journal of the Neurological Sciences, 283: ral variant of frontotemporal dementia. Journal of
139–142. Neuropsychiatry and Clinical Neurosciences, 18 (1):
Korner, A., Lauritzen, L., Abelskove, K. et al. (2006). The 100–107.
geriatric depression scale and the Cornell scale for Miller, B.L., Darby, A., Benson, D.F. et al. (1997).
depression in dementia: A validation study. Nordic Aggressive socially disruptive and antisocial behaviour
Journal of Psychiatry, 60: 360–364. associated with fronto-temporal dementia. British
Kumar, C.S. and Kuriakose, J.R. (2013). End-of-life care Journal of Psychiatry, 170: 150–154.
issues in advanced dementia. Mental Health in Family Mjorud, M., Kirkevold, M., Rosvik, J. and Engedal, K.
Medicine, 10 (3): 129–132. (2014). Principle component analysis of the Norwegian
Lam, L.C.W., Leung, T., Lui, V.W.C. et al. (2006). version of the quality of life in late stage dementia
Association between cognitive function, behav- scale. Dementia and Cognitive Disorder, 37: 265–275.
ioural syndromes and two-year clinical outcome in Moyle, W., Murfield, J., Venturto, L. et al. (2014). Dementia
Chinese subjects with late-onset Alzheimer’s disease. and its influence on quality of life and what it means to
International Psychogeriatrics, 18: 517–526. be valued: Family members’ perceptions. Dementia,
Lautenschlager, N.T. and Forstl, H. (2007). Personality 13 (3): 412–425.
change in old age. Current Opinions in Psychiatry, 20: Nilsson, L.V. (1983). Personality changes in the aged.
62–66. A transectional and longitudinal study with Eysenk
Lawton, M.P. (1991). A multi-dimensional view of qual- Personality Inventory. Acta Psychiatrica Scandinavica,
ity of life in frail elders. In J.E. Birren, J. Lubben, J.C. 68 (3): 202–211.
Rowe and D.E. Deutschmann (eds.), The Concept and Nilsson, K., Palmsteirna, B. and Wistedt, B. (1988).
Measurement of Quality of Life in the Frail Elderly. San Aggressive behaviour in hospitalised psychogeriatric
Diego, CA: Academic Press, pp. 3–27. patients. Acta Psychiatrica Scandinavica, 78: 172–175.
Lawton, M.P. (1994). Quality of life in Alzheimer’s disease. O’Connor, D.W. (2000). Epidemiology of behavioural and
Alzheimer Disease and Associated Disorders, 5: 21–32. psychological symptoms of dementia. International
Lawton, M.P. (1997). Assessing quality of life in Alzheimer Psychogeriatrics, 12 (Suppl. 1): 41–45.
disease research. Alzheimer Disease and Associated Ott, B.R. and Fogel, B.S. (1992). Measurement of depres-
Disorders, 11 (Suppl. 6): 91–99. sion in dementia: Self vs clinician rating. International
Logsdon, R.G. and Teri, L. (1997). The pleasant events Journal of Geriatric Psychiatry, 7 (12): 899–904.
schedule ‒ AD: Psychometric properties and rela- Palmsteirna, B. and Wistedt, B. (1987). Staff observation
tionship to depression and cognition in Alzheimer’s aggression scale: Presentation and evaluation. Acta
disease patients. The Gerontologist, 37: 40–45. Psychiatrica Scandinavica, 76: 657–663.
Lukovits, T.G. and McDaniel, K.D. (1992). Behavioural Papastavrou, E., Andreou, P., Middleton, N. et al. (2014).
disturbance in severe Alzheimer’s disease: A com- Factors associated with quality of life among fam-
parison of family member and nursing staff report- ily members of patients with dementia in Cyprus.
ing. Journal of the American Geriatric Society, 40: International Psychogeriatrics, 26: 443–452.
891–895. Patel, V. and Hope, R.A. (1992a). A rating scale for
Lyketsos, C.G., Sheppard, J.M.E., Steinberg, M. et al. aggressive behaviour in the elderly ‒ The RAGE.
(2001). Neuropsychiatric disturbance in Alzheimer’s Psychological Medicine, 22: 211–221.
disease clusters into three groups: The Cache county Pearson, P.R. (1990). Impulsiveness and spiral maze perfor-
study. International Journal of Geriatric Psychiatry, 16: mance in elderly psychiatry patients. The Personality
1043–1053. and Individual Differences, 11: 1309–1310.
Measurement of behaviour disturbance, non-cognitive symptoms and quality of life 77

Pearson, P.R. (1992). The relationship of impulsiveness to Selai, C.E., Trimble, M.R., Rossor, M.N. and Harvey, R.J.
psychoticism and extraversion in elderly psychiatric (2001). Assessing quality of life in dementia: Preliminary
patients. Journal of Psychology, 126: 443–444. psychometric testing of the quality of life assessment
Petry, S., Cummings, J., Hill, M. and Shapira, J. (1988). schedule (QOLAS). Neuropsychological Rehabilitation:
Personality alterations in dementia of the Alzheimer’s An International Journal, 11(3-4): 219–243.
type. Archives of Neurology, 45 (11): 1187–1190. Selbaek, G., Engedal, K., Benth, J.S. and Bergh, S. (2014).
Petry, S., Cummings, J., Hill, M. and Shapira, J. (1989). The course of neuropsychiatric symptoms in nursing
Personality alterations in dementia of Alzheimer’s home patients with dementia over a 53-month follow-
type: A three-year follow-up study. Journal of Geriatric up period. International Psychogeriatrics, 26: 81–91.
Psychiatry and Neurology, 2: 203–207. Seltzer, B. and Shrewin, I. (1983). A comparison of clinical
Pinto, C. and Seethalakshmi, R. (2006). Behavioural and features in early onset and late onset primary degen-
psychological symptoms of dementia in an Indian erative dementia. Archives of Neurology, 40: 143–146.
population: Comparison between Alzheimer’s disease Senanarong, V., Poungvarin, N., Jamjumras, P. et al. (2005).
and vascular dementia. International Psychogeriatrics, Neuropsychiatric symptoms, functional impairment
18: 87–94. and executive ability in Thai patients with Alzheimer’s
Rabins, P. and Kasper, J.D. (1997). Measuring quality of disease. International Psychogeriatrics, 17: 81–90.
life in dementia: Conceptual and practical issues. Shah, A.K. (1995). Violence among psychogeriatric inpa-
Alzheimer Disease and Related Disorders, 11 (Suppl. 6): tients with dementia. International Journal of Geriatric
100–104. Psychiatry, 10: 887–891.
Rabins, P.V., Kasper, J.D., Kleinman, L. et al. (1999). Shah, A.K. (1999a). Aggressive behaviour in the elderly.
Concepts and methods in the development of the International Journal of Psychiatry in Clinical Practice,
ADQRL: An instrument for assessing health related 3: 85–103.
quality of life in persons with Alzheimer’s disease. Shah, A.K. (1999b). Some methodological issues in using
Journal of Mental Health and Aging, 5: 33–48. aggression rating scales in institutions. International
Rankin, K.P., Baldwin, E., Pace-Savitsky, C. et al. (2005). Psychogeriatrics, 4: 439–444.
Self-awareness and personality change in dementia. Shah, A.K. (2000). What are the necessary characteris-
Journal of Neurology, Neurosurgery and Psychiatry, 76: tics of a behavioural and psychological symptoms in
632–639. dementia rating scale? International Psychogeriatrics,
Ready, R.E., Ott, B.R., Grace, J. and Fernandez, I. (2002). 12 (Suppl. 1): 205–209.
The Cornell-Brown scale for quality of life in dementia. Shah, A.K. and Allen, H. (1999). Is improvement possible in
Alzheimer’s Disease and Associated Disorders, 16: the measurement of behaviour disturbance in demen-
109–115. tia? International Journal of Geriatric Psychiatry, 14:
Reifler, B., Larson, E., Teri, L. and Poulsen, L. (1986). 512–519.
Dementia of Alzheimer’s type and depres- Shah, A.K., Chui, E. and Ames, D. (1997). The relation-
sion. Journal of the American Geriatric Society, ship between two aggression scales in nursing homes.
34: 855–859. International Journal of Geriatric Psychiatry, 12: 628–631.
Rindlisbacher, P. and Hopkins, R.W. (1992). An investi- Shah, A.K., Foli, S. and Nnatu, I. (2005). Measurement
gation of the sundowning syndrome. International of behavioural disturbance, non-cognitive symptoms
Journal of Geriatric Psychiatry, 7 (1): 15–23. and quality of life. In J. O’Brien, D. Ames and A. Burns
Rubins, E.H. and Kinscherf, D.A. (1989). Psychopathology (eds.), Dementia. London: Arnold Hodder, pp. 72–80.
of very mild dementia of alzheimer’s type. American Shah, A.K. and Gray, T. (1997). Screening for depression
Journal of Psychiatry, 146: 1017–1021. on continuing care psychogeriatric wards. International
Rubins, E.H., Morris, J.C. and Berg, L. (1987a). The pro- Journal of Geriatric Psychiatry, 12: 125–127.
gression of personality change in senile dementia of Shah, A.K., Suh, G.K. and Elanchenny, N. (2004). A cross-
Alzheimer’s type. Journal of the American Geriatric national comparative study of behavioural and psycho-
Society, 35: 721–725. logical symptoms of dementia between UK and Korea.
Rubins, E.H., Morris, J.C., Storandt, M. and Berg, L. International Psychogeriatrics, 16: 219–236.
(1987b). Behavioural change in patients with mild Shah, A.K., Suh, G.K. and Elanchenny, N. (2005). A
senile dementia of Alzheimer’s type. Psychiatric comparative study of behavioural and psychological
Research, 21: 55–62. symptoms of dementia in patients with Alzheimer’s
Ryden, M. (1988). Aggressuve behaviour in persons with disease and vascular dementia referred to psycho-
dementia who live in the community. Alzheimer’s geriatric services in Korea and the UK. International
Disease and Associated Disorders, 2: 345–355. Psychogeriatrics, 17: 207–219.
Savva, G.M., Zaccai, J., Matthews, F.E. et al. (2009). Shuter, P., Beattie, E. and Edwards, H. (2013). An explor-
Prevalence correlates and course of behavioural and atory study of grief and health related quality of life for
psychological symptoms of dementia in the popula- caregivers of people with dementia. American Journal of
tion. British Journal of Psychiatry, 194: 212–219. Alzheimer’s Disease and Other Dementias, 29: 379–385.
78 Dementia

Siegler, I.C., Welsh, K.A., Fillenbaum, G.G. et al. (1991). initiative, and national Akzheimer’s coordinating cen-
Ratings of personality change in patients being evalu- tre cohorts. American Journal of Geriatric Society, 21:
ated for memory disorders. Alzheimers Disease and 607–622.
Associated Disorders, 5 (4): 240–250. Van der Linde, R.M., Dening, T., Mathhews, F.E. and
Smith, S., Lamping, D., Banerjee, S. et al. (2005). Brayne, C. (2014). Grouping of behavioural and
Measurement of health related quality of life for peo- psychological symptoms of dementia. International
ple with dementia: Development of a new instrument Journal of Geriatric Psychiatry, 29: 562–568.
(DEMQOL) and an evaluation of current methodology. Verhey, F.R.J. and Visser, P.J. (2000). Phenomenology of
Health Technology Assessment, 9: 1–93. depression in dementia. International Psychogeriatrics,
Smith, S.C., Lamping, D.L., Banerjee, S. et al. (2007). 12 (Suppl. 1): 129–134.
Development of a new measure of health-related qual- Weiner, M.F., Martin-Cook, K., Svetlik, D.A. et al. (2000).
ity of life for older people with dementia: DEMQOL. The quality of life in late-stage dementia (QUALID)
Psychological Medicine, 37 (5): 737–746. scale. Journal of the American Medical Directors
Starkstein, S.E., Ingram, L., Garau, M.L. et al. (2005). An Association, 1 (3): 114–116.
overlap between apathy and depression in dementia. Weiner, M.F., Svetlik, D. and Risser, R.C. (1997). What
Journal of Neurology, Neurosurgery and Psychiatry, 76: depressive symptoms are reported in Alzheimer’s
1070–1074. patients. International Journal of Geriatric Psychiatry,
Strauss, M.E., Lee, M.M. and Di Filippo, J.M. (1997). 12 (6): 648–652.
Premorbid personality and behavioural symptoms Welleford, E.A., Harkins, S.W. and Taylor, J.R.
in Alzheimer’s disease: Some cautions. Archives of (1995). Personality change in dementia of
Neurology, 54: 257–259. Alzheimer’s type: Relations to care giver personal-
Strauss, M.E. and Pasupathi, M. (1994). Primary caregivers ity and burden. Experimental Ageing Research,
descriptions of Alzheimer patients’ personality traits: 21: 295–314.
Temporal stability and sensitivity to change. Alzheimer Whall, A.L., Kim, H., Colling, K.B. et al. (2013).
Disease and Related Disorders, 8 (3): 166–176. Measurement of aggressive behaviours in dementia:
Sultzer, D.L., Levin, H.S., Mahler, M.E. et al. (1993). A Comparison of the physical aggression subscales of
comparison of psychiatric symptoms in vascular the Cohen-Mansfield Agitation inventory and the
dementia and Alzheimer’s disease. American Journal Ryden Aggression Scale. Research Gerontological
of Psychiatry, 150: 1806–1812. Nursing, 6 (3): 171–177.
Sunderland, T., Alterman, I.S., Yount, D. et al. (1988). A Whitehouse, P., Patterson, M.B., Strauss, M.E. et al. (1996).
new scale for the assessment of depressed mood in Hallucinations. International Psychogeriatrics, 8 (Suppl.
dementia patients. American Journal of Psychiatry, 30): 387–392.
145: 955–959. WHOQOL Group (1995). The World Health
Talassi, E., Cipriani, G., Bianchetti, A. and Trabucchi, M. Organization Quality of Life Assessment Position
(2007). Personality change in Alzhiemer’s disease. Paper from the WHO. Social Science and Medicine,
Ageing and Mental Health, 11: 526–531. 41: 1403–1409.
Trabucchi, M. and Bianchetti, A. (1996). Clinical per- Winger, J., Schirm, V. and Stewart, D. (1987). Aggressive
spectives: What should we be studying? Deluisons. behaviour in long term care. Journal of Psychosocial
International Psychogeriatrics, 8 (Suppl. 3): 383–385. Nursing and Mental Health, 25: 28–33.
Trigg, T., Skevington, S.M. and Jones, R.W. (2007). How Wragg, R.E. and Jeste, D. (1989). Overview of depres-
can we best assess the quality of life of people with sion and psychosis in Alzheimer’s disease. American
dementia? The bath assessment of subjective quality of Journal of Psychiatry, 146 (5): 577–587.
life in dementia (BASQID). Gerontologist, 47: 789–797. Youn, J.C., Lee, D.Y., Lee, J.H. et al. (2008). Development
Truzzi, A., Ulstein, I., Valente, L. et al. (2013). Patterns of a Korean version of the behaviour rating scale for
of neuropsychiatric sub-syndromes in Brazilian and dementia (BRSD-K). International Journal of Geriatric
Norwegian patients with dementia. International Psychiatry, 23: 677–684.
Psychogeriatrics, 25: 228–235. Zaudig, M. (1996). Assessing behavioural symptoms
Trzepacz, P.T., Saykin, A., Yu, P. et al. (2013). Subscale of Alzhemer’s type: Categorical and quantita-
validation of neuropsychiatric inventory questionnaire: tive approaches. International Psychogeriatrics,
Comparison of Alzheimer’s disease neuroimaging 8 (Suppl. 2): 183–200.
9
Cross-cultural issues in the assessment of
cognitive impairment

AJIT SHAH

Education can influence performance on cognitive tests


9.1 INTRODUCTION (Choi et al., 2014), including the mini-mental state examina-
tion (MMSE) (Folstein et al., 1975). The Chinese MMSE has
Cross-cultural studies comparing two or more ethnic developed different cut-off scores predicting dementia for
groups in one country and those comparing populations different educational levels of respondents with good speci-
across two or more countries will be discussed with partic- ficity and sensitivity (Katzman et al., 1988; Zhang et al.,
ular reference to methodology, measurement instruments, 1990; Sahadevan et al., 2000); this has also been suggested
aetiology, protective and risk factors. for other cultural groups (Murden et al., 1991; Gurland et al.,
1992) and for different cognitive measurement instruments
(Gagnon et al., 2013), but may reduce sensitivity (Gagnon
et al., 2013). Illiterate subjects may be unable to complete
9.2 INTERNATIONAL DEMOGRAPHIC tests which require reading, writing and drawing (Katzman
CHANGES AND THEIR et al., 1988; Chandra et al., 1994; Rajkumar and Kumar, 1996)
IMPLICATIONS and illiteracy may reduce access to information related to
orientation in time and general knowledge (Lindesay, 1998;
The size of the elderly population is increasing worldwide Khan and Tadros, 2013). Innumerate subjects may be unable
because of increase in life expectancy and fall in the birth to perform tests involving calculations (Chandra et al., 1994;
rates (Oeppen and Vaupel, 2002; Shah and MacKenzie, 2007) Livingston and Sembhi, 2003). Subjects with lower levels of
and there is an increase in 80-year-olds (Jeune and Skythe, education may have difficulties in understanding the nature
2001; Shah, 2007a). The prevalence of dementia doubles every of the test and may feel that the questions are irrelevant and
5.1 years after the age of 60 (Hofman et al., 1991). Thus, with of little practical value (Chandra et al., 1994; Bhatnagar and
the increase in the elderly population, the absolute number Frank, 1997; Khan and Tadros, 2013).
of dementia cases will also increase worldwide. Tests identifying the discrepancy between age and date
of birth (Bhatnagar and Frank, 1997; McCrakken et al.,
1997) may disadvantage those born in remote villages
with poor birth registration facilities and those who have
9.3 DIAGNOSTIC ISSUES altered age and date of birth to facilitate migration and
entry into various institutions (Rait et al., 1997). Culture-
Cognitive tests standardized in one cultural group may not specific questions (e.g. about monarchy or politicians) also
be appropriate for another because they are influenced by cul- disadvantage ethnic minority elders (Bhatnagar and Frank,
ture, education, language, literacy, numeracy, sensory impair- 1997; McCrakken et al., 1997), although these can be modi-
ment, unfamiliarity with test situations and anxiety (Chandra fied (e.g. date of independence of the country of origin)
et al., 1994; Lindesay et al., 1997; Lindesay, 1998; Stewart et al., (Chandra et al., 1994; Rait et al., 1997). Cultural concepts of
2003; Prince et al., 2003, 2004; Crane et al., 2006; Inzelberg orientation in time and place (Ganguli et al., 1995; Lindesay
et al., 2007; Jacob et al., 2007; Kalaria et al., 2008; Gagnon et al., 1997) and preferential use of western or traditional
et al., 2013; Khan and Tadros, 2013; Choi et al., 2014). calendars (Kua, 1992; Lindesay et al., 1997; Rait et al., 1997)

79
80 Dementia

can also influence performance. Orientation items work (CAMCOG) component of the Cambridge Examination
well within the dominant culture but less so in some ethnic for Mental Disorders of the Elderly (CAMDEX) interview
minority groups (Rait et al., 1997; Livingston and Sembhi, schedule (Roth et al., 1986) have all been evaluated among
2003); this was the case noted among Singapore Chinese elderly members of the African Caribbean group in the
population (Kua, 1992), but not among Liverpool Chinese United Kingdom (Richards and Brayne, 1996; Richards
(McCrakken et al., 1997). et al., 2000). In this population, normative data is available
for the orientation items of the MMSE, selected items of
the CERAD neurospychological test battery and the clock
drawing test (Stewart et al., 2001a). The 10-word learning
9.4 MEASUREMENT INSTRUMENTS test from the CERAD battery has been proved successful in
establishing a diagnosis of dementia study among popula-
Several instruments measuring cognitive impairment have tions in India, China, Southeast Asia, Latin America and
been developed by adapting existing instruments (mostly the Caribbean (Prince et al., 2003). The Alzheimer’s Disease
those developed in England) by either using a Delphi panel Questionnaire (Breitner and Folstein, 1984) which gathers
of experts from the culture of interest or more widespread information on history of cognitive impairment among
consultation within the culture of interest, translation and first degree relatives, has been translated into Chinese and
back-translation by separate groups of bilingual translators Spanish and has been administered by bilingual workers
and iterative field pretesting to ensure content, semantic, (Silverman et al., 1992). The Informant Questionnaire on
technical, criterion and conceptual equivalence with the Cognitive Decline in the Elderly (IQCODE) (Jorm et al.,
parent version of the instrument for each item (Chandra 1991) has been developed for use among illiterate Chinese
et al., 1994; Shah et al., 2005b). The aim is to produce a population (Fuh et al., 1995). A dementia screening instru-
culture-fair, education-free and analogous instrument. This ment, the Community Screening Interview for Dementia
should be followed by pilot testing to determine the distri- (CSI-D), a cognitive test for subjects and an informant
bution of the scores and the ability to discriminate between interview has been developed for use among Cree Indians
dementias with various levels of severity (Chandra et al., in Canada (Hendrie et al., 1993; Hall et al., 2000), English-
1994; Gagnon et al., 2013). Clear validation against a gold speaking Canadians (Hendrie et al., 1993; Hall et al., 2000)
standard diagnosis of dementia is also required, together and Yoruba Nigerians in Ibadan (Hendrie et al., 1995b;
with determination of various psychometric properties Hall et al., 2000), Jamaicans (Hall et al., 2000), African
(Livingston and Sembhi, 2003; Shah et al., 2005b). Ideally, Americans in Indianapolis (Hall et al., 2000), Koreans
the psychometric profile of the newly developed instrument (Liu et al., 2005) and in study populations in India, China,
should be similar to or better than the parent version. Southeast Asia, Latin America and the Caribbean (Prince
The MMSE has been developed in several languages et al., 2003), with good psychometric properties. The Chula
including Arabic (Inzelberg et al., 2007), Chinese (Serby Mental Test, developed for use in Thailand by selecting and
et al., 1987; Katzman et al., 1988; Salmon et al., 1989; Yu adapting items from several existing screening tests, has
et al., 1989; Xu et al., 2003), Korean (Park and Kwon, 1990; been found to reduce the influence of literacy on scores
Park et al., 1991), Finnish (Salmon et al., 1989), Italian (Rocca (Jitapunkul et al., 1996). A Spanish (for use among Mexican
et al., 1990), Yoruba (Hendrie, 1992), Spanish (Escobar et al., Americans) (Royall et al., 2003) and Brazilian Portuguese
1986; Anzola-Perez et al., 1996), Thai (Phanthumchinda version (Fuzikawa et al., 2003, 2007) of the clock drawing
et al., 1991), Hindi (Ganguli et al., 1995; Rait et al., 2000a), test have been evaluated; both these versions were reported
Punjabi (Rait et al., 2000a), Urdu (Rait et al., 2000a), Bengali to aim at being education-free instruments. A clock reading
(Kabir and Herlitz, 2000; Rait et al., 2000a), Malyalam test, developed in Germany, has been reported to be reli-
(Shaji et al., 1996), Gujarati (Lindesay et al., 1997; Rait et al., able and sensitive in detecting Alzheimer’s disease (AD)
2000a) and Sinhalese (De Silva and Gunatilake, 2002). and Lewy body dementia (Schmidtke and Olbrich, 2007). A
Comparisons between these different versions are problem- cognitive assessment instrument, the Kimberely Cognitive
atic because not all have been developed using rigorous pro- Assessment Tool, has been developed for indigenous
cedures and/or a satisfactory psychometric evaluation. Australians and has good psychometric properties, includ-
The abbreviated Mental Test Score (Quereshi and ing good sensitivity and specificity in detecting dementia
Hodkinson, 1974) has been developed in several south (LoGuidice et al., 2006).
Asian languages and have been put to use among African A unique approach involving use of three instruments,
Caribbean population in the United Kingdom (Rait et al., the Geriatric Mental State examination (GMS) (Copeland
1997, 2000a, 2000b). A Korean version of the Alzheimer’s et al., 1976), the Community Screening Interview for
Disease Assessment Scale (ADAS) (Rosen et al., 1984) has Dementia (CIS-D) and the 10-word list-learning task from
been developed (Youn et al., 2002). The MMSE, selected items the CERAD battery appropriately translated into native lan-
of the Consortium to Establish a Registry for Alzheimer’s guages and an algorithm derived from these instruments
Disease (CERAD), a neurospychological test battery (Morris have been shown to have high sensitivity and specificity in
et al., 1989) and The Cambridge Cognitive Examination the diagnosis of dementia in culturally diverse populations
Cross-cultural issues in the assessment of cognitive impairment 81

like India, Iran, China, Southeast Asia, Africa, Latin relatives of Jews and Italians compared to Chinese and
America and the Caribbean (Prince et al., 2003, 2008). Puerto Ricans (Silverman et al., 1992). This raises the issue of
discrepant environmental exposure, as the first two groups
had migrated to the United States significantly earlier.
In a Canadian study, the prevalence of all dementias and
9.5 THE DIFFERENTIAL PREVALENCE AD was 4.2% and 0.5%, respectively, in Cree Indians and
AND INCIDENCE OF DEMENTIA 4.2% and 3.5%, respectively, in English-speaking Canadians
SUBTYPES (Hall et al., 1993). An Israeli study of presenile AD, using
hospital discharge diagnosis as a proxy for incidence rates,
Published studies of the prevalence of dementia in UK eth- reported age- and sex-adjusted incidence rates to be higher
nic minority groups generally do not report on the relative in European and American Jews than those from Asia or
prevalence of individual subtypes of dementia (Bhatnagar Africa (Treves et al., 1986), raising the possibility of both
and Frank, 1997; Lindesay et al., 1997; McCrakken et al., environmental and genetic risk factors. Pollitt (1997), quot-
1997; Livingston et al., 2001). Anecdotally, it is believed that ing Zann (1994), reported that the prevalence of dementia in
vascular dementia (VaD) may be more common than AD Aborigines in Northern Queensland was 20%, with alcohol-
among elders in the Indian subcontinent (Bhatnagar and related dementia accounting for majority of the cases.
Frank, 1997) and African Caribbean population (Richards A recent review of developing countries reported that
et al., 2000; Livingston et al., 2001; Stewart et al., 2001a; the age-adjusted prevalence of dementia in 65-year-olds
Livingston and Sembhi, 2003); one study demonstrated this and above was consistently low in India and sub-­Saharan
in the UK African Caribbean population (Stevens et al., Africa, although it was high in some Asian and Latin
2004). In the United Kingdom some risk factors for VaD are American countries (Kalaria et al., 2008). Although, the
more prevalent among the ethnic groups than in the indig- rates are variable, the overall report shows low prevalence
enous population, such as diabetes in Asians (Mather and of dementia in Asian and African countries. Similar find-
Keen, 1985; Samantha et al., 1987) and hypertension as well ings were observed in the 10/66 Dementia Research Group
as cardiovascular diseases in Asians and African Caribbean study when the DSM-IV diagnosis was used, but the preva-
population (Balarajan, 1996; Ritch et al., 1996; Taylor et al., lence rates were more consistent across countries when the
2013). Native Americans with AD in the United States are 10/66 dementia algorithm and diagnosis was used (Llibre
reported to have a higher prevalence of cardiovascular risk Rodriguez et al., 2008). Almost universally, the prevalence
factors including diabetes, hypertension and heart disease of dementia increases with age and generally it is more
than white Americans (Weiner et al., 2003). common in women. A meta-analysis of the world literature
The prevalence of dementia in ethnic minority groups reported that the incidence of VaD and AD exponentially
in other countries is also variable. A study of Chinese resi- increases with age (Jorm and Jolley, 1998). Eastern coun-
dents in U.S. nursing homes reported a prevalence of 95% tries had a lower incidence of AD, but there was no differ-
dementia patients with the ratio of VaD to AD estimated ence in the incidence of VaD between European and eastern
as 4.4 to 1 (Serby et al., 1987), but there was no indigenous countries (Jorm and Jolley, 1998). The prevalence of AD is
group with which this study could be compared. Studies generally lower in Japan and China (Hasegawa et al., 1986;
in the United States have reported a higher prevalence of Shibayama et al., 1986; Li et al., 1989, 1991) and that of VaD
dementia in African Americans (Still et al., 1990; Heyman generally higher (Hasegawa et al., 1986; Shibayama et al.,
et al., 1991; Perkins et al., 1997) and Hispanics (Perkins 1986; Li et al., 1989), although the ratio of the prevalence of
et al., 1997) than in white Americans, although this was not AD to VaD is increasing in these countries (Fujishima and
found in a study conducted on severe dementia in African Kiyohara, 2002; Li et al., 2007) with the passage of time. The
Americans (Schoenberg et al., 1985). African Americans and 10/66 incidence studies, using similar methodology across
Latinos had significantly higher prevalence and incidence all studied countries and when using the less controversial
of dementia than non-Latino white Americans (Gurland DSM IV diagnosis of dementia, reported low incidence for
et al., 1995, 1999), but these ethnic differences disappeared dementia in several Latin American countries and China
after applying background controls for age and educational (Prince et al., 2012).
attainment. Also, there was no difference between the three The variable prevalence rates for dementia and its sub-
groups selected for studying individual subtypes of demen- types across different ethnic groups in a single country
tia. Another study reported a higher prevalence of VaD and across different countries may be due to methodologi-
coupled with higher prevalence of strokes and hypertension cal issues including: the sensitivity of screening instru-
among African Americans (Heyman et al., 1991; Perkins ments and case-finding methods (Osuntokun et al., 1992;
et al., 1997). Hall et al., 1993; Prince et al., 2003); diagnostic criteria
A U.S. study, using an informant risk questionnaire (Osuntokun et al., 1992; Hall et al., 1993; Prince et al., 2003;
(Alzheimer’s Disease Risk Questionnaire) administered to Llibre Rodriguez et al., 2008; Prince et al. 2012); sampling
non-demented elderly day-care centre attenders, reported frames and procedures (Osuntokun et al., 1992); screen-
a significantly increased risk of dementia in first degree ing personnel (Hall et al., 1993) and interrater reliability
82 Dementia

(Osuntokun et al., 1992); age and demographic character- mellitus and cerebrovascular disease (Meguro et al., 2001b).
istics of study populations (Osuntokun et al., 1992); educa- The authors have posited that the prevalence of dementia
tional attainment and literacy (Hall et al., 1993; Prince et al., was not affected by environmental factors because environ-
2003) and cultural factors (Shah et al., 2005b). mental effects may take considerable time to emerge, per-
haps showing symptoms in subsequent generations.
The Indo-U.S. cross-national study reported a prevalence
of 0.84% of all dementias among rural Indians near Delhi,
9.6 GENETIC AND ENVIRONMENTAL a significantly lower figure than a community sample in
INFLUENCES Pennsylvania (Chandra et al., 1998). An incidence study in
the same geographical area reported one of the lowest inci-
If the differences in the prevalence and incidence persist dences for AD in the world (Chandra et al., 2001). A study of
after overcoming the methodological issues, then cross- Singapore Malays and Chinese reported a higher prevalence
cultural studies have the potential to allow investigation of dementia among Malays than Chinese (Kua and Ko,
concerning the underlying genetic and environmental 1995); the difference was mainly accounted for by a higher
risks, protective factors and the interaction between them prevalence of multi-infarct dementia in Malay women
(Amaducci et al., 1991; Osuntokun et al., 1992; Hall et al., compared with Malay men and Chinese women. A preva-
1993). If the risk of dementia was solely due to genetic factors lence and incidence study comparing rates among Yoruba
then migrants would share the same incidence as in their Nigerians in Ibadan and African Americans in Indianapolis
country of origin (Graves et al., 1994). Thus, comparison reported lower prevalence and incidence rates for dementia
of the incidences faced by same ethnic groups in different and AD among the Nigerians (Hendrie et al., 1995a, 2001),
communities, at different stages of socio-economic devel- again suggesting the importance of environmental aetio-
opment and differing environments, while maintaining logical factors. The 10/66 cross-national study using similar
genetic homogeneity, may allow identification of environ- methodologies across different countries reported relatively
mental risk factors (Graves et al., 1994). To meet this end, low incidence of dementia in Latin American countries and
cross-cultural incidence and prevalence studies designed to China when using DSM-IV criteria (Prince et al., 2012).
overcome the methodological difficulties, using screening
and diagnostic instruments that are culture-fair, education- 9.6.2 GENETICS
free and analogous instruments with similar case-finding
methods in age- and sex-stratified samples have emerged An early pilot population-based prevalence study reported
(Prince et al., 2003; Llibre Rodriguez et al., 2008; Prince a strong association between Apo lipoprotein E (ApoE)
et al., 2012). ∈4 allele and AD in African Americans (Hendrie et al.,
1995b) suggesting that ApoE ∈4 is a risk factor independent
9.6.1 ENVIRONMENT of ethnicity. However, this was not observed in Nigerians
(Osuntokun et al., 1995; Gureje et al., 2006), Kenyans (Chen
Despite comparable overall rates of dementia in Japan et al., 2009) and Cubans of African ancestry (Teruel et al.,
and the United States, the prevalence of VaD is highest in 2011), despite a high prevalence of ∈4 allele in the commu-
Japan, intermediate in Hawaii and the lowest in mainland nity in these groups; in the Cuban population sample the
United States, with an opposite trend observed in the rates effect of the ApoE ∈4 allele was attenuated (Teruel et al.,
of AD (Graves et al., 1994). The prevalence of AD among 2011). Similarly, the prevalence of ApoE ∈4 allele is higher
Japanese Americans in Washington (Graves et al., 1996) in AD in those people form North America and Northern
and Honolulu (White et al., 1996) is closer to the num- Europe and lower in those from Asian, Southern European
bers found in white American population than Japanese and Mediterranean countries (Crean et al., 2011). This dis-
population in Japan, suggesting an environmental aetiol- crepancy may be explained by differences in the expres-
ogy. A prospective cohort study of Japanese Americans sion of ApoE alleles or of ApoE receptors, interaction with
reported that those who led a traditional Japanese lifestyle unidentified modifier genes or environmental factors inter-
had a slower decline in cognition over 2 years (Graves et al., acting with genes. Furthermore, there was lower deposition
1999), also suggesting the importance of environmental fac- of A4 β-amyloid found in post-mortem brains of Nigerian
tors. Additionally, the lower prevalence of cardiovascular subjects without dementia compared with a similar series
disease among Japanese Americans may also explain slower from Australia (Osuntokun et al., 1994). Additionally, ApoE
rates of cognitive decline (Graves et al., 1999). Furthermore, ∈4 alleles may have a differential effect on different types of
increasing physical activity was associated with lower rates memory in different ethnic groups (Barnes et al., 2013); the
of dementia in the same Japanese American cohort (Taafe presence of this allele predicted greater decline in seman-
et al., 2008). However, a study of first-generation Japanese tic and working memories in whites compared with blacks.
migrants from Miyagi prefecture to Brazil (Meguro et al., These observations may help to explain a lower prevalence
2001a) reported a similar prevalence of dementia, VaD and and incidence of AD in Nigeria.
AD to that in Miyagi prefecture in Japan (Ishii et al., 1999), A strong association between ApoE ∈4 homozygous
despite the migrants having a higher prevalence of diabetes status and AD has been observed among Caucasians
Cross-cultural issues in the assessment of cognitive impairment 83

(non-Hispanic white Americans), African Americans and despite a decline in ApoE ∈4 frequency in AD with increase in
Hispanics in population-based prevalence studies in the age, other genetic and environmental factors may increase the
United States (Maestre et al., 1995; Tang et al., 1996; Sahota risk of developing AD in African Americans and Hispanics
et al., 1997); despite these associations, the relative risk of in the absence of this allele (Tang et al., 1998). A population-
developing AD was found to be the highest in Caucasians based incidence study of Japanese American men reported
compared to the other two groups suggesting a gene–­ a significant association between ApoE ∈4 and AD (Havlik
environment interaction. However, when considering het- et al., 2000). Also, ApoE ∈4 was found to be a risk factor for
erozygous ApoE ∈4 status, this relation was either weaker AD in women but not men in Venezuela suggesting that gen-
(Maestre et al., 1995) or absent in African Americans der may also be an important factor (Molero et al., 2001).
(Tang et al., 1996; Sahota et al., 1997) in comparison with Chandra and Pandav (1998) have speculated that high
Caucasians and Hispanics. A similar weak association was serum cholesterol levels may interact with ApoE ∈4 alleles
found between ApoE ∈4 status and cognitive impairment to produce AD based on evidence of low serum cholesterol
in African Caribbean population in the United Kingdom levels in rural Indians and a low prevalence of AD (Chandra
(Stewart et al., 2001c) and Cubans of African ancestry. et al., 1998). Moreover, low serum levels of cholesterol was
The presence of depressive symptoms and ApoE ∈4 allele observed at least 15 years before the onset of dementia, par-
markedly increased the risk of dementia, including AD, in ticularly in AD and subsequently remained low in Japanese
Japanese American men (Irie et al., 2008). American men and was suggested to be associated with
A population-based incidence study, with controlling early stages in the development of dementia (Stewart et al.,
apparatus for education, family history of AD and other risk 2007). However, the Indian observations were not supported
factors like hypertension, demonstrated a strong association by the ApoE type in the same population-based Indo-U.S.
between combined homozygous and heterozygous ApoE ∈4 prevalence study, where the frequency of ApoE ∈4 was sig-
status and AD in white Americans, African Americans and nificantly lower in the Indian sample than in the U.S. sample
Hispanics in the United States (Tang et al., 1998); Japanese and the strength of association between ApoE ∈4 and AD
(Ohara et al., 2011), Tunisian (Rassas et al., 2012) and Indian was same in both samples (Ganguli et al., 2000). These find-
(Bharath et al., 2010) studies with regard to controls applied ings collectively may derive one explanation because one of
to similar variables, demonstrated a strong link between the lowest prevalence and incidence rates for AD in the world
ApoE ∈4 allele and AD. Furthermore, in the presence of is the Indian sample (Chandra et al., 1998, 2001). The authors
at least one ApoE ∈4 allele, the cumulative risk of develop- concluded that the different prevalence in the two samples
ing AD up to the age of 90 years was higher in the African cannot be explained by differential risk or modifier risk fac-
American and Hispanic groups compared with the white tors pertaining to ApoE polymorphism and they speculated
American group, although a Norwegian study reported that there may be additional risk or protective factors, sur-
that the effect of ApoE ∈4 on AD becomes weaker with vival effects or contributory environmental factors.
increase in age (Sando et al., 2008a). These observations col- In a convenience sample of Cherokee Indians in Texas
lectively suggest that the effect of ApoE ∈4 on AD is weaker (United States), the risk of developing AD declined with an
and ‘dose dependent’ in African Americans compared with increase in the genetic degree of Cherokee ancestry and this
white Americans and to a lesser extent Hispanics. Moreover, relationship was independent of ApoE ∈4 allele (Rosenberg
a Cuban study demonstrated high prevalence of ApoE ∈4 et al., 1996). However, the protective effect of Cherokee
alleles in those with a strong influence of African ancestry, ancestry declined with age. These findings suggest a com-
but the effect of this allele were attenuated (Teruel et al., 2011). plex interaction between genetic and environmental factors
It is possible that the presence of other environmental factors determining AD.
or modifier genes may reduce the ApoE ∈4–­associated risk An increased risk for VaD in the presence of APOE E4
of developing AD in African Americans (Sahota et al., 1997; allele has been reported in India and a meta-analysis of 29
Tang et al., 1998). These environmental factors or modifier studies conducted (Bharath et al., 2010; Yin et al., 2012), but
genes may be able to alter the risk of one dose of ApoE ∈4 it still requires careful research.
more easily than two doses (Maestre et al., 1995).
Another explanation for these findings is that there may
be differential linkage disequilibrium between unidentified
AD susceptibility gene and ApoE ∈4 alleles in different eth-
9.7 EDUCATION
nic groups (Maestre et al., 1995). A further explanation is the
differential survival of those with ApoE ∈4 alleles, although The role of educational attainment as a risk factor for
this observation is not supported by reports that ApoE ∈4 in dementia is controversial, but its examination across dif-
white subjects is unchanged with age (Maestre et al., 1995; ferent cultural groups may shed further light on the role of
Tang et al., 1996) and either unchanged (Tang et al., 1996) or environmental factors or gene–environment interaction.
increased (Maestre et al., 1995) with age in African Americans. Low levels of education were associated with very mild and
Also, the frequency of ApoE ∈4 in AD in whites and African mild dementia in Hong Kong (Lam et al., 2008). Illiteracy
Americans is reported to decline (Maestre et al., 1995) or was identified as a risk factor for incident cases of AD in a
remain unchanged (Tang et al., 1996) with age. However, South Korean sample and this link was more pronounced
84 Dementia

with increase in age (Lee et al., 2008). Lower levels of educa- ●● High incidence–low mortality society
tion were associated with AD even when ApoE ∈4 status was ●● Low incidence–low mortality society
controlled for in a Norwegian sample (Sando et al., 2008b).
Levels of education were strongly correlated with the preva- Within this model several factors contribute to the
lence of dementia in Latinos, African Americans and non- ­prevalence of dementia:
Latino white Americans in the United States (Gurland et al.,
1995, 1999), but the differences in prevalence disappeared if ●● The overall incidence (and thus, the prevalence) will be
age and education controlling apparatus was used, suggest- low in societies where life expectancy is short because
ing that education may be an important environmental risk fewer subjects will reach the age where there is a risk of
factor independent of ethnicity. In a community study of dementia. The selective survival of those not at risk of
African Caribbean population in London, hypertension and dementia may further influence such a trend. It is pos-
diabetes were specifically associated with cognitive impair- sible that early mortality rate, selects for genetic and/or
ment in those with low levels of education (Stewart et al., constitutional factors that protect against neurodegen-
2001c). In a South Korean study, a similar association was erative disorders is responsible.
found between cognitive impairment and systolic blood ●● Mortality and survival after the onset of dementia also
pressure, with patients diagnosed earlier suffering from dia- influences prevalence rates. In societies where survival
betes and random glucose levels in subjects with no formal after the onset of dementia is short, the prevalence may
education (Stewart et al., 2003). be low even if the incidence is not.
These findings may be explained by several hypotheses ●● The incidence itself is important because uneven dis-
which require rigorous testing: tribution of protective or risk factors for dementia will
vary the incidence.
●● Subjects with lower levels of education may have less ●● In socio-economically less developed societies, infec-
well-controlled hypertension or diabetes (Gurland et al., tious diseases associated with poor socio-economic
1995, 1999; Stewart et al., 2001c, 2003). factors will reduce life expectancy with fewer indi-
●● Cognitive batteries may not be sufficiently sensitive to viduals reaching the age of risk for dementia. Such
measure cognitive impairment in those with high levels a society has been described as low incidence–high
of education (Stewart et al., 2001c). mortality society. With improvement in basic ­medical
●● Those with high levels of education may have developed care, the average life expectancy will increase and
sufficient cognitive reserve to be less vulnerable to the reach the threshold age where there is risk for demen-
effects of hypertension and diabetes (Gurland et al., tia (Li et al., 2007; Lam et al., 2008). This will lead
1995, 1999; Stewart et al., 2001c, 2003). to a gradual transition from a low incidence–high
●● Cerebral damage due to hypertension or diabetes mortality society to high incidence–high mortality
may be severe in those with lower levels of education society. The observed high prevalence of demen-
(Stewart et al., 2003). tia in a socio-economically deprived area of Brazil
may represent this stage of epidemiological transi-
Paradoxically, in a study of subjects with familial AD, tion (Scazufca et al., 2008). Furthermore, in socio-­
high levels of education were associated with earlier onset of economically developed societies, the mortality
dementia (Mejia et al., 2003). This was explained by earlier associated with dementia may decrease because of
detection of symptoms in those with higher levels of educa- greater availability of medical care and ­technology.
tion due to greater intellectual and environmental demands This will lead to a gradual transition from high
(Mejia et al., 2003). ­i ncidence–high ­mortality society to a high incidence–
low mortality society. In socio-economically well-off
societies, efforts to improve the control of risk factors
for dementia (such as those for VD) may reduce the
9.8 LIFE EXPECTANCY AND THE
incidence of dementia leading to a gradual transition
EPIDIMIOLOGICAL TRANSITION
to low incidence–low mortality society. There is evi-
HYPOTHESIS dence that the ratio of AD to that of vascular disease
is increasing in some Asian countries due to reduction
A theoretical model of the epidemiological transition in in the incidence of strokes (Fujishima and Kiyohara,
dementia has been developed using incidence and preva- 2002; Li et al., 2007).
lence data from different countries (Suh and Shah, 2001; ●● The transition from a low incidence–high mortality
Shah and Zarate-Escudero, 2014). According to this model society to low incidence–low mortality society may
all societies sequentially move through four hierarchical unfold in several ways depending upon availability of
stages: medical services, advances in medical care and technol-
ogy, public health policies and efforts undertaken to
●● Low incidence–high mortality society control risk factors for dementia and promote protective
●● High incidence–high mortality society factors for dementia in a given society.
Cross-cultural issues in the assessment of cognitive impairment 85

A similar epidemiological transition model has been REFERENCES


developed for other mental disorders in old age (Shah,
2007b) and for the elderly suicide rates (Shah, 2010; Shah Amaducci, L., Baldereschi, M., Amato, M.P. et al. (1991).
and Zarate Escudero, 2014). The World Health Organization cross-national research
program on age-associated dementias. Aging
(Milano), 3 (1): 89–96.
Anzola-Perez, E., Bangdiwala, S.I., De Llano, G.B. et al.
9.9 FUTURE DIRECTIONS (1996). Towards community diagnosis of dementia:
Testing cognitive impairment in older persons in
Cross-cultural studies of dementia have focussed primar- Argentina, Chile and Cuba. International Journal of
ily on cognitive impairment. Behavioural and psychological Geriatric Psychiatry, 11: 429–438.
symptoms of dementia (BPSD) have not been well-studied Balarajan, R. (1996). Ethnicity and variation in mortal-
in cross-cultural studies (Shah and Mukherjee, 2000; Shah ity from cardiovascular disease. Health Trends,
et al., 2005b). Cross-cultural measures of BPSD that are 28: 45–51.
culture-fair and analogous to those used in western coun- Barnes, L.L., Arvanitakis, Z., Yu, L. et al. (2013).
tries are emerging (Shah et al., 2004, 2005a, 2005c). BPSD Apolipoprotein E and change in episodic memory in
require close examination in cross-sectional and longitudi- blacks and whites. Neuroepidemiology, 40: 211–219.
nal studies conducted to study the prevalence and incidence Bharath, S., Purushottam, M., Mukherjee, O. et al. (2010).
of dementia in cross-cultural settings; these studies should Apolipoprotein E polymorphism and dementia: A
be designed to examine genetic and environmental risk fac- hospital-based study from southern India. Dementia
tors and the interaction between them for both cognitive and Geriatric Cognitive Disorder, 30 (6): 455–460.
impairment and BPSD. Bhatnagar, K.S. and Frank, J. (1997). Psychiatric disorders
Very few cross-cultural studies have examined neuro- in the elderly from the Indian subcontinent living in
chemical, neuroanatomical and neurohistological changes Bradford. International Journal of Geriatric Psychiatry,
in dementia (Amaducci et al., 1991) and structural and 12: 907–912.
functional imaging have hardly been used in cross-cultural Breitner, J.C.S. and Folstein, M. (1984). Familial
studies. These techniques should be applied in rigorous Alzheimer’s dementia: A prevalent disorder with spe-
population-based cross-cultural studies of both cognitive cific clinical features. American Journal of Psychiatry,
impairment and BPSD, using cross-culturally validated 14: 63–80.
measures of cognitive impairment and BPSD. This approach Chandra, V. and Pandav, R. (1998). Gene-environment
would provide powerful means of exploring environmental interaction in Alzheimer’s disease: A poten-
and genetic factors and the interaction between them. The tial role for cholesterol. Neuroepidemiology,
clustering of less common dementias, such as Huntington’s 17 (5): 225–232.
disease, prion dementias and Parkinsonism–dementia Chandra, V., Ganguli, M., Pandav, R. et al. (1998).
complex of Guam in some cultural groups may also shed Prevalence of Alzheimer’s and other dementias in
light on genetic and environmental interactions (Graham rural India: The Indo-US study. Neurology, 51 (4):
et al., 1998). 1000–1008.
The epidemiological transition hypothesis for dementia Chandra, V., Ganguli, M., Ratcliff, G. et al. (1994). Studies
also requires further study. This can initially be achieved of the epidemiology of dementia: Comparison
using data from incidence and prevalence studies from between developed and developing countries. Ageing
individual countries and data on socio-economic sta- Clinical and Experimental Research, 6: 307–321.
tus (e.g. gross national domestic product), life expectancy Chandra, V., Pandav, R., Dodge, H.H. et al. (2001).
and proxy measures of the quality of medical services (e.g. Incidence of Alzheimer’s disease in a rural community
infant mortality rates) ‒ all available from the World Health in the Indo-US study. Neurology, 57 (6): 985–989.
Organization. One statistical model of the proposed epi- Chen, C.H., Mizuno, T., Elston, R. et al. (2009). A compara-
demiological transition hypothesis is that of an inverted tive study to screen and APOE genotypes in an ageing
U-shaped curve following a quadratic equation Y = A + East African population. Neurobiology of Ageing, 31
BX - CX 2 (where Y is the incidence of dementia, X is the (5): 732–740.
socio-economic status and A, B and C are constants). This Choi, H.J., Lee D.Y., Seo, E.H. et al. (2014). A norma-
approach could be combined with measurement of genetic tive study of the digit span in an educationally
and environmental factors, which, in turn, would enable diverse elderly population. Psychiatry Investigation,
careful examination of these factors and their interaction as 11: 39–43.
societies develop socio-economically. Copeland, J.R.M., Kelleher, M.J., Kellett, J.M. et al. (1976).
The development of these sophisticated international A semi-structured interview for the assessment of
research programmes will require considerable develop- diagnosis and mental state in the elderly: The Geriatric
ment of specialist services for these groups worldwide if Mental State Schedule: I. Development and reliability.
they are to be feasible (Shah, 2008, Shah et al., 2008). Psychological Medicine, 6: 439–449.
86 Dementia

Crane, P.K., Gibbons, L.E., Jolley, L. et al. (2006). Differential Graves, A.B., Larson, E.B., Edland, S.D. et al. (1996).
item functioning related to education and age in the Prevalence of dementia and its subtype in the
Italian version of the Mini-mental State Examination. Japanese American in King County, Washington State.
International Psychogeriatrics, 18 (3): 505–515. American Journal of Epidemiology, 144: 760–771.
Crean, S., Ward, A., Mercaldi, C.J. et al. (2011). Graves, A.B., Larson, E.B., White, L.R. et al. (1994).
Apolipoprotein E ∈4 prevalence in Alzheimer’s disease Opportunities and challenges in international col-
patients varies across global populations: A systematic laborative epidemiological research of dementia and
literature review and meta-analysis. Dementia and its subtypes: Studies between Japan and the US.
Geriatric Cognitive Disorders, 31: 20–30. International Psychogeriatrics, 6: 209–223.
De Silva, H.A. and Gunatilake, S.B. (2002). Mini-Mental Graves, A.B., Rajaram, L., Bowen, J.D. et al. (1999).
State Examination in Sinhalese: A sensitive test to Cognitive decline and Japanese culture in a cohort
screen for dementia in Sri Lanka. International Journal of older Japanese Americans in King Country, WA:
of Geriatric Psychiatry, 7: 134–139. The Kame Project. The Journals of Gerontology Series
Escobar, J.I., Burnham, A., Karno, M. et al. (1986). Use of B-Psychological Sciences and Social Sciences, 54 (3):
Mini-mental State Examination (MMSE) in a commu- S154–S161.
nity population of mixed ethnicity. Journal of Nervous Gureje, O., Ogunniyi, A., Baiyewu, O. et al. (2006). APOE
and Mental Diseases, 174: 607–614. epsilon 4 is not associated with Alzheimer’s disease in
Folstein, M.F., Folstein, S.E. and McHugh, P.R. (1975). elderly Nigerians. Annals of Neurology, 59: 182–185.
‘Mini-mental State’: A practical method for grading Gurland, B.J., Wilder, D.E., Cross, P. et al. (1995). Relative
the cognitive state of patients for the clinician. Journal rates of dementia by multiple definitions, over two
of Psychiatric Research, 12: 189–198. prevalence periods in three sociocultural groups.
Fuh, J.L., Teng, E.L., Lin, K.N. et al. (1995). The informant American Journal of Geriatric Psychiatry, 3: 6–20.
questionnaire on cognitive decline in the elderly Gurland, B.J., Wilder, D.E., Cross, P. et al. (1992).
(IQCODE) as a screening tool for dementia for a pre- Screening scales for dementia: Towards reconcilia-
dominantly illiterate Chinese population. Neurology, 45: tion of conflicting cross-cultural findings. International
92–96. Journal of Geriatric Psychiatry, 7: 105–113.
Fujishima, M. and Kiyohara, Y. (2002). Incidence and risk Gurland, B.J., Wilder, D.E., Lantigua, R. et al. (1999). Rates
factors of dementia in a defined elderly Japanese of dementia in three ethnoracial groups. International
population: The Hisayama study. Annals of the New Journal of Geriatric Psychiatry, 14 (6): 481–493.
York Academy of Science, 977: 1–8. Hall, K.S., Gao, S., Emsley, C.L. et al. (2000). Community
Fuzikawa, C., Lima-Costa, F.M., Uchoa, E. et al. (2003). screening interview for dementia (CIS-D): Performance
A population-based study of intra and inter-rater reli- in five disparate study sites. International Journal of
ability of the clock drawing test in Brazil: The Bambui Geriatric Psychiatry, 15: 521–531.
Health and Ageing Study. International Journal of Hall, K.S., Hendries, H.C., Brittain, H.M. et al. (1993). The
Geriatric Psychiatry, 18: 450–456. development of dementia screening interview in two
Fuzikawa, C., Lima-Costa, F.M., Uchoa, E. et al. (2007). distinct languages. International Journal of Methods in
Correlation and agreement between the Mini-Mental Psychiatric Research, 3: 1–28.
State Examination and the clock drawing test in Hasegawa, K., Homma, A. and Imai, Y. (1986). An epide-
older adults with low levels of schooling: The Bambui miological study of age-related dementia in the com-
Health and Ageing Study (BHAS). International munity. International Journal of Geriatric Psychiatry, 1:
Psychogeriatrics, 19: 657–668. 45–55.
Gagnon, G., Hansen, K.T., Woolmore-Goodwin, S. et al. Havlik, R.J., Izmirlian, G., Petrovitch, H. et al. (2000).
(2013). Correcting the MoCA for education: Effect on ApoE ∈4 predicts incident AD in Japanese American
sensitivity. Canadian Journal of Neurological Science, men: The Honolulu-Asia Aging Study. Neurology, 54:
40: 678–683. 1526–1529.
Ganguli, M., Chandra, V., Kamboh, I. et al. (2000). Hendrie, H. (1992). Indianapolis-Ibadan research project.
Apolipoprotein E polymorphism and Alzheimer’s dis- In: J.D. Curb and A.B. Graves (eds.), Multi-national
ease. Archives of Neurology, 57: 824–830. studies of dementia. The Gerontologist, 32 (Suppl. 2):
Ganguli, M., Ratcliff, G., Chandra, V. et al. (1995). A 219.
Hindi version of the MMSE: Development of a cogni- Hendrie, H.C., Hall, K.S., Hui, S. et al. (1995a).
tive screening instrument for a largely illiterate rural Apolipoprotein E genotypes and Alzheimer’s disease
population in India. International Journal of Geriatric in a community study of elderly African Americans.
Psychiatry, 10: 367–377. Annals of Neurology, 37 (1): 118–120.
Graham, C., Howard, R. and Ya, Y. (1998). Studies on Hendrie, H., Hall, K.S., Pillay, N. et al. (1993). Alzheimer’s
dementia: Dementia and ethnicity. International disease is rare in Cree. International Psychogeriatrics,
Psychogeriatrics, 10: 183–191. 5: 5–15.
Cross-cultural issues in the assessment of cognitive impairment 87

Hendrie, H.C., Ogunniyi, A., Hall, K.S. et al. (2001). Katzman, R., Zhang, M., Qu, Q.Y. et al. (1988). A Chinese
Incidence of dementia and Alzheimer’s disease in 2 version of the Mini-mental State Examination: Impact
communities: Yoruba residing in Ibadan, Nigeria, and of illiteracy in a Shanghai dementia survey. Journal of
African Americans residing in Indianapolis, Indiana. Clinical Epidemiology, 41: 971–978.
Journal of the American Geriatric Society, 285: 739–747. Khan, F. and Tadros, G. (2013). Complexity in cognitive
Hendrie, H.C., Osuntokun, B.O., Hall, K.S. et al. (1995b). assessment of elderly British minority ethnic groups:
Prevalence of Alzheimer’s disease and demen- Cultural perspective. Dementia, 13: 467–482.
tia in two communities: Nigerian Africans and Kua, E.H. (1992). A community study of mental disor-
African Americans. American Journal of Psychiatry, ders in elderly Singaporean Chinese using the GMS-
152: 1485–1492. AGECAT package. The Australian and New Zealand
Heyman, A., Fillenbaum, G. and Prosnitz, B. (1991). Journal of Psychiatry, 26 (3): 502–506.
Estimated prevalence of dementia among elderly Kua, E.H. and Ko, S.M. (1995). Prevalence of demen-
black and white community residents. Archives of tia among elderly Chinese and Malay residents of
Neurology, 48: 594–598. Singapore. International Psychogeriatrics, 7: 439–446.
Hofman, A., Rocca, W.A., Brayne, C. et al. (1991). The Lam, L.C.W., Tam, C.W.C., Lui, V.W.C. et al. (2008).
prevalence of dementia in Europe: A collaborative Prevalence of very mild and mild dementia in com-
study of 1980–1990 findings. International Journal of munity-dwelling older Chinese people in Hong Kong.
Epidemiology, 20: 736–748. International Psychogeriatrics, 20: 135–148.
Inzelberg, R., Schechtman, E., Abuful, A. et al. (2007). Lee, J.Y., Chang, S.M., Jang, H.K. et al. (2008). Illiteracy
Education effects on cognitive function in a healthy and the incidence of Alzheimer’s disease in the
aged Arab population. International Psychogeriatrics, Yonchon County survey, Korea. International
19: 593–606. Psychogeriatrics, 20: 976–985.
Irie, F., Masaki, K.H., Petrovitch, H. et al. (2008). Li, G., Shen, Y.C., Chen, C.H. et al. (1989). An epidemio-
Apolipoprotein E epsilon 4 allele genotype and the logical survey of age-related dementia in an urban
effect of depressive symptoms on the risk of dementia area of Beijing. Acta Psychiatrica Scandinavica, 79:
in men: The Honolulu-Asia Aging Study. Archives of 557–563.
General Psychiatry, 65: 906–912. Li, G., Shen, Y.C., Zhau, Y.W. et al. (1991). A three-year
Ishii, H., Meguro, K., Ishizaki, J. et al. (1999). Prevalence follow-up study of age-related dementia in an urban
of senile dementia in a rural community in Japan: The area of Beijing. Acta Psychiatrica Scandinavica, 83:
Tajiri project. Archives of Gerontology and Geriatrics, 99–104.
29: 249–265. Li, S., Yan, F., Chen, C. et al. (2007). Is the dementia rate
Jacob, K.S., Senthil Kumar, P., Gayathri, K. et al. (2007). increasing in Beijing? Prevalence and incidence of
The diagnosis of dementia in the community. dementia 10 years later in an urban elderly population.
International Psychogeriatrics, 19: 669–678. Acta Psychiatrica Scandinavica, 115: 73–79.
Jeune, B. and Skytthe, A. (2001). Centenarians in Denmark Lindesay, J. (1998). The diagnosis of mental illness in
in the past and the present. Population an English elderly people from ethnic minorities. Advances in
Selection, 13: 75–94. Psychiatric Treatment, 4: 219–226.
Jitapunkul, S., Lailert, C., Worakul, P. et al. (1996). Chula Lindesay, J., Jagger, C. and Mlynik-Szmid, M. (1997). The
Mental Test: A screening test for elderly people in less mini-mental state examination (MMSE) in an elderly
developed countries. International Journal of Geriatric immigrant Gujarati population in the United Kingdom.
Psychiatry, 11: 715–720. International Journal of Geriatric Psychiatry, 12:
Jorm, A.F. and Jolley, D. (1998). The incidence of demen- 1155–1167.
tia: A meta-analysis. Neurology, 51: 728–733. Liu, S.I., Prince, M., Chiu, M.J. et al. (2005). Validity and
Jorm, A.F., Scott, R. and Cullen, J.S. (1991). Performance reliability of a Taiwan Chinese version of the com-
of informant questionnaire on Cognitive Decline in the munity screening instrument for dementia. American
Elderly (IQCODE) as a screening test for dementia. Journal of Geriatric Psychiatry, 13 (7): 581–588.
Psychological Medicine, 21 (3): 785–790. Livingston, G. and Sembhi, S. (2003). Mental health of the
Kabir, Z.H. and Herlitz, A. (2000). The Bangla adapta- ageing immigrant population. Advances in Psychiatric
tion of mini-mental state examination (BMASE): An Treatment, 9: 31–37.
instrument to assess cognitive function in illiterate and Livingston, G., Leavey, G., Kitchen, G. et al. (2001). Mental
literate individuals. International Journal of Geriatric health of migrants: The Islington study. The British
Psychiatry, 15: 441–440. Journal of Psychiatry, 179: 361–366.
Kalaria, R.N., Maestre, G.S., Arizaga, R. et al. (2008). Llibre Rodriguez, J.J., Ferri, C.P., Acosta, D. et al. (2008).
Alzheimer’s disease and vascular dementia in devel- Prevalence of dementia in Latin America, India and
oping countries: Prevalence, management and risk China: A population-based cross-sectional survey.
factors. Lancet Neurology, 7: 812–826. Lancet, 372: 464–474.
88 Dementia

LoGuidice, D., Smith, K., Thomas, J. et al. (2006). Osuntokun, B.O., Sahota, A., Ogunniyi, A.O. et al. (1995).
Kimberely Indigenous Cognitive Assessment Tool Lack of association between apolipoprotein E ∈4 allele
(KICA): Development of a cognitive assessment and Alzhimer’s disease in elderly Nigerians. Annals of
tool for older indigenous Australians. International Neurology, 38: 463–465.
Psychogeriatrics, 18: 269–280. Park, J.H. and Kwon, Y.C. (1990). Modification of the
Maestre, G., Ottman, R., Stern, Y. et al. (1995). Mini-mental State Examination for use in the elderly
Apolipoprotein E and Alzheimer’s disease: Ethnic in a non-western society. Part I. Development of the
variation in genotypic risks. Annals of Neurology, 37: Korean version of the Mini-Mental State Examination.
254–259. International Journal of Geriatric Psychiatry, 5: 381–387.
Mather, H. and Keen, M. (1985). The Southall dia- Park, J.H., Park, Y.N. and Ko, H.J. (1991). Modification
betes survey: Prevalence of known diabetes in of the Mini-mental State Examination for use in the
Asians and Europeans. British Medical Journal, 291: elderly in a non-western society. Part II: Cut-off points
1081–1084. and their diagnostic validities. International Journal of
McCrakken, C.F.M., Boneham, M.A., Copeland, J.R.M. Geriatric Psychiatry, 6: 875–882.
et al. (1997). Prevalence of dementia and depression Perkins, P., Annegers, J.F., Doody, R.S. et al. (1997).
among elderly people in black and ethnic groups. The Incidence and prevalence of dementia in a m ­ ultiethnic
British Journal of Psychiatry, 171: 269–273. cohort of municipal retirees. Neurology, 49 (1): 44–50.
Meguro, K., Meguro, M., Caramelli, P. et al. (2001a). Phanthumchinda, K., Jitapunkul, S., Sitthi-Amorn, C.
Elderly Japanese emigrants to Brazil before World War et al. (1991). Prevalence of dementia in an urban slum
II: II. Prevalence of dementia. International Journal of population in Thailand: Validity of screening meth-
Geriatric Psychiatry, 16 (8): 775–779. ods. International Journal of Geriatric Psychiatry, 6:
Meguro, K., Meguro, M., Caramelli, P. et al. (2001b). 639–646.
Elderly Japanese emigrants to Brazil before World War Pollitt, P. (1997). The problem of dementia in Australian
II: I. Clinical profiles based on specific historical back- Aboriginal and Torres Strait islander communities: An
ground. International Journal of Geriatric Psychiatry, overview. International Journal of Geriatric Psychiatry,
16: 768–774. 12: 155–163.
Mejia, S., Giraldo, M., Pineda, D. et al. (2003). Nongenetic Prince, M., Acosta, D., Chiu, H. et al. (2003). Dementia
factors as modifiers of the age of onset of familial diagnosis in developing countries: A cross-cultural
Alzheimer’s disease. International Psychogeriatrics, 15: validation study. Lancet, 361: 909–917.
337–349. Prince, M., Acosta, D., Chiu, H. et al. (2004). Effects of
Molero, A.E., Pino-Ramirez, G. and Maestre, G.E. (2001). education and culture on the validity of the Geriatric
Modulation by age and gender of risk of Alzheimer’s Mental State and its AGECAT algorithm. The British
disease and vascular dementia associated with apoli- Journal of Psychiatry, 185: 429–436.
poprotein E4 allele in Latin Americans: Findings from Prince, M., Acosta, D., Ferris, C.P. et al. (2012). Dementia
the Maracaibo Ageing Study. Neuroscience Letters, incidence and mortality in middle income countries,
307: 5–8. and associations with indicators of cognitive reserve:
Morris, J., Heyman, A., Mohs, R. et al. (1989). The A 10/66 Dementia Research Group population-based
Consortium to Establish a Registry for Alzheimer’s cohort study. Lancet, 380: 50–58.
disease (CERAD). Part 1: Clinical and neuropsychologi- Prince, M.J., de Rodriguez, J.L., Noriega, L. et al.
cal assessment of Alzheimer’s disease. Neurology, 39: (2008). The 10/66 Dementia Research Group’s
149–155. fully operational dementia computerised algo-
Murden, R.A., McRae, T.D., Kaner, S. et al. (1991). Mini- rithm, comparison of 10/66 dementia algorithm
mental state exam scores vary with education in blacks a clinician diagnosis validation study. BMC Public
and whites. Journal of the American Geriatric Society, Health, 8 (1): 1. Quereshi, K.N. and Hodkinson, H.M.
39: 149–155. (1974). Evaluation of a ten-question mental test in
Oeppen, J. and Vaupel, J.W. (2002). Broken limits to life institutionalised elderly. Age and Ageing, 3: 152–157.
expectancy. Science, 296: 1029–1031. Rait, G., Burns, A., Baldwin, R. et al. (2000a). Validating
Ohara, T, Ninomiya, T., Kubo, M. et al. (2011). screening instruments for cognitive impairment
Apolipoprotein genotype for prediction of Alzheimer’s in older south Asians in the United Kingdom.
in older Japanese: The Hisayama study. Journal of the International Journal of Geriatric Psychiatry, 15: 54–62.
American Geriatric Society, 59: 1074–1079. Rait, G., Morley, M., Burns, A. et al. (2000b). Screening
Osuntokun, B.O., Hendrie, H.C. and Ogunniyi, A.O. for cognitive impairment in older African-Caribbeans.
(1992). Cross-cultural studies in Alzheimer’s diesease. Psychological Medicine, 30: 957–963.
Ethnicity and Diseases, 2: 352–357. Rait, G., Morley, M., Lambat, I. and Burns, A. (1997).
Osuntokun, B.O., Ogunniyi, A.O., Akang, E.E.U. et al. Modification of brief cognitive assessments for use
(1994). A4-amyloid in brains of non-demented with elderly people from the South Asian sub-conti-
Nigerians. Lancet, 343: 56. nent. Ageing and Mental Health, 1: 356–363.
Cross-cultural issues in the assessment of cognitive impairment 89

Rajkumar, S. and Kumar, S. (1996). Prevalence of d ­ ementia Sando, S.B., Melquist, S., Cannon, A. et al. (2008a). APOE
in the community: A rural urban comparison from epsilon 4 lowers the age of onset and is a high risk fac-
Madras, India. Australian Journal on Ageing, 15: 9–13. tor for Alzheimer’s disease: A case control study from
Rassas, A.A., Mrabet Khiari, H., Hadj Fredj, S. et al. (2012). central Norway. BMC Public Health, 16: 9.
High APOE epsilon 4 allele frequencies associated Sando, S.B., Melquist, S., Cannon, A. et al. (2008b). Risk-
with Alzheimer’s disease in a Tunisian population. reducing effect of education in Alzheimer’s disease.
Neurological Science, 33: 33–37. International Journal of Geriatric Psychiatry, 23:
Richards, M. and Brayne, C. (1996). Cross-cultural research 1156–1162.
into cognitive impairment and dementia: Some practi- Scazufca, M., Menezes, P.R., Vallada, H.P. et al. (2008).
cal experiences. International Journal of Geriatric High prevalence of dementia among older adults from
Psychiatry, 11: 383–387. poor socioeconomic backgrounds in Sao Paulo, Brazil.
Richards, M., Brayne, C., Dening, T. et al. (2000). Cognitive International Psychogeriatrics, 20: 394–405.
function in UK community dwelling African Caribbean Schmidtke, K. and Olbrich, S. (2007). The Clock Reading
and white elders: A pilot study. International Journal of Test: Validation of an instrument for the diagnosis of
Geriatric Psychiatry, 15: 621–630. dementia and disorders of visuo-spatial cognition.
Ritch, A.E.S., Ehtisham, M., Guthries, S. et al. (1996). International Psychogeriatrics, 19: 307–322.
Ethnic influence on health and dependency of elderly Schoenberg, B.S., Anderson, D.W. and Haerer, A.F. (1985).
innercity residents. Journal of the Royal College of Severe dementia: Prevalence and clinical features in
Physicians of London, 30: 215–220. a biracial US population. Archives of Neurology, 42:
Rocca, W.A., Bonaiuto, S., Lippi, A. et al. (1990). 740–743.
Prevalence of clinically diagnosed Alzheimer’s disease Serby, M., Chou, J.C. and Franssen, E.H. (1987). Dementia
and other dementing disorders. A door-to-door survey in an American Chinese nursing home population.
in Appigano, Macerata Province, Italy. Neurology, 40: American Journal of Psychiatry, 144: 811–881.
626–631. Shah, A.K. (2007a). Demographic changes among ethnic
Rosen, W.G., Mohs, R.C. and Davis, K.L. (1984). A new rat- minority elders in England and Wales. Implications
ing scale for Alzheimer’s disease. American Journal of for development and delivery of old age psychiatry
Psychiatry, 141: 1356–1364. services. International Journal of Migration, Health and
Rosenberg, R.N., Richter, R.W., Risser, R.C. et al. (1996). Social Care, 3: 22–32.
Genetic factors for the development of Alzheimer’s Shah, A.K. (2007b). The importance of socio-economic
disease in Cherokee Indians. Archives of Neurology, status of countries for mental disorders in old age: A
53: 997–1000. development of an epidemiological transition model.
Roth, M., Tym, E., Mountjoy, C.Q. et al. (1986). CAMDEX: International Psychogeriatrics, 19: 785–787.
A standardised instrument for diagnosis of mental Shah, A.K. (2008). Do socio-economic factors, elderly
disorder in the elderly with special references to population size and service development factors influ-
early detection of dementia. The British Journal of ence the development of specialist mental health pro-
Psychiatry, 149: 698–709. grams for older people? International Psychogeriatrics,
Royall, D.R., Espino, D.V., Polk, M.J. et al. (2003). Validation of 20 (6): 1238–1244.
a Spanish translation of the CLOX for use in Hispanic Shah, A.K. (2010). The possible evidence for epide-
samples: The Hispanic EPESE study. International Journal miological transition hypothesis for elderly suicides.
of Geriatric Psychiatry, 18: 135–141. International Psychogeriatrics, 22: 219–226.
Sahadevan, S., Lim, P.P.J., Tan, N.J.L. and Chan, S.P. Shah, A.K. and MacKenzie, S. (2007). Disorders of
(2000). Diagnostic performance of the mental status ageing across cultures. In D. Bhugra and K. Bhui
tests in older Chinese: Influence of education and age (eds.), Textbook of Cultural Psychiatry. Cambridge:
on cut-off values. International Journal of Geriatric Cambridge University Press, pp. 323–344.
Psychiatry, 15: 234–241. Shah, A.K. and Mukherjee, S. (2000). Cross-cultural issues
Sahota, A., Yang, M. Gao, S. et al. (1997). Apolipoprotein in the measurement of behavioural and psychological
E-associated risk of Alzheimer’s disease in the African signs and symptoms of dementia (BPSD). Ageing and
American population is genotype dependent. Annals Mental Health, 4: 244–252.
of Neurology, 42: 659–661. Shah, A.K. and Zarate-Escudero, S. (2014). Can some
Salmon, D.P., Reikkinen, P.J., Katzman, R. et al. (1989). aspects of the epidemiology of suicide be applied
Cross-cultural studies of dementia ‒ A comparison of to dementia? International Psychogeriatrics, 26 (2):
the Mini-Mental state Examination performance in 185–191.
Finland and China. Archives of Neurology, 46: 769–772. Shah, A.K., Dalvi, M. and Thompson, T. (2005a).
Samantha, D.P., Burden, A.C. and Fent, B. (1987). Behavioural and psychological signs and symptoms
Comparative prevalence of non-insulin-dependent of dementia across cultures: Current status and the
diabetes mellitus in Asian and white Caucasian adults. future. International Journal of Geriatric Psychiatry, 20:
Diabetes Research and Clinical Practice, 4: 1–6. 1187–1195.
90 Dementia

Shah, A.K., Doe, P. and Deverill, K. (2008). Ethnic minor- Still, C.N., Jackson, K.L., Brandes, D.A. et al. (1990).
ity elders: Are they neglected in published geriatric Distribution of major dementias by race and sex in
psychiatry literature. International Psychogeriatrics, 20: South Carolina. Journal of South Carolina Medical
1041–1045. Association, 86: 453–456.
Shah, A.K., Lindesay, J. and Nnatu I. (2005b). Cross- Suh, G.K. and Shah, A.K. (2001). A review of epidemiologi-
cultural issues in the assessment of cognitive cal transition in dementia: Cross-national comparisons
impairment. In J. O’Brien, D. Ames and A. Burns of the indices related to Alzheimer’s disease and
(eds.), Dementia. London: Arnold Hodder, pp. vascular dementia. Acta Psychiatrica Scandinavica, 104:
147–164. 4–11.
Shah, A.K., Suh, G.K. and Elanchenny, N. (2005c). A Taafe, D.R., Irie, F., Masaki, K.H. et al. (2008). Physical
comparative study of behavioural and psychological activity, physical function, and incident dementia in
symptoms of dementia in patients with Alzheimer’s elderly men: The Honolulu-Asia Aging Study. Journal
disease and vascular dementia referred to psycho- of Gerontology Series A Biological Sciences and
geriatric services in Korea and the UK. International Medical Sciences, 63: 529–535.
Psychogeriatrics, 17: 207–219. Tang, M.X., Maestre, G., Tsai, W.Y. et al. (1996). Relative
Shah, A.K., Suh, G.K. and Elanchenny, N. (2004). A cross- risk of Alzheimer’s disease and age-at-onset distribu-
national comparative study of behavioural and psycho- tions based on APOE genotypes among elderly African
logical symptoms of dementia between UK and Korea. Americans, Caucasians, and Hispanics in New York City.
International Psychogeriatrics, 16: 219–236. American Journal of Human Genetics, 58 (3): 574–584.
Shaji, S., Promodu, K., Abraham, T. et al. (1996). An epi- Tang, M.X., Stern, Y., Marder, K. et al. (1998). The ApoE
demiological study of dementia in a rural community ∈4 allele and the risk of Alzheimer’s disease among
in Kerala, India. The British Journal of Psychiatry, 168: African Americans, whites and Hispanics. Journal of
747–780. the American Geriatric Society, 279: 751–755.
Shibayama, K., Kashara, Y., Kobyashi, H. et al. (1986). Taylor, C., Tillin, T., Chatuvedi, N., Dewey, M. et al. (2103).
Prevalence of dementias in a Japanese elderly popula- Midlife hypertension status and cognitive function
tion. Acta Psychiatrica Scandinavica, 74: 144–151. 20 years later: The Southall and Brent revisited study.
Silverman, J.M., Li, G., Schear, S. et al. (1992). A cross- Journal of American Geriatric Society, 61: 1489–1498.
cultural family history study of primary progres- Teruel, B.M., Rodriguez, J.J., McKeigue, P. et al. (2011).
sive dementia in relatives of non-demented elderly Interactions between genetic admixture, ethnic iden-
Chinese, Italians, Jews and Peurto Ricans. Acta tity, APOE genotype and dementia prevalence in an
Psychiatrica Scandinavica, 85: 211–217. admixed Cuban sample: A cross-sectional population
Stevens, T., Leavey, G. and Livingston, G. (2004). survey and nested case-control study. BMC Medical
Dementia and hypertension in African/Caribbean Genetics, 12: 43.
elders. Age Ageing, 33: 193–195. Treves, T., Korczyn, A.D., Zilber, N. et al. (1986). Presenile
Stewart, R., Kim, J., Shin, I. et al. (2003). Education and the dementia in Israel. Archives of Neurology, 43: 26–29.
association between vascular risk factors and cognitive Weiner, M.F., Rosenberg R.N., Svetlik, D. et al. (2003).
function: A cross-sectional study in older Koreans with Comparison of Alzheimer’s disease in native and
cognitive impairment. International Psychogeriatrics, white Americans. International Psychogeriatrics,
15: 27–36. 15: 367–375.
Stewart, R., Richards, M., Brayne, C. et al. (2001a). White, L., Petrovitch, H., Ross, G.W. et al. (1996).
Cognitive function in UK community-dwelling/African Prevalence of dementia in older Japanese-American
Caribbean elders: Normative data for a test bat- men in Hawaii. Journal of the American Medical
tery. International Journal of Geriatric Psychiatry, 16: Association, 276: 955–960.
518–527. Xu, G., Meyer, J.S., Huang, Y. et al. (2003). Adapting
Stewart, R., Richards, M., Brayne, C. and, Mann, A. mini-mental state examination for dementia screen-
(2001b). Vascular risk and cognitive impairment in an ing among illiterate or minimally educated Chinese.
older British, African-Caribbean population. Journal of International Journal of Geriatric Psychiatry, 18:
the American Geriatric Society, 49: 263–269. 609–616.
Stewart, R., Russ, C., Richards, M. et al. (2001c). Yin, Y.W., Li, J.C., Wang, J.Z. et al. (2012). Association
Apolipoprotein E genotype, vascular risk and early between apolipoprotein E gene polymorphism
cognitive impairment in an African Caribbean popula- and the risk of vascular dementia: A meta-analysis.
tion. Dementia and Geriatric Cognitive Disorders, 12: Neuroscience Letter, 514: 6–11.
251–256. Youn, J.C., Lee, D.Y., Kim, K.W. et al. (2002). Development
Stewart, R., White, L.R., Xue, Q.L. et al. (2007). Twenty- of the Korean version of Alzheimer’s Disease
six-year change in total cholesterol levels and incident Assessment Scale (ADAS-K). International Journal of
dementia. Archives of Neurology, 64: 103–107. Geriatric Psychiatry, 17: 797–803.
Cross-cultural issues in the assessment of cognitive impairment 91

Yu, E.S.H., Liu, W.T., Levy, P. et al. (1989). Cognitive impair- Zhang, M., Katzman, R., Salmon, D. et al. (1990). The
ment among elderly adults in Shanghai, China. Journal prevalence of dementia and Alzheimer’s disease in
of Gerontology, 44: S97–S106. Shanghai, China: Impact of age, gender and educa-
Zann, S. (1994). Identification of Support, Education and tion. Annals of Neurology, 27: 428–437.
Training Needs of Rural/Remote Health Care Service
Providers Involved in Dementia Care: Rural Health,
Support, Education and Training (RHSET). Project
Progress Report. Townsville, Queensland: Northern
Regional Health Authority.
10
Structural brain imaging

ROBERT BARBER and JOHN T. O’BRIEN

develop dementia. It has also been used in clinical trials of


10.1 INTRODUCTION putative disease modifying agents to aid sample selection
(stratification/enrichment) and as a surrogate measure to
This chapter focuses on the use of structural imaging, ­monitor patient safety and adverse events.
­magnetic resonance imaging (MRI) and computed tomog- There are a number of important longitudinal studies
raphy (CT) in dementia. in progress trying to improve the clinical utility of neuro-
From a clinical-orientated perspective, the ability to imaging and evaluate how best to combine different imag-
visualize the structure of the live brain directly means ing modalities as well as their relationship with bio-fluid
MRI and CT scans play a central role in diagnosis, pro- markers and clinical phenotypes. At present, however,
viding both positive and negative predictive information. there is a lack of standardization in how imaging biomark-
Traditionally, imaging in dementia was applied to ‘rule ers are acquired and measured to draw definitive conclu-
out’ pathologies that may be responsible for cognitive sions regarding combination of biomarkers which offer the
impairment, including the so-called treatable or reversible best diagnostic and prognostic accuracy (Jack et al., 2011a;
causes. Increasingly, the application of imaging recognizes Frisoni et al., 2013). An added obstacle, especially in demen-
dementia subtypes have characteristic changes that can be tia, stems from the relative lack of pathological validation of
identified at various stages of the illness. This shift to use in vivo imaging findings – either because of the difficulty
imaging to ‘rule in’ these changes can increase diagnostic in obtaining a donor for brain and/or the gap in time (often
accuracy and confidence, and is increasingly becoming a years) between scan acquisition and brain tissue autopsy
part of the new clinical diagnostic criteria for dementia analysis, if the scan is available.
subtypes. Reflecting this change, structural imaging is now
regarded as a core investigation in the clinical assessment
of patients with possible dementia: a practice endorsed
by international consensus guidelines for the assessment
10.2 BACKGROUND TO CT AND
of those suspected of suffering from dementia (see Table MRI AND THEIR CLINICAL
10.1). Indeed, expectations are growing and imaging can APPLICATION
be central to helping individuals and their families under-
stand and adjust to the diagnosis of dementia. To optimize 10.2.1 COMPUTED TOMOGRAPHY
the benefits of imaging, clinicians should know how best
to incorporate these techniques into their practice, includ- This chapter focuses on X-ray-based CT; other CT modali-
ing how to interpret their findings in the wider context of ties, notably, single-photon emission computer tomography
global clinical presentation and communicate their rel- (SPECT) and positron emission tomography (PET), are cov-
evance to patients and carers. ered in Chapter 11.
From a research-oriented perspective, imaging provides The first human X-rays from the 1890s were crude ‘sha­
a window to help understand the neurobiology of demen- dow’ images from a hand placed on a photographic plate.
tia in life. To date, studies have focused on identifying bio- Fast-forward to the 1970s and advances in computer tech-
markers that aid the differential diagnosis of dementia and nology allowed the first patient brain scan to be acquired by
the early identification of patients who are on a trajectory to combining single images or ‘slices’ (‘tomography’ is derived

92
Structural brain imaging 93

Table 10.1 Examples of guidelines for use of imaging in dementia

U.S./Canadian guidelines UK/European guidelines


Canadian Consensus Conference on Dementia (Patterson EFNS: Guidelines for the diagnosis and Management of
et al., 2001) Alzheimer’s disease (AD) (Hort et al., 2010)
Cranial computed tomography (CT) scan is recommended if Recommends CT or magnetic resonance imaging (MRI)
one or more of the following criteria is present. performed once for every patient. For AD, hippocampal
atrophy is best seen with the help of MRI scan but may
A. Age less than 60 years
also be visualized on the more modern type CT scanner
B. Rapid, (e.g. over 1–2 months) unexplained decline in
and yields sensitivity and specificity values between 80%
cognition or function
and 90% in most studies. For practice purposes, a
C. Short duration of dementia (less than 2 years
standard magnetic resonance protocol involves at least
D. Recent and significant head trauma
coronal T1 and axial T2 or fluid-attenuated inversion
E. Unexplained neurological symptoms, e.g. new onset of
recovery (FLAIR) sequences.
severe headache or seizures
The UK National Institute of Health and Care Excellence
F. History of cancer (especially in sites and types that
(NICE) Guidance on Dementia (guidance.nice.org.uk/
metastasize to the brain)
cg42, 2006)
G. Use of anticoagulants or history of a bleeding disorder
Structural imaging should be used in the assessment of
H. History of urinary incontinence and gait disorder in the
people with suspected dementia to exclude other
early stages of dementia (as may be found in
cerebral pathologies and to help establish the subtype
normopressure hydrocephalus)
diagnosis. MRI is the preferred modality to assist early
I. Any new localizing sign (e.g. hemiparesis or Babinski
diagnosis and detect subcortical vascular changes,
reflex)
although CT scanning could be used. Imaging may not
J. Unusual or atypical cognitive symptoms or presentation
always be needed in those presenting moderate to
(e.g. progressive aphasia, gait disturbance)
severe dementia, if the diagnosis is already clear.
Practice Parameters: Diagnosis of dementia (evidence- Specialist advice should be taken when interpreting scans
based guidelines) from the American Academy of in people with learning disabilities.
Neurology (Knopman et al., 2001)
Recommends structural neuroimaging with either a non-
contrast CT or MRI scan in the routine initial evaluation of
patients with dementia, usually is appropriate.

from the Greek word tomos meaning ‘slice’). On-going with small vessel disease in comparison to newer magnetic
advances have continued to improve patient experience resonance (MR) sequences. Clinically, CT is used more
and safety, as well as technical features such as spatial and widely than MRI, probably reflecting its greater availability,
temporal resolution and processing times allow the brain to ease of use and lower cost, though the evidence base relating
be imaged in seconds. The development of multi-slice CT to the utility of structural imaging in dementia is predomi-
has made three-dimensional (3D) reconstruction possible, nantly from MRI studies.
so images are no longer limited to the axial plane. As with
the first X-ray, the CT image ultimately depends on electron
density and the differential absorption of X-rays by body 10.2.2 MAGNETIC RESONANCE IMAGING
tissue, in such a way that high-density structures like bone
appears white and low-density tissue, whereas, water and fat This chapter focuses on structural MRI; other MRI modali-
appear black. ties, notably, functional MRI, are covered in Chapter 20.
CT and MRI have various advantages and disadvan- MRI is a non-invasive chemical probe that utilizes the
tages as summarized in Table 10.2. As a general rule, CT electromagnetic properties of protons to generate high-
is adequate for ‘ruling out’ most intra-cerebral lesions such resolution 3D images of the body’s internal structures. MRI
as tumours, metastases, strokes, subdural haemtomas, scanners use powerful magnetic fields, up to 60,000 times
hydrocephalus and bone trauma, but has a limited role to greater than the earth’s magnetic field and under the con-
play in comparison to MRI for ‘ruling in’ degenerative dis- trolled environment of the MR scanner, protons inside body
orders such as Alzheimer’s disease (AD), though CT coro- tissues are induced to act like microscopic radio transmit-
nal reconstruction now allows reasonable assessment of the ters. 3D images are then constructed by detecting and pro-
degree of temporal atrophy. CT is a lesser sensitive marker cessing these signals. About two-thirds of the human body is
of vascular disease, especially when mild and/or associated water, composed of hydrogen nuclei or protons. The ability
94 Dementia

of MRI to distinguish different structures and pathologi- normal and abnormal tissue. A major strength of MRI is its
cal changes is based upon the variation in the water con- versatility to visualize changes in both grey and white mat-
tent (protons) and alterations in the cellular environment ter by changing the type of imaging sequences. The princi-
that affects the behaviour of protons: a difference in water pal sequences and their application to study brain diseases
content of less than one per cent is enough to distinguish are summarized in Table 10.3.
Clinically, MRI provides a method to study atrophy and
cerebrovascular disease (CVD), as well as various causes of
Table 10.2 Comparison between computed tomography
grey and white matter abnormalities. These include demy-
(CT) and magnetic resonance imaging (MRI)
elinating, infective, inflammatory, toxic or metabolic condi-
CT MRI tions. There are a number of important contraindications to
Shorter scan times (<10 Higher spatial and MRI, as summarized in Table 10.4.
seconds, good for restless anatomical resolution
patients)
Widely available Superior soft tissue 10.3 ASSESSMENT OF BRAIN ATROPHY
definition and contrast AND VASCULAR DISEASE
Lower cost Uses non-ionizing
radiation (in that way In the patients suspected of suffering from dementia, a primary
safer) use of structural imaging is to investigate the contribution of
Better tolerated by patients Wider application for atrophy and vascular disease in the clinical presentation.
quantitative analysis
Better for lesions with little Image in multiple planes;
or no water content (e.g. therefore superior
10.3.1 ASSESSING AND MEASURING
meningiomas) and views of middle and
ATROPHY
detection of acute posterior fossa, Imaging provides information about the structural integ-
intracerebral haemorrhage pituitary, brain stem, rity and size of the brain. Any reduction in the size of the
and spinal cord brain is a visual end point of cell death and MRI is an excel-
Good for bone abnormalities Greater sensitivity to lent tool to assess and track atrophy. In this way, MRI can be
and detecting calcification detect white matter viewed as a biomarker for neuronal degeneration or injury,
pathology and lesions such as neuron loss, shrinkage and synapse loss (Jack, 2011).
causing epilepsy (such When interpreting scans, it is important to be systematic.
as inflammatory disease A useful algorithm to follow has been proposed by Harper
and hippocampal et al. (2014). Before assessing atrophy, this algorithm guides
sclerosis) the observer to start checking for any ‘surgical pathologies’,

Table 10.3 Main types of MRI sequences

Main sequences:

Conventional sequences:
• T1-weighted: Commonly used clinical scan provides good definition of anatomy and grey/white matter contrast
• T2-weighted: Provides high contrast and definition of soft tissue pathology (increased water content) – useful to detect
ischaemic changes
• Proton density: Provides good brain/cerebrospinal fluid (CSF) contrast
• Fluid-attenuated inversion recovery (FLAIR): Sensitive to changes in white matter (while suppressing the signal from the
CSF) and useful to detect ischaemic changes

Additional sequences:
• T2* gradient echo (or susceptibility weighted imaging): Can identify micro-haemorrhages or micro-bleeds suggestive
of cerebral amyloid angiopathy or small vessel disease (SVD)
• Magnetization transfer imaging (MTI): Sensitive to changes in structural integrity of tissue
• Diffusion-weighted imaging (DWI): Provides information about the microscopic diffusion of water molecules and
integrity of axons. Diffusion tensor imaging (DTI) is a type of diffusion scan taken in multiple planes that can identify
white matter tracts – so called tractography. It gives an impression of microstructure, coherence and connectivity of
white matter tracks – see Chapter 11 for further details
• Perfusion-weighted Imaging (PWI) including arterial spin labelling (ASL): Provides information about the status of brain
tissue perfusion – see Chapter 11 for further details
Structural brain imaging 95

Table 10.4 Contraindications of MRI a standardized way, automated approaches offer reproduc-
• MRI scanners can be noisy, patients feel confined and ibility and efficiency, especially advantageous in large-scale
they are required to remain still for varying amounts multicentre studies. Techniques such as voxel-based mor-
of time to avoid motion artefacts: approximately phometry (VBM) can examine changes in all parts of the
5% –10% of people feel that they are unable to brain, detect grey matter loss and make detailed comparisons
tolerate the scan and may experience claustrophobia. between diagnostic groups. Newer techniques allow the cor-
• Ferromagnetic implants, e.g. pacemakers, tical grey matter to be segmented from the rest of the brain to
intracranial aneurysm clips, cochlear implants, derive a measure of cortical thickness in regions of interest,
neurostimulators: cardiac pacemakers can demonstrating subtle differences as small as 0.2 mm in thick-
deprogram and misfire so structural imaging with ness can be significant (Dickerson et al., 2011).
CT scan is the only option. Atrophy can be measured one point at a time or sequen-
• Ferromagnetic objects may irritate eyes. tially over a long period of time. Cross-sectional studies allow
• Orthopaedic implants are not contraindicated comparisons to be made between diagnostic groups usually
though may introduce local artefacts. with healthy age-matched controls, whereas, longitudinal
studies can measure the same structure in the same individ-
ual at two different time points, thereby reducing the vari-
such as tumours, hematomas and arteriovenous malforma- ance due to inter-subject differences. This technique, usually
tions. This can be followed by an assessment of any signal focused on serial measurements of hippocampal and whole-
changes (mainly relevant in MRI) based on the premise brain volume, is sufficiently sensitive to detect very small vol-
that the appearance within a single tissue type should be ume changes in the order of a few millilitres, which can be
relatively uniform and the presence of hyperintensity or important during the pre-symptomatic or early stages of the
hypointensity within the tissue can often reflect pathology. disease when changes can be subtle (Scahill and Fox, 2007).
When evaluating atrophy, its pattern and extent should be Regulatory authorities are keen for biomarkers to be used in
determined, including areas where brain tissue appears per- clinical trials so the biological impact and safety of new treat-
severed. This would involve checking for general and lobar ments can be appraised. Used as surrogate endpoint, serial
patterns of cortical atrophy, noting any asymmetry and MRI imaging can reduce the number of subjects required
anterior-posterior gradient, as well as any evidence of basal compared to clinical outcome measures (Hampel et al., 2011).
ganglia, brainstem or cerebellum atrophy. T1-weighed MRI Notwithstanding the advantages of serial scanning, there are
or CT is commonly used for detecting atrophic changes. a number of methodological challenges: atrophy rates can
There are a number of ways to assess atrophy, each with vary considerably between subjects, serial scanning of patients
advantages and disadvantages. Visual rating scales offer the with advancing dementia can be problematic; currently, a gap
advantage of speed, simplicity and ease of application in of at least 6 months between scans is required before volume
clinical settings, though they are inherently subjective and change can be consistently detected. There are technical and
limited in terms of quantification. Nevertheless, in the case cost implications, especially for studies that require many
of Alzheimer’s disease (AD), visual rating scales of medial years to determine the predictive relevance of MRI biomark-
temporal lobe atrophy (MTA) can distinguish AD from ers; and ultimately there is a need for post-mortem validation.
controls with a sensitivity and specificity of around 85% With advancing age, approximately 0.5% of brain vol-
(Scheltens et al., 2002), findings that also correlate with neu- ume is lost every year, and as our brains become smaller our
ropathological changes in autopsy (which in turn indicate ventricles become larger, often at a faster rate. The pace of
atrophy in AD is more closely linked to abnormalities in tau this change accelerates with age, particularly after 70 years
pathology than amyloid burden). of age and appears to be associated with greater cognitive
Moving away from visual rating scales, MRI can be used to decline. The hippocampus appears to be particularly sensi-
measure atrophy with a high degree of spatial resolution (<1 tive to the effects of ageing (as well as AD pathology) and
mm), with the development of high resolution imaging allow- atrophies at a faster rate than other structures.
ing the sub-field architecture of the structures such as hippo- These age-related changes are common but not inevitable
campus to be measured. Research centres have developed a and there is considerable variation between changes in indi-
myriad of techniques to try and reliably measure specific struc- viduals. Atrophy is likely to be influenced by a number of
tures using manual, semi-automated or automated approaches. factors, including genetic status (such as presence of apoli-
As expected, these methodologies are more adept at quantify- poprotein E [ApoE] ε4 alleles), health status (such as hyper-
ing structural changes, but require higher levels of technical tension and diabetes), socio-economic status and possibly,
expertise and are less transferable to the clinical setting. dietary and hormonal factors too.
In research settings, measurements are increasingly based Compared to age-matched controls, increased rates
on automated imaging software packages. These approaches of brain atrophy and ventricular enlargement have been
are aimed at achieving quicker, in-depth and less operator- documented in most major forms of dementia, including
dependent way of measuring brain structures, though there AD, dementia with Lewy bodies (DLB), vascular dementia
has been a lack of standardization between research centres (VaD), frontotemporal dementia (FTD), corticobasal degen-
in their design and application. However, when applied in eration (CBD), progressive supranuclear palsy (PSP) and
96 Dementia

Huntington’s disease (HD) (O’Brien et al., 2001; Whitwell vascular cognitive impairment. However, interpreting the
et al., 2007a). However, serial measures of global atrophy relationship between CVD detected on imaging and the
are unlikely to distinguish individual patients with different clinical presentation of an individual patient can be prob-
types of dementia, although, measures of regional atrophy lematic and these reasons are discussed later in the chapter.
may be more discriminating. For example, in AD, the rate of CVD is inherently heterogeneous with diverse underly-
hippocampal atrophy is greater than the global atrophy rate – ing pathophysiological mechanisms. These changes can be
approximately 3%–8% compared to 1%–3% depending on detected on imaging as cortical ischaemic or haemorrhagic
the study. Conversely, patients with FTD have a greater rate strokes due to large vessel disease, subcortical infarcts, lacunes,
of frontal lobe atrophy compared to those with AD (Krueger white matter hyperintensities (WMH) (on MRI), prominent
et al., 2010). Individuals who develop mild cognitive impair- perivascular spaces and cerebral micro-bleeds. In addition, vas-
ment (MCI) also have accelerated patterns of regional atro- cular disease may occur due to a ­subarachnoid haemorrhage,
phy compared to cognitively normal individuals. subdural haematoma or vascular ­ malformations. Hypoxic
damage can result from a significant cerebral hypoperfusion,
10.3.2 ASSESSING AND MEASURING for example, following a cardiac arrest, which may cause wide-
VASCULAR DISEASE spread ischaemic damage, particularly, in ‘watershed’ areas.
MRI studies have recognized that small vessel disease
Neuroimaging plays a central role in characterizing the bur- (SVD) is both prevalent with advancing age and the most com-
den of CVD. Indeed, findings from population-based MRI mon vascular cause of dementia which has emphasized the
studies have helped to shape and redefine our understand- need to define the imaging characteristics of SVD in a more
ing of the spectrum of pathologies that are associated with consistent way. As summarized in Figure 10.1 and discussed

Recent small subcortical White matter Lacune Perivasular space Cerebral microbleed
infarct hyperintensity

Example image

T2
Schematic

DWI FLAIR FLAIR T1/FLAIR T2*/SWI

Usual diamater ≤20 mm Variable 3–15 mm ≤2 mm ≤10 mm

Comment Best identified on DWI Located in white matter Usually have Most linear without Detected on GRE seq.,
hyperintense rim hyperintense rim round or ovoid, blooming
DWI ↑ ↔ ↔/(↓) ↔ ↔
FLAIR ↑ ↑ ↓ ↓ ↔

T2 ↑ ↑ ↑ ↑ ↔

T1 ↓ ↔/(↓) ↓ ↓ ↔
T2*-weighted GRE ↔ ↑ ↔ (↓if haemorrhage) ↔ ↓↓

Figure 10.1 Magnetic resonance imaging (MRI) findings for lesions related to small vessel disease show examples (upper)
and schematic representation (middle) of MRI features for changes related to small vessel disease, with a summary of imag-
ing characteristics (lower) for individual lesions. DWI, diffusion-weighted imaging; FLAIR, fluid-attenuated inversion recovery;
SWI, susceptibility-weighted imaging; GRE, gradient-recalled echo. ↑ Increased signal, ↓ Decreased signal, ↔ Iso–intense
signal. (Reprinted from The Lancet, 12(8), Wardlaw, J.M. et al., Neuroimaging standards for research into small vessel disease
and its contribution to ageing and neurodegeneration, 822–838, Copyright 2013, with permission from Elsevier.)
Structural brain imaging 97

in detail below, Wardlaw et al. (2013) have proposed a stan- That being said, as white matter changes it becomes more
dard set of definitions for the MRI characteristics of SVD. extensive, extending bilaterally into the deeper white matter
tracts as confluent hyperintense areas on T2-weighted and
10.3.2.1 White matter disease FLAIR, then current convention is to infer these changes
likely to result from underlying vascular disease (so-called
In a CT scan, white matter lesions (WMLs) appear as areas ‘WMH of presumed vascular origin’ by Wardlaw et al.,
of reduced attenuation, commonly reported as white matter 2013). The location of DWMH could have an important
hypoattenuation, hypodensities or leukoaraiosis. bearing on the clinical phenotype with the “disconnection”
In MRI scan, WML appear as hyperintense (WMH) on of key cortical–subcortical circuits mediating their effect on
proton density, T2-weighted and fluid-attenuated inversion brain function. PVH, on the other hand, may have less bear-
recovery (FLAIR) MR images, as illustrated in Figure 10.2. ing on the clinical outcome and is less closely coupled with
There have been a number of approaches undertaken to vascular mechanisms.
characterize and rate WMH. Broadly, WHM can be divided The progression of white matter changes are likely to be
into those immediately adjacent to the ventricles (periven- influenced by various cardiovascular factors. The emerging
tricular hyperintensities [PVH]) and those located in the deep picture suggests WML and atrophy share similar risk fac-
white matter (deep white matter hyperintensities [DWMH]). tors, and there may well be an inter-relationship between
As shown in Figure 10.3, PVH can be further subdivided these MRI variables; for example, higher rates of cerebral
into caps (frontal and occipital) and bands and DWMH into atrophy and ventricular dilatation have been associated
punctate foci, early confluent foci and large confluent areas with severe WML. Importantly, WML could also be a target
(Fazekas et al., 1987). In a similar way that is used to measure for treatment, including treatment of hypertension associ-
atrophy, WMH can be assessed using simple rating scales, ated with reduced risk of WML progression in cognitively
semi-quantitative scales or automated software programmes. normal subjects (Firbank et al., 2007).
When interpreting the significance of any change to the
appearance of white matter, it is important to assess the pat- 10.3.2.2 Other changes associated with
tern, location and severity of these changes along with any
small vessel disease
other abnormalities affecting the cortical areas, basal gan-
glia and brain stem. Technical factors, such as, the imag- 10.3.2.2.1 LACUNAE OR LACUNAR INFARCTS
ing parameters used, as well as clinical variables like the
These are small discrete changes deep within the brain
patient’s age and known medical co-morbidities also need
that are thought to be the legacy of an old infarct. As sum-
to be considered. This is necessary because different scan
marized in Figure 10.1, Wardlaw et al. (2013) proposed
sequences and parameters vary in their sensitivity to detect
the definition of a ‘lacune of presumed vascular origin’ as
small vessel disease; WML are extremely common with
a round or ovoid, subcortical, fluid-filled (similar signal
advancing age (8%–95% depending on the study) and ulti-
as cerebrospinal fluid [CSF]) cavity, between 3 mm and
mately they are unlikely to result from a single cause; also
WML commonly coexist with other changes. Indeed, WML
can be found in all major forms of dementia, as well as a host
of other conditions, including multiple sclerosis (MS), amy-
loid angiopathy, hydrocephalus, various leukodystrophies,
cerebral oedema, neurosarcoid and conditions resembling
late life depression. Potential pathological mechanisms for
WMH are shown in Table 10.5. It is important to distin-
guish infarcts from WMH as summarized in Table 10.6 and (a) (b) (c)
illustrated in Figure 10.4.

(d) (e)

FLAIR PD T2 Figure 10.3 Examples of white matter lesions on FLAIR.


(a) No periventricular hyperintensities (PVH). (b) Mild PVH
Figure 10.2 Comparison of FLAIR, proton density (frontal and occipital caps). (c) Moderate to severe PVH
(PD) and T2-weighted images from the same indi- (frontal and occipital caps). (d) Punctate deep white mat-
vidual: note differences in appearance of white matter ter hyperintensities (DWMH) in frontal and parietal lobes.
hyperintensities. (e) Large, early confluent DWMH.
98 Dementia

Table 10.5 Histopathology associated with white matter lesions in dementias


Lesion type Pathology Possible mechanisms
Periventricular hyperintensities Less extensive PVH: loss of In part, secondary to atrophy processes
(PVH) ependymal lining, increased Frontal caps extremely common in late life
interstitial fluid, gliosis, myelin could be considered ‘normal’ and
pallor and dilatation of perivascular secondary to age-related changes
spaces. More extensive PVH probably linked to
More extensive irregular PVH, which vascular risk factors.
extend into the deep white matter:
likely to correspond to areas of
reactive gliosis, lacunar infarction,
loss of myelin, perivascular spaces
and arteriolar thickening.
Deep white matter hyperintensities Punctuate WMH: reduced myelination Punctate lesions are probably not
(DWMH) and perivenous damage. associated with ischaemia/infarction.
Early confluent areas: show evidence Ischaemic damage implicated as lesions
of perivascular rarefaction of become larger and more confluent.
myelin, fibre loss and gliosis.
Confluent WMH: have a similar Other possible causes of white matter
histopathology as irregular PVH, change include focal cerebral oedema,
including microcystic infarcts. hypoxia, acidosis, chronic perfusion
changes, Wallerian degeneration of
axons secondary to neuronal loss in
neocortex.

Table 10.6 Differences between cerebral infarcts and DWMH on MRI

Infarcts DWMH (of presumed vascular origin)


• Hypointense on T1 and proton density weighted images • Hyperintense on T2/FLAIR and proton density
weighted images
• Hyperintense on T2-weighted images • Isodense on T1-weighted images (can be hypointense)
• Well defined, wedge shape if peripheral • Range from punctuate to diffuse confluent lesions
• Often single or low numbers • Often multiple
• Often evidence of cortical extension • Restricted to white matter with no cortical extension
• May be associated with focal enlargement of ventricles • Ventricles and sulci remain unchanged
and sulci

about 15 mm in diameter, consistent with a previous acute gradient-echo (or susceptibility weighted imaging). These
small deep brain infarct or haemorrhage in the territory changes are thought to be due to a small leakage of blood
of one perforating arteriole. Lacunar infracts occur in which has left a residue of iron (haemosiderin). Population-
about 20% of older people considered healthy without any based MRI studies indicate CMB increases significantly
significant symptom, though they are associated with an with advancing age and presence of ApoE 4 allele reaches as
increased risk of stroke (Vermeer et al., 2007) and their high as nearly 40% of those over 80 years (Sveinbjornsdottir
impact may be greater if there are pre-existing patholo- et al., 2008; Vernooij et al., 2008; Romero et al., 2014).
gies, such as AD. However, whether CMB has a direct impact on cognitive
function is still to be determined, especially because they
10.3.2.2.2 MICRO-BLEEDS often coexist with other vascular pathologies.
Interest in the significance, and therefore, detection of It has been hypothesized that CMB result from at
cerebral microhaemorrhages or micro-bleeds (CMBs) has least two pathological mechanisms. CMB occurring in
increased over the recent years based on a convergence of deeper structures, such as, the thalamus, basal ganglia,
findings that indicate their prevalence and clinical relevance brainstem and cerebellum, result from vascular micro-
is potentially greater than previously appreciated. Partly, angiopathy caused by hypertension and atherosclerosis.
this is due to the application of newer MR sequences which In addition to this, deep CMB often coexists with other
are much more sensitive at detecting these small (2–5 mm) types of SVD, such as WMH and lacunar infarcts. CMB
round foci of low but homogeneous signal, notably T2* are also associated with vascular risk factors and an
Structural brain imaging 99

increased risk of stroke recurrence (Cordonnier et al., though, often prominent in the medial and posterior brain
2007; Vernooij et al., 2008). regions, asymmetry may, however, be present. It is also pro-
Conversely, lobar CMBs are thought to be more closely gressive: individuals with AD lose brain volume at approxi-
related to cerebral amyloid angiopathy (CAA). The presence mately two-to-four times more than the rate of healthy
of CAA could be a possible factor in explaining why some controls (range 1%–4% vs. 0.3%–0.7% depending on study).
patients with AD treated with β-amyloid immunothera- Broadly, atrophy is envisaged to progress in a way that mirrors
pies experienced an increase in CMBs: the hypothesis of an the established version of Braak staging. Indeed, atrophy on
antibody-mediated inflammation which further compro- MRI in AD can be seen as a marker for neurofibrillary tangles
mised the integrity of blood vessels already affected by CAA and tau related neurodegeneration (Jack et al., 2010, 2011b).
is plausible but yet to be corroborated. In response to con- That being said, generalized atrophy per se can be rela-
cerns that patients in AD immunotherapy trials were expe- tively non-specific, and characteristically, AD is associated
riencing ‘vasogenic oedema’ and microhaemorrhages, the with early and accelerated atrophy of medial temporal lobe
amyloid-related imaging abnormalities (ARIA) MR criteria structures. By the time an individual is diagnosed with AD,
was introduced to standardized safety monitoring (Sperling he/she can have hippocampi, parahippocampi, entorhinal
et al., 2011). cortex and amygdalae that are 40%–50% smaller than nor-
mal. This level of atrophy can easily be visualized on MRI,
as illustrated in Figures 10.5 and 10.6.
MTA is also common, occurring in 80%–90% of patients
10.4 ALZHEIMER’S DISEASE with AD, compared to 5%–10% occurrence in healthy sub-
jects. MTA has been defined in numerous ways, but on an
MRI has been used to study established clinical AD (as a average it has a specificity of 80% to distinguish AD from
diagnostic marker) and increasingly, the preclinical stages controls (Frisoni et al., 2013), with some studies reaching
of AD (as a prognostic marker), variably referred to as the accuracy levels of 85%–100% (Jack, 2011).
asymptomatic, presymptomatic, preclinical and prodromal MTA is a relatively sensitive marker of AD pathology,
phases of AD progression (Dubois et al., 2010). especially tau pathology, which is more closely linked to the
rate of brain loss than β-amyloid burden (Whitwell et al.,
2008). Importantly, MTA is not unique to AD and can occur
10.4.1 EARLY AND ESTABLISHED AD in a number of other conditions, including other types of
dementia, such as DLB, VaD and FTD, as described below.
Generalized brain atrophy and ventricular enlargement are
The overlap in tau pathologies between different types of
common in AD. Typically, these changes are symmetrical,
dementia is one factor why MTA is less specific and links

(a)

(b)

(a) (b)

(c)

(d)

(c) (d)

Figure 10.4 Example of frontal stroke on (a) axial FLAIR,


(b) proton density, (c) coronal T1-weighted and Figure 10.5 (a–d) Examples of graded medial temporal
(d) T2-weighted. atrophy (MTA): arrows point to hippocampi.
100 Dementia

(a) 10.4.2 PROGNOSTIC SIGNIFICANCE


OF MRI IN AD
AD
Accelerated atrophy rates, notably affecting the hippo-
campus, can be evident in about 3–10 years prior to transi-
(b)
tion to dementia, depending on the area of study. By the
Control time mild symptoms are apparent, hippocampal volume
reductions may exceed 25%. Jack and collaborators from
the Alzheimer’s Disease Neuroimaging Initiative (ADNI)
have proposed an AD biomarker model to map the stag-
(c)
ing of the illness. This model, evolving as new evidence
DLB
emerges, broadly hypothesizes that changes to topo-
graphical structures as measured by MRI are secondary
to neuronal injury and in particular, tau related neurode-
Figure 10.6 Comparison of medial temporal atrophy in generation. These structural changes are a downstream
(a) Alzheimer’s disease (AD) (b) dementia with Lewy consequence of other pathologies, putatively abnormali-
­bodies (DLB) and (c) normal controls. ties in β-amyloid metabolism detectable via CSF and PET
amyloid biomarkers years before the MRI changes are evi-
between atrophy and neuropathology are probably, less dent (Jack et al., 2010). This model may account for the
closely associated with advancing age (Burton et al., 2009, reason why MRI appears to perform less, as both a diag-
2012). Lesser degrees of MTA are also seen in Parkinson’s nostic and prognostic marker for AD when compared to
disease, temporal lobe epilepsy, schizophrenia and depres- amyloid imaging, with the caveat that head to head com-
sion. Ultimately, MRI can provide secondary, not primary parisons are few in number and that the accuracy of each
evidence of AD (de Leon et al., 2007). AD biomarker as much dependent on how the biomarker
Furthermore, as MTA occurs with advancing age the measured on the biomarker itself (Frisoni et al., 2013). In
specificity of MTA as a biomarker for AD declines with the latest revision of this model, 18F-fluroodeoxyglucose
increase in age, especially above 80 years. There is evidence PET and MRI changes are modelled to occur coinciden-
that the link between atrophy and neuropathology disso- tally as the last biomarkers become abnormal, but they are
ciates advancing age and becomes less predictable (Savva most closely coupled to the emergence and progression of
et al., 2009; Burton et al., 2012). Conversely, individuals cognitive impairment (Jack et al., 2013). This model helps
who develop early onset AD may have less noticeable MTA to conceptualize the potential interrelationship between
(especially the non-­amnestic form of AD) but prominent the different imaging biomarkers but the clinical utility of
posterior cortical atrophy, affecting the posterior cingulated predicting outcome for individual patients is still inaccu-
and precuneus (Frisoni et al., 2007). The pattern of atrophy rate. This temporal ordering of MRI findings in relation
in individuals affected by autosomal dominant AD (ADAD) to other biomarkers and clinical symptoms is supported
can also differ from individuals with sporadic AD, with evi- by findings from two separate cohorts involving subjects
dence of smaller basal ganglia, notably putamen and thala- with sporadic AD and subjects with ADAD Presenilin 1
mus which may be evident in asymptomatic subjects (Cash E280A mutation carriers (Villemagne et al., 2013; Fleisher
et al., 2013; Lee et al., 2013). et al., 2015).
Although MTA appears to be necessary for the develop- Individuals with MCI are at an increased risk of devel-
ment of AD, it is not sufficient to explain all symptomatic oping dementia (see Chapter 20), with such individuals
changes and as Jack et al. (2002) speculate, MTA is a good increasingly thought to represent a group with ‘prodromal
marker for early AD but as the disease progresses beyond AD’. Detection of MCI due to prodromal AD is, therefore,
the limbic areas, measures of neocortical atrophy may be a important and the role of neuroimaging in MCI has been
better marker of advanced disease. extensively studied. Overall, baseline temporal lobe atro-
To summarize, MTA occurs early in the course of AD phy correlates with future cognitive decline and conversion
and increases with increasing severity of dementia. As MTA from MCI to AD with the changes in MCI usually being
is associated with the clinical diagnosis of AD and transition intermediate between normal subjects and AD subjects.
from MCI to AD, there have been recommendations to include The intermediate nature of these results is reflected in the
estimates of MTA in revised criterion for the diagnosis of AD relatively lower sensitivity (62%) and specificity (73%) of
(Dubois et al., 2007) and MCI due to AD (Albert et al., 2011). measures of MTA on MRI for the discrimination of patients
In terms of WML, most studies show that patients with with MCI, who subsequently progress to AD from patients
AD have more extensive PVH and DWMH than controls. with MCI who do not progress to AD (Frisoni et al., 2013).
Loss of white matter integrity probably occurs for a number It is possible that composite MRI measures involving MTA
of reasons, including a secondary downstream event to the plus other grey matter regions and white matter changes
primary cortical pathology, amyloid angiopathy and con- can improve the prediction of individual subjects with MCI
current ischaemic damage. at risk of developing AD (Misra et al., 2009).
Structural brain imaging 101

More recent studies have focused on evaluating whether pattern of relatively focused atrophy of the midbrain, hypo-
biomarkers can help identify subjects who are cognitively thalamus and substantia innominata, with a relative spar-
normal but destined to develop impairments. Emerging ing of the hippocampus and temporoparietal cortex, is more
findings suggest that this may be possible with mea- likely to occur in DLB compared to AD (Whitwell et al.,
sures of cortical thickness predictive of cognitive decline 2007b).
over 3 years in clinically normal individuals at baseline Structural imaging changes in DLB are broadly similar
(Dickerson et al., 2012). to those seen in patients with Parkinson’s disease dementia
(Watson et al., 2009). Subjects with DLB probably share a
similar distribution, prevalence and severity of WML to AD
on MRI, intermediate in severity between normal controls
10.5 DEMENTIA WITH LEWY BODIES and those with VaD (Barber et al., 1999), though studies
conducted till now are not conclusive (Burton et al., 2006).
The most consistent MRI finding in patients with DLB is
greater preservation of medial temporal lobe (MTL) struc-
tures compared to those with AD (Barber et al., 2000, 2001;
Burton et al., 2009). These autopsy verified differences have 10.6 FRONTOTEMPORAL DEMENTIA
been observed visually, as shown in Figure 10.6, and on a
range of more detailed morphometric measures, includ- FTD is characterized by atrophy of the frontal and anterior
ing cortical thickness (Watson et al., 2015). Preservation temporal lobes. A normal MRI scan does not necessarily
of MTL has been incorporated into the revised criteria exclude the illness, especially early stages of the illness, and
for clinical diagnosis of DLB (McKeith et al., 2005), and is in such instances functional imaging may assist the diagno-
consistent with the finding that patients with DLB tend to sis. If both structural and functional imagings are normal,
maintain memory function for a longer time compared to however, the risk of making a false positive diagnosis of FTD
patients with AD. may be increased (Kipps et al., 2009). As subjects with FTD
Therefore, observing preservation of MTL can be use- can have varying degrees of MTA, the pattern of prominent
ful in distinguishing DLB and AD, as it would be unusual frontotemporal anterior atrophy can assist in its differentia-
to find hippocampal volumes significantly preserved in tion from other types of dementia, such as AD. Annual rates
AD: the inference being that a patient without MTL (or of whole-brain atrophy are increased compared to controls
indeed global atrophy) is unlikely to have significant bur- in all types of dementia. Asymmetrical pattern of atrophy
den of underlying AD-type pathology. Over a long period is also suggestive of FTD pathology than AD pathology
of time, patients with DLB also show less progression in (Harper et al., 2014). FTD consists of a number of clinical
MTA (and probably global atrophy) than those with AD: phenotypes with differing, though, overlapping MRI find-
again the inference here is accelerated atrophy rates reflect ings, as summarized in Table 10.7 (see also Figure 10.7).
AD-pathology more than DLB-pathology (Mak et al., Approximately 20% of patients with FTD have an auto-
2015a,b; Nedelska et al., 2015). somal dominant pattern of inheritance, primarily linked
However, the clinical utility of MTA to distinguish DLB to one of three mutations (progranulin, microtubule-­
and AD is likely to weaken with advancing age and dis- associated protein tau and chromosome nine open reading
ease progression and this finding in isolation is not reliable frame 72). Different patterns of atrophy have been described
enough to make a specific diagnosis of DLB for an individ- for these three groups compared to sporadic FTD (Rohrer
ual patient. In this regard, a dopamine transporter scan is and Warren, 2011; Whitwell and Josephs, 2012), as summa-
likely to provide greater discrimination at distinguishing rized in Table 10.8. As seen in subject with ADAD, individ-
DLB from AD than MRI scan. uals who carry one of these genetic mutations can develop
From a research perspective, findings suggest combin- subtle structural changes; in this instance in the insula and
ing measures of cortical thickness from different brain temporal lobes onset of symptoms was predicted 10 years
areas that can enhance the discrimination of DLB from AD, before (Rohrer et al., 2015).
achieving a sensitivity and specificity of over 80% (Lebedev Provisional findings indicate WML is common with AD
et al., 2013). In the future, multimodal imaging protocols and DLB, less severe than VaD though. Their pathological
that combine structural and functional modalities may also basis, however, is likely to be different and reflect gliosis
improve discrimination further. For example, a combined rather than ischaemic damage or amyloid angiopathy.
measure of hippocampal atrophy, cortical amyloid and
occipital lobe metabolism achieved an accuracy of 98% for
distinguished DLB from AD, better than any of the mea-
surements (Kantarci et al., 2012).
10.7 VASCULAR DEMENTIA
There is also a general trend among patients with DLB
who have less global atrophy compared to patients with The term vascular dementia implies that there is a defin-
AD, but greater atrophy of subcortical structures, including able threshold above which the presence of dementia can
the substantia innominata and dorsal midbrain. Indeed, a be attributed to the burden of CVD. The inherent difficulty
102 Dementia

in demarcating this threshold, coupled with a broadening Broadly, the research criteria for VaD (Chui et al., 1992;
in our understanding of the spectrum and continuum of Roman et al., 1993) is based on linking the presence of
CVD, has led many to prefer the concept of ‘vascular cog- dementia (which may be non-amnestic in profile) to CVD
nitive impairment’ for defining this relationship (Gorelick manifest in imaging as cortical infarcts, infarcts in strategic
et al., 2011). brain areas (such as the thalamus), multiple lacunar infarcts,
extensive white matter change (usually defined as area >25%),
or combinations thereof (so-called mixed cortical and sub-
Table 10.7 MRI findings in frontotemporal dementia cortical dementia). CVD may also be associated with atro-
(FTD) by clinical phenotype phy and ventricular enlargement corresponding to an area of
infarction with subsequent local cell loss. SVD is now consid-
Characteristic Structural ered to be the commonest form of VaD and associated ‘sub-
Clinical Phenotype Change cortical’ vascular pathologies, as described in Figure 10.1.
Behavioural variant of Varying degrees of atrophy Implicit in the criteria for VaD is that the clinical sig-
FTD occurring along the frontal- nificance of CVD can be inferred by considering their size,
to-temporal lobe axis, number and location. But the precise relationship between
occasionally including the cognitive impairment and vascular change needs fur-
parietal lobes ther exploration and can be confounded by the presence
Progressive nonfluent Usually symmetrical, left-sided of other pathologies, such as AD. Probably, the strongest
aphasia (PA) perisylvian atrophy (inferior evidence linking the severity of CVD with greater cogni-
frontal and antero-superior tive impairment relates to the burden of WML and SVD.
temporal regions) Bearing in mind the effect different imaging modalities and
Semantic dementia Usually asymmetrical (especially sequences have on the detection of CVD, the more exten-
(SD) as shown in anterior left temporal pole) sive these changes, the more we can infer the presence of
Figure 10.7 and significant (‘knife edge’ vascular pathology and the fact that this change is likely to
appearance of gyri) be clinically relevant. In the case of WML, this would be
Atrophy may become bilateral the presence of large confluent DWMH. However, as dis-
as the illness progresses, but cussed beforehand, mild degrees of WML are almost uni-
usually atrophy still left versal in cognitively normal people with advancing age and
> right hemisphere commonly seen in patients with degenerative dementia, so,
given the varied pathologies that can cause WML, assigning
the clinical relevance to this level of change is less straight-
Table 10.8 MRI findings in FTD by genotype forward. Determining the clinical relevance of other types
Characteristic structural of SVD in individual patients can also be a challenge. For
Genotype – mutations changea example, incidental ‘silent’ lacunar infarcts occur in 20%–40%
of elderly people and CMB is equally common later in life.
Progranulin gene Asymmetrical pattern of
Discerning the impact of large vessel strokes can raise
atrophy
similar challenges. Approximately 30% of people who have
Early involvement of the
suffered a stroke progress to develop a dementia, but the
temporal, inferior frontal
and inferior parietal lobes
Microtubule-associated Symmetrical pattern
protein tau gene Involvement of anteromedial
temporal and orbitofrontal
lobes
Chromosome 9 open Symmetrical pattern
reading frame 72 Variable pattern of cortical
atrophy – usually involving
dorsolateral, medial and
orbitofrontal lobes, with
additional loss in anterior
temporal lobes, parietal
lobes, occipital lobes and
cerebellum in some
subjects
Sporadic FTD Frontal and anterior temporal Figure 10.7 Patient with clinical diagnosis of seman-
tic dementia showing bilateral anterior temporal lobe
atrophy
atrophy, left > right side: coronal and axial T1-weighted
a
Rohrer and Warren, 2011; Whitwell and Josephs, 2012 images. (Courtesy of Prof. T. Griffiths.)
Structural brain imaging 103

findings from studies examining the relationship between


topography and severity (number and volume) have been 10.8 OTHER DEMENTIAS
inconsistent. It is also likely, that, ‘non-vascular’ variables
influence whether cognitive deficits develop in the presence Less common types of dementia with relatively distinct
of vascular disease. For example, generalized changes such neuroradiological features on MRI are summarized in
as cortical atrophy and MTA as well as variables, such as, Table 10.9. Uncommon types of dementia are further dis-
age and level of education may play an important role in the cussed in Chapter 20.
development of post-stroke dementia.
Finally, the prevalence of pathologically confirmed mixed
dementias (AD and VaD) is now widely recognized and it is
possible that the two pathologies have a synergistic effect 10.9 FINAL COMMENTS
such that the presence of AD-type pathology can increase
the clinical sequelae of CVD, and vice-versa. Furthermore, There is a convergence of opinion that structural neuro-
there is evidence, that, when both cortical atrophy (such as imaging should be considered in the investigation of all
MTA) and CVD coexist, measures of atrophy more closely patients with suspected dementia and where possible per-
explain the variance in cognitive function, suggesting con- formed at least once during their illness. Functional imag-
comitant AD pathology has a significant impact on the ing can also have an important, complimentary role in
emerging clinical presentation. The recognition that vas- enhancing diagnostic accuracy (see Chapter 11). An impor-
cular and degenerative diseases share similar risk factors tant question is perhaps, ‘If I need to make a diagnosis and
introduces another level of complexity to their relationship. plan treatment, what sort of imaging do I need to perform?’
As Scheltens (2009) states, the absence of operational crite- A CT scan will, for cost and practical reasons, usually be
ria for mixed dementia means that the clinician is often left the imaging modality most often used, although an MRI, if
to judge which diagnostic category best fits! accessible and feasible, would almost always be the superior

Table 10.9 MRI findings in other diseases associated with cognitive impairment

Disorder MRI findings


Variant Creutzfeldt–Jakob disease (vCJD) Symmetrical hyperintensity in the pulvinar (posterior) nuclei of
the thalamus relative to the anterior thalamus (pulvinar sign).
Other features are hyperintensity of the dorsomedial
thalamic nuclei, caudate head and periaqueductal gray
matter (PG)
Sporadic CJD (sCJD) High signal changes in the putamen, caudate and/or thalamus
and cortical gyri (cortical ribboning). DWI has a high
sensitivity and specificity for sCJD, (especially in detecting
cortical changes compared with other MRI sequences)
Huntington Chorea (HC) Bilateral caudate atrophy may also have reduced volume of
other basal ganglia structures and frontal lobes
Progressive supranuclear palsy (PSP) Brain stem atrophy and non-specific mild atrophy
Corticobasal degeneration May have bilateral (symmetrical) frontal lobe atrophy possibly
extending posteriorly (paracentral region)
Multisystem atrophy (MSA) Olivopontocerebellar or basal ganglia atrophy depending on
variant
Normal pressure hydrocephalus (NPH) Marked ventricular enlargement, with rounding of anterior
horns of lateral ventricles. Ventricular enlargement is
proportionate to sulcal widening – sulci normal, no
significant white matter pathology
CADASIL (cerebral autosomal dominant arteriopathy Evidence subcortical infarcts and confluent hyperintensities
with subcortical infarcts and leukoencephalopathy) potentially extending into the temporal poles (which may be
non-enhancing)
Multiple sclerosis (demyelinating disease) Usually, one T2 lesion at least in two of four locations
(juxtacortical, periventricular, infratentorial, and spinal cord)
and a new T2 lesion on a follow-up scan. Consider using
contrast agent (e.g. gadolinium) T1 to identify new lesions
Infections/inflammation May appear as multifocal/confluent areas of hyperintensities
104 Dementia

imaging modality. Having selected a modality, it is impor- Cordonnier, C., Al-Shahi Salman R. and Wardlaw, J. (2007).
tant to interpret what we ‘see’ in the context of clinical his- Spontaneous brain microbleeds: Systematic review,
tory in order to arrive at a clinical diagnosis that fits best. subgroup analyses and standards for study design and
reporting. Brain, 130 (Pt 8): 1988–2003.
de Leon, M.J., Mosconi, L., Blennow, K. et al. (2007).
Imaging and CSF studies in the preclinical diagnosis of
REFERENCES Alzheimer’s disease. Annals of the New York Academy
of Sciences, 1097: 114–114.
Albert, M.S., DeKosky, S.T., Dickson, D. et al. (2011). Dickerson, B.C., Stoub, T.R., Shah, R.C. et al. (2011).
The diagnosis of mild cognitive impairment due Alzheimer-signature MRI biomarker predicts AD
to Alzheimer’s disease: Recommendations from dementia in cognitively normal adults. Neurology, 76
the National Institute on Aging-Alzheimer’s Association (16): 1395–1402.
workgroups on diagnostic guidelines for Alzheimer’s Dickerson, B.C., Wolk, D.A. and Alzheimer’s Disease
disease. Alzheimer’s Dementia, 7 (3): 270–279. Neuroimaging Initiative (2012). MRI cortical
Barber, R., Ballard, C., McKeith, I.G., et al. (2000). MRI thickness biomarker predicts AD-like CSF and
volumetric study of dementia with Lewy bodies: A cognitive decline in normal adults. Neurology, 78 (2):
comparison with Alzheimer’s disease and vascular 84–90.
dementia. Neurology, 54: 1304–1309. Dubois, B., Feldman H.H., Jacova, C. et al. (2007).
Barber, R., McKeith, I.G., Ballard, C. et al. (2001). A com- Research criteria for the diagnosis of Alzheimer’s
parison of medial and lateral temporal lobe atrophy in disease: Revising the NINCDS-ADRDA criteria. Lancet
dementia with Lewy bodies and Alzheimer’s dis- Neurology, 6 (8): 734–746.
ease: MRI volumetric Study. Dementia and Geriatric Dubois, B., Feldman, H.H. Jacova, C. et al. (2010). Revising
Cognitive Disorders, 12: 198–205. the definition of Alzheimer’s disease: A new lexicon.
Barber, R., Scheltens, P., Gholkar, A. et al. (1999). Lancet Neurology, 9 (11): 1118–1127.
White matter lesions on magnetic resonance imag- Fazekas, F., Chawluk, J.B., Alavi, A. et al. (1987). MR signal
ing in dementia with Lewy bodies, Alzheimer’s abnormalities at 1.5T in Alzheimer’s disease and normal
disease, vascular dementia, and normal aging. ageing. American Journal of Roentgenology, 149:
Journal of Neurology, Neurosurgery and Psychiatry 67: 351–356.
66–72. Firbank, M.J., Wiseman, R.M., Burton, E.J. et al. (2007).
Burton, E.J., Barber, R., Mukaetova-Ladinska, E.B. et al. Brain atrophy and white matter hyperintensity change
(2009). Medial temporal lobe atrophy on MRI differ- in older adults and relationship with blood pressure.
entiates Alzheimer’s disease from dementia with Lewy Journal of Neurology, 254: 713–721.
bodies and vascular cognitive impairment: A prospec- Fleisher, A.S., Chen, K., Quiroz, Y.T. et al. (2015).
tive study with pathological verification of diagnosis. Associations between biomarkers and age in the
Brain, 132 (Pt 1): 195–203. Presenilin 1 E280A autosomal dominant Alzheimer
Burton, E.J., McKeith, I.G., Burn, D.J. et al. (2006). disease kindred: A cross-sectional study. JAMA
Progression of white matter hyperintensities in Neurology, 72 (3): 316–324.
Alzheimer disease, dementia with Lewy bodies, and Frisoni, G.B., Bocchetta, M., Chételat, G. et al. (2013).
Parkinson disease dementia: A comparison with normal Imaging markers for Alzheimer disease: Which vs how.
aging. The American Journal of Geriatric Psychiatry, 14 Neurology, 81 (5): 487–500.
(10): 842–849. Frisoni, G.B., Pievani, M., Testa, C. et al. (2007). The
Burton, E.J., Mukaetova-Ladinska, E.B., Perry, R.H. topography of grey matter involvement in early
et al. (2012). Quantitative neurodegenerative and late onset Alzheimer’s disease. Brain, 130:
pathology does not explain the degree of hippo- 720–730.
campal atrophy on MRI in degenerative dementia. Gorelick, P.B., Scuteri, A., Black, S.E. et al. (2011). Vascular
International Journal of Geriatric Psychiatry, 27 (12): contributions to cognitive impairment and demen-
1267–1274. tia: A statement for healthcare professionals from
Cash, D.M., Ridgway, G.R., Liang, Y. et al. (2013). the American Heart Association/American Stroke
The pattern of atrophy in familial Alzheimer dis- Association. Stroke, 42 (9): 2672–2713.
ease: Volumetric MRI results from the DIAN study. Hampel, H., Wilcock, G. Andrieu, S. et al. (2011).
Neurology, 81 (16): 1425–1433. Biomarkers for Alzheimer’s disease therapeutic trials.
Chui, H.C., Victoroff, J.I., Margolin, D. et al. (1992). Criteria Progress in Neurobiology, 95 (4): 579–593.
for the diagnosis of ischaemic vascular dementia pro- Harper, L., Barkhof, F., Scheltens, P. et al. (2014). An algo-
posed by the State of California Alzheimer’s Disease rithmic approach to structural imaging in dementia.
Diagnostic and Treatment Centres. Neurology, 42: Journal of Neurology, Neurosurgery, & Psychiatry, 85
473–480. (6): 692–698.
Structural brain imaging 105

Hort, J., O’Brien, J.T., Gainotti, G. et al. (2010). EFNS Mak, E., Su, L., Williams, G.B. et al. (2015a). Longitudinal
guidelines for the diagnosis and management of assessment of global and regional atrophy rates in
Alzheimer’s disease. European Journal of Neurology, Alzheimer’s disease and dementia with Lewy bodies.
17 (10): 1236–1248. Neuroimage: Clinical, 7: 456–462.
Jack, C.R. Jr. (2011). Alliance for aging research AD bio- Mak, E., Su, L., Williams, G.B. et al. (2015b). Progressive
markers work group: Structural MRI. Neurobiology of cortical thinning and subcortical atrophy in demen-
Aging, 32 (Suppl. 1): S48–S57. tia with Lewy bodies and Alzheimer’s disease.
Jack, C.R. Jr., Barkhof, F., Bernstein, M.A. et al. (2011a). Neurobiology of Aging, 36 (4): 1743–1750.
Steps to standardization and validation of hippocam- McKeith, I.G., Dickson, D.W., Lowe, J. et al. (2005).
pal volumetry as a biomarker in clinical trials and diag- Diagnosis and management of dementia with
nostic criterion for Alzheimer’s disease. Alzheimer’s & Lewy bodies: Third report of the DLB Consortium.
Dementia, 7 (4): 474–485. Neurology, 65 (12): 1863–1872.
Jack, C.R. Jr., Dickson, D.W., Parisi, J.E. et al. (2002). Misra, C., Fan, Y. and Davatzikos, C. (2009). Baseline and
Antemortem MRI findings correlate with hippocam- longitudinal patterns of brain atrophy in MCI patients,
pal neuropathology in typical aging and dementia. and their use in prediction of short-term conver-
Neurology, 58: 750–757. sion to AD: Results from ADNI. Neuroimage, 44 (4):
Jack, C.R. Jr., Knopman, D.S., Jagust, W.J. et al. (2010). 1415–1422.
Hypothetical model of dynamic biomarkers of the National Institute of Health and Care Excellence (NICE)
Alzheimer’s pathological cascade. Lancet Neurology, 9 (2006). Dementia: Supporting people with dementia
(1): 119–128. and their carers in health and social care 2006 NICE
Jack, C.R. Jr., Knopman, D.S., Jagust, W.J. et al. (2013). guidelines [CG42] https://www.nice.org.uk/guidance/
Tracking pathophysiological processes in Alzheimer’s cg42. Accessed 6 June 2016.
disease: An updated hypothetical model of dynamic Nedelska, Z., Ferman, T.J. and Boeve, B.F. (2015). Pattern
biomarkers. Lancet Neurology, 12 (2): 207–216. of brain atrophy rates in autopsy-confirmed demen-
Jack, C.R. Jr., Vemuri, P., Wiste, H.J. et al. (2011b). tia with Lewy bodies. Neurobiology of Aging, 36 (1):
Evidence for ordering of Alzheimer disease biomark- 452–461.
ers. Archives of Neurology, 68 (12): 1526–1535. O’Brien, J.T., Paling, S., Barber, R. et al. (2001). Progressive
Kantarci, K., Lowe, V.J., Boeve, B.F. et al. (2012). brain atrophy on serial MRI in dementia with Lewy
Multimodality imaging characteristics of dementia bodies, AD, and vascular dementia. Neurology, 56:
with Lewy bodies. Neurobiology of Aging, 33 (9): 1386–1388.
2091–2105. Patterson, C., Gauthier, S., Bergman, H. et al. (2001).
Kipps, C.M., Hodges, J.R., Fryer, T.D. et al. (2009). The recognition, assessment and management of
Combined magnetic resonance imaging and positron dementing disorders: conclusions from the Canadian
emission tomography brain imaging in behavioural Consensus Conference on Dementia. Canadian
variant frontotemporal degeneration: Refining the Journal of Neurological Sciences, 28 (Suppl. 1):
clinical phenotype. Brain, 132 (Pt 9): 2566–2578. S3–S16.
Knopman, D.S., DeKosky, S.T., Cummings, J.L. et al. Rohrer, J.D., Nicholas, J.M., Cash, D.M. et al. (2015).
(2001). Practice parameters: Diagnosis of dementia Presymptomatic cognitive and neuroanatomical
(an evidence-based review). Report of the Quality changes in genetic frontotemporal dementia in the
Standards Subcommittee of the American Academy of Genetic Frontotemporal Dementia Initiative (GENFI)
Neurology. Neurology, 56: 1143–1153. study: A cross-sectional analysis. Lancet Neurology, 14
Krueger, C.E., Dean, D.L., Rosen, H.J. et al. (2010). (3): 253–262.
Longitudinal rates of lobar atrophy in frontotemporal Rohrer, J.D. and Warren, J.D. (2011). Phenotypic signa-
dementia, semantic dementia, and Alzheimer’s disease. tures of genetic frontotemporal dementia. Current
Alzheimer Disease & Associated Disorder, 24 (1): 43–48. Opinion in Neurology, 24 (6): 542–549.
Lebedev, A.V., Westman, E., Beyer, M.K. et al. (2013). Roman, G.C., Tatemichi, T., Erkinjuntti, T. et al. (1993).
Multivariate classification of patients with Alzheimer’s Vascular dementia: Diagnostic criteria for research
and dementia with Lewy bodies using high-dimen- studies. Report of the NINDS-AIRENS international
sional cortical thickness measurements: An MRI sur- workshop. Neurology, 43: 250–260.
face-based morphometric study. Journal of Neurology, Romero, J.R., Preis, S.R., Beiser, A. et al. (2014). Risk fac-
260 (4): 1104–1115. tors, stroke prevention treatments, and prevalence of
Lee, G.J., Lu, P.H., Medina, L.D. et al. (2013). Regional cerebral microbleeds in the Framingham Heart Study.
brain volume differences in symptomatic and pres- Stroke, 45 (5): 1492–1494.
ymptomatic carriers of familial Alzheimer’s disease Savva, G.M., Wharton, S.B., Ince, P.G. et al. (2009). Age,
mutations. Journal of Neurology, Neurosurgery & neuropathology, and dementia. The New England
Psychiatry, 84 (2): 154–162. Journal of Medicine, 360 (22): 2302–2309.
106 Dementia

Scahill, R.I. and Fox, N.C. (2007). Longitudinal imaging Wardlaw, J.M., Smith, E.E., Biessels, G.J. et al. (2013).
in dementia. The British Journal of Radiology, 80: STandards for ReportIng Vascular changes on nEu-
592–598. roimaging (STRIVE v1). Neuroimaging standards for
Scheltens, P. (2009). Imaging in Alzheimer’s disease. research into small vessel disease and its contribution
Dialogues in Clinical Neuroscience, 11 (2): 191–199. to ageing and neurodegeneration. Lancet Neurology,
Scheltens, P., Fox, N., Barkhof, F. et al. (2002). Structural 12 (8): 822–838.
magnetic resonance imaging in the practical assess- Watson, R., Blamire, A.M. and O’Brien, J.T. (2009).
ment of dementia: Beyond exclusion. Lancet Magnetic resonance imaging in lewy body dementias.
Neurology, 1: 13–21. Dementia and Geriatric Cognitive Disorders, 28 (6):
Sperling, R.A., Jack, C.R., Jr., Black, S.E. et al. (2011). 493–506.
Amyloid-related imaging abnormalities in amyloid- Watson, R., Colloby, S.J., Blamire, A.M. et al. (2015).
modifying therapeutic trials: Recommendations from Assessment of regional gray matter loss in dementia
the Alzheimer’s Association Research Roundtable with Lewy bodies: A surface-based MRI analysis. The
Workgroup. Alzheimer’s Dementia, 7 (4): 367–385. American Journal of Geriatric Psychiatry, 23 (1): 38–46.
Sveinbjornsdottir, S., Sigurdsson, S., Aspelund, A. et al. Whitwell, J.L., Jack, C.R., Jr., Parisi, J.E. et al. (2007a).
(2008). Cerebral microbleeds in the population based Rates of cerebral atrophy differ in different degenera-
AGES-Reykjavik study: Prevalence and location. tive pathologies. Brain, 130: 1148–1158.
Journal of Neurology, Neurosurgery, & Psychiatry, 79 Whitwell, J.L. and Josephs, K.A. (2012). Recent advances
(9): 1002–1006. in the imaging of frontotemporal dementia. Current
Vermeer, S.E., Longstreth, W.T., Jr. and Koudstaal, P.J. Neurology and Neuroscience Reports, 12 (6):
(2007). Silent brain infarcts: A systematic review. 715–723.
Lancet Neurology, 6 (7): 611–619. Whitwell, J.L., Josephs, K.A., Murray, M.E. et al. (2008).
Vernooij, M.W., van der Lugt, A., Ikram, M.A. et al. (2008). MRI correlates of neurofibrillary tangle pathology
Prevalence and risk factors of cerebral microbleeds: The at autopsy: A voxel-based morphometry study.
Rotterdam Scan Study. Neurology, 70 (14): 1208–1214. Neurology, 71 (10): 743–749.
Villemagne, V.L., Burnham, S., Bourgeat, P. et al. (2013). Whitwell, J.L., Weigand, S.D., Shiung, M.M. et al. (2007b).
Amyloid β deposition, neurodegeneration, and cogni- Focal atrophy in dementia with Lewy bodies on MRI:
tive decline in sporadic Alzheimer’s disease: A prospec- A distinct pattern from Alzheimer’s disease. Brain, 130
tive cohort study. Lancet Neurology, 12 (4): 357–367. (Pt 3): 708–719.
11
Functional brain imaging and connectivity in
dementia

KLAUS P. EBMEIER, NICOLA FILIPPINI, CLARE E. MACKAY, SANA SURI AND


VYARA VALKANOVA

three-dimensional reconstruction of tracer distribution in


11.1 INTRODUCTION the brain gives a biologically meaningful map of blood flow,
receptor binding capacity or similar measures. Collimators
Neuroimaging offers a window into the living brain and thus are used to admit photons coming from a defined direc-
provides unique access to the understanding of normal and tion to the photosensitive crystal. By design, this method
pathological processes associated with ageing. Structural of directional filtering only allows for a small proportion of
imaging has diagnostic and, to some extent, prognostic value emitted photons to be detected, which limits the sensitiv-
in dementia (see Chapter 10). This chapter reviews the litera- ity of SPECT. The attenuation of γ rays during their path
ture on functional imaging in dementia and also considers through brain is modelled making simple assumptions of
new methods for looking at both structural and functional homogenous attenuation across the brain and head. Some
connectivity in the brain. It explores whether these tech- common applications of SPECT are listed in Table 11.1.
niques – some of which are novel, others have been used for Gamma emitters with half-lives of 6–12 hours can be pro-
several decades – have added to our understanding of patho- duced some distance from the scanner. Because of the long
logical ageing. In particular we will focus on whether any of half-life, only limited numbers of exposures are possible.
the many imaging acquisition and analysis techniques can be The isotopes employed (123I and 99mTc) are relatively large
used as biomarkers to help the clinician with diagnosis, prog- and tend to change the pharmacology of the substituted
nosis or evaluating treatments (Valkanova and Ebmeier, 2014). molecule. The extensive pharmacological development
Although several different image modalities will be work may, therefore, be one of the explanations for the rela-
described, neuroimaging studies of brain function in tively limited number of SPECT-ligands available.
dementia largely fall into two categories: (1) the study of rest-
ing blood flow and (2) measurement of brain changes due to 11.2.2 POSITRON EMISSION
a specific task. This chapter starts with a brief description TOMOGRAPHY (PET)
of methods of emission tomography, functional magnetic
resonance imaging (fMRI) and diffusion tensor imaging PET uses positron emitters to label physiological brain pro-
(DTI) before describing applications in patients. cesses. Positrons are non-stable elementary particles; within
millimetres’ travel they react with an electron to generate
two photons with a defined energy moving at approximately
180° away from each other. The detection of such coinci-
11.2 IMAGING METHODS dence signals with a detector ring makes it possible to iden-
tify spatial as well as intensity information. Collimators are
11.2.1 SINGLE PHOTON EMISSION not required and signal detection is more sensitive. Positron
COMPUTED TOMOGRAPHY (SPECT) emitting nuclei, particularly 11C, are easily incorporated
into biological molecules, without changing their chemical
SPECT uses γ-emitting biologically relevant molecules characteristics. Positron emission is relatively energetic –
injected intravenously to be distributed to the brain. The only short exposure to radiation is possible, the short decay
107
108 Dementia

Table 11.1 Some common applications of SPECT and PET


Tracer Isotope Mode Physiology Clinical Use
HMPAO a 99m Tc-SPECT Perfusion Yes
IMPb 123I-SPECT Perfusion Yes
ECDc 99mTc-SPECT Perfusion Yes
Water 15O-PET Blood flow Rare
Oxygen 15O-PET Oxygen uptake No
FDGd 18F-PET Glucose uptake (aerobic + Yes
anaerobic)
Glucose 11 C-PET Glucose uptake (aerobic) No
Abbreviations: SPECT, single photon emission computed tomography; PET, positron emission tomography.
a Hexamethylpropyleneamine oxime.

b N-isopropyl-(iodine-123)p-iodoamphetamine.

c Ethylene cysteinate dimer.

d Fluoro-deoxyglucose.

half-lives require on-site radiochemistry to incorporate the 11.2.4 BLOOD FLOW MAGNETIC


nuclei into physiologically active compounds. In turn, short RESONANCE IMAGING (ARTERIAL
half-lives allow for repeated application of tracers. Usually SPIN LABELLING)
transmission scans with a γ source in the detector rings are
used to quantify the attenuation across the brain. Arterial spin labelling (ASL) makes it possible to obtain
quantitative information about local tissue blood flow
11.2.3 FUNCTIONAL MAGNETIC (Wong et al., 1997). It is based on the modification of blood
RESONANCE IMAGING magnetization in an artery (e.g. carotid) by magnetically
labelling blood flowing into the brain. Thus, ASL uses, as
fMRI investigates brain function in a non-invasive fash- a ‘tracer’, magnetically labelled water in flowing blood.
ion. It relies on the blood-oxygen-level dependent (BOLD) Blood flowing into the imaging slice exchanges with tissue
contrast to measure haemodynamic signal changes related water, also altering tissue magnetization (Detre et al., 1992).
to neural activity. Oxy- and deoxy-haemoglobin have dif- A ­perfusion-weighted image can be generated by subtract-
ferent ferromagnetic properties and affect a magnetic field ing an image in which inflowing water molecules have been
differently. The relative concentration of oxyhaemoglobin spin labelled from an image without spin labelling. ASL
increases in active brain areas, as blood flow increases more can be used to obtain either a static measurement of rest-
than oxygen extraction. ing blood flow, or can be measured dynamically to obtain a
Repeated examinations are usual, as within each sub- more quantitative fMRI signal. Compared with the BOLD
ject high signal-to-noise ratios can be achieved by repeat- signal, ASL offers certain advantages, such as improved
ing scans many times. Brain activations with well-defined sensitivity to slow changes in neural activity, reduced inter-
BOLD signal have been obtained for functions such as lan- subject variability (Wang et al., 2003) and generally reduced
guage, movement, hearing and memory. Signal changes sensitivity to susceptibility artefacts (Detre and Wang,
may be generated by synaptic activity that can be excitatory 2002). Furthermore, ASL provides a quantitative measure
or inhibitory (Arthurs and Boniface, 2002) and located at of the CBF, which may be more closely related to neuronal
a distance from the site of neuronal body activity. As the activation than the BOLD signal (Miller et al., 2001). It also
BOLD signal is an indirect measure of neuronal activ- allows for a more specific functional localization because
ity greatly dependent on vascular components (i.e. cere- the blood flow signal can be better spatially localized to
bral blood flow [CBF], cerebral blood volume and vascular the site of neuronal activity, the ASL signal reaches the
compliance), brain oxygen metabolism and the coupling activity peak earlier and has less variance in peak latency
between these measures, any change in one of these factors compared with the BOLD signal. Perfusion-related altera-
may affect the magnitude of the signal. While this is not tions have been observed across a variety of neurodegen-
a major problem when BOLD fMRI is used for anatomical erative disorders (Hu et al., 2010), and these changes may
localization and mapping of neuronal activity, quantitative reflect disease-related pathophysiological processes. ASL has
interpretation of the signal in clinical populations is less been less frequently used in research compared with fMRI
straightforward. Baseline perfusion measures and a more because of its low signal-to-noise ratio, poorer temporal
accurate fMRI analysis model may be essential for correctly resolution, mainly due to the pairwise acquisition of label
interpreting group differences (Uludag et al., 2004). FMRI and control images, as well as the required delay time for the
analyses continue to be developed in a research context and tagged blood to flow into imaging slices. It has smaller sig-
are by no means standardized. The clinical use of fMRI in nal changes (typically less than 1%) compared with BOLD,
dementia is therefore an emerging field. and only few established imaging sequences.
Functional brain imaging and connectivity in dementia 109

11.2.5 DIFFUSION TENSOR IMAGING than across it (perpendicular); thus, it has a higher FA than
grey matter (Figure 11.1b). In contrast, grey and white mat-
As described in Chapter 10, most of the research into struc- ter has very similar values of ADC, because the amount of
tural deficits associated with dementia concerns grey mat- water movement is approximately equal (Figure 11.1a).
ter. However, white matter pathology has also been reported Alterations of ADC and FA have been reported in many
in Alzheimer’s disease (AD) and is noted in many other neu- diseases using a variety of methods: a region of interest (ROI)
rodegenerative diseases. DTI is a structural imaging tech- across which mean ADC or FA is averaged can be compared
nique that is used to investigate the integrity of white matter between individuals. This technique is simple, but deter-
pathways in the brain. Diffusion-weighted images measure mining the ROI is subjective, can be difficult to replicate
the movement of water molecules within tissue, and when and information available in other areas of the brain may be
acquired in multiple directions, are sensitized to diffusion in missed. Voxelwise techniques allow comparisons to be made
all directions in the brain. Two quantitative measurements over the whole brain, but multi-subject registration is par-
are typically obtained from DTI data: (1) the total amount of ticularly problematic for FA data because of the variability
diffusion in a voxel variously referred to as the apparent dif- of white matter tracts. Tract-based spatial statistics (TBSS)
fusion coefficient (ADC) or mean diffusivity (MD; to avoid is a conservative voxelwise technique that reduces registra-
confusion with major depression we will use the acronym tion problems by comparing FA or ADC projected to the
ADC for this quantity) and (2) a measurement of the extent centre of each major tract (Smith et al., 2006). Finally, by cal-
to which diffusion is constrained to a particular direction culating the tensor of the diffusion direction (Figure 11.1c),
and is usually called fractional anisotropy (FA). In white reconstructions of white matter tracts can be made (Figure
matter, the orientation of axons and myelin sheaths deter- 11.1d). This tractography is used to define individual tracts
mine that diffusion will be easier along the tract (parallel) of interest for calculating mean FA or ADC.

11.3 BRAIN ACTIVITY PATTERNS USED


(a) (b)
IN CLINICAL PRACTICE

There are no strictly applicable ‘rules’ for diagnostic imag-


ing, as Alzheimer-like pathology can be found in a sub-
stantial proportion of the brains of non-demented patients
(Neuropathology Group, 2001). In addition, the diagnoses
of AD and vascular dementia (VaD) are not mutually exclu-
sive so that mixed patterns are likely to occur in a signifi-
cant proportion of patients. Finally, at an advanced stage,
all dementias tend to involve large portions of the brain,
(c) (d) preferentially (and in all likelihood) association cortex, so
any differential features will disappear.
AD is said to initially be present with posterior cingulate
(Minoshima et al., 1997) or medial temporal (Callen et al.,
2002) reductions in brain activity, but soon after bilateral
posterior temporo-parietal reductions in brain activity also
appear (Holman et al., 1992). In blood flow and glucose
metabolism studies of patients with mild cognitive impair-
ment (MCI), temporo-parietal association areas, posterior
cingulate and hippocampus are associated with a higher
Figure 11.1 (See colour insert.) Diffusion tensor imaging
provides quantitative measures of (a) apparent diffusion risk of progressive cognitive decline (Wolf et al., 2003).
coefficient (ADC) and (b) fractional anisotropy (FA). Brighter During later stages, reductions in prefrontal activity occur,
signal means more average diffusion and more constrained so that some authors have suggested computing a ratio of
diffusion for ADC and FA, respectively. Note that grey and association cortex activity over primary sensory-motor
white matter do not differ in ADC but have very different cortex activity as a diagnostic index for AD (Herholz et al.,
FA values, such that FA provides a map of the myelinated 1999; Herholz, 2014). Frontotemporal dementias (FTDs),
axons of white matter. By calculating the tensor informa- i.e. those that initially present with functional impairment
tion, the direction of diffusion can be determined in each of anterior parts of the brain, have a variety of underlying
voxel (c), from which tracts can be reconstructed (d). Panel
pathologies, from Pick’s disease to AD and other tauopa-
(c) shows the principal diffusion direction in the region
including the genu of the corpus callosum depicted by thies to ­ubiquitin-positive, tau-negative cases. Vascular dis-
the red box in panel (b). In panel (d), a tract of the forceps ease is likely to result in patchy lesions of brain perfusion,
minor (yellow) has been reconstructed by placing a single often asymmetrical in distribution, or localized in ‘water-
‘seed’ voxel (blue) in the genu of the corpus callosum. shed’ regions of the brain.
110 Dementia

Experience of imaging clinical dementia with Lewy bod- sensory-motor cortex for classification, initial metabolic
ies (DLB) patients suggests little difference in perfusion pat- impairment was significantly associated with subsequent
terns from AD, although occipitotemporal changes have clinical deterioration, thus predicting for patients with mild
been described (Ishii et al., 1999; Minoshima et al., 2001). cognitive deficits the progression to AD (Herholz et al., 1999).
In advanced AD, bilateral parieto-temporal perfusion
deficits are reported to be more frequent and severe (Nitrini
et al., 2000) with a reported odds ratio (OR) of 17.0 (95%
11.4 DIAGNOSTIC SENSITIVITY AND confidence interval [CI]: 3.1–94.2) for severe AD (Mini-
SPECIFICITY Mental State Examination [MMSE]: <10) and an OR of 5.2
(95% CI: 1.1–24.4) for moderate AD (MMSE: 10–17).
As described in more detail in Chapter 57, the clinical and Initial studies using ASL and magnetic resonance imag-
pathological diagnoses of dementia are not always congruent. ing (MRI) to measure blood flow in AD and FTD confirm
The question about the true ‘gold standard’ for the evalua- the established emission computed tomography results (Du
tion of imaging studies, therefore, arises: Should functional et al., 2006; Takahashi et al., 2014).
imaging reflect the distribution and severity of brain cell Reduced CBF in AD patients relative to healthy controls
loss or pathological deposits, or should it be representative has been demonstrated with ASL in brain regions typically
of patients’ functional impairment or their symptoms and affected by Alzheimer type pathology, such as precuneus, pos-
signs? Either association would be of interest, but the most terior cingulate, parietal association cortex and inferior tem-
important purpose of imaging is of course the facilitation of poral lobe (Johnson et al., 2005; Alsop et al., 2008). Alsop et al.
effective treatment. Often the predictive validity of imaging also reported significantly greater CBF values in AD patients
methods is not known, but it should be ascertainable using relative to controls in the hippocampus, and more broadly
standard empirical methods. By the time the brain comes to the medial temporal complex, after correcting for grey mat-
post-mortem, a large number of confounding mechanisms ter differences (Alsop et al., 2008). This finding emphasizes
will have intervened, associated with the natural history of the necessity to account for morphological differences when
dementia, additional illness and treatment, and the selection investigating functional or physiological differences in stud-
bias resulting from low uptake of post-mortem examinations. ies involving AD patients. Moreover, it suggests that poten-
tial compensatory or pathological mechanisms may modulate
neural activity in brain regions where atrophy occurs. In
another study comparing AD, MCI and healthy controls, the
11.5 DIAGNOSTIC ACCURACY AD patients showed the lowest CBF values within the poste-
OF FUNCTIONAL IMAGING rior cingulate, precuneus and parietal lobes relative to healthy
CURRENTLY USED IN CLINICAL controls and MCI, whereas the MCI patients had reduced CBF
PRACTICE values in the right inferior parietal lobe compared with the
healthy participants (Xu et al., 2007). There is also some evi-
11.5.1 ALZHEIMER’S DISEASE dence to suggest that metabolic PET is able to distinguish pos-
terior cortical atrophy from AD and DLB (Spehl et al., 2014).
Bilateral temporo-parietal hypo-metabolism has been fre-
quently reported for AD compared with normal volun- 11.5.1.1 Pathologically confirmed studies
teers. A systematic review and meta-analysis of literature
in SPECT and dementia (Dougall et al., 2003) found that A multicentre study of PET in dementia with diagnostic
using data pooled from 27 studies, SPECT successfully verification by 3-year clinical follow-up (Silverman et al.,
discriminated between healthy elderly controls and AD 2001) concluded that regional brain metabolism was a sen-
with a pooled sensitivity of 77% against a pooled specific- sitive indicator of AD. In the same study, PET predicted a
ity of 89%. Additional significant abnormalities have been pathologically confirmed diagnosis of AD with a sensitivity
reported for AD in posterior cingulate (Minoshima et al., of 94% (91 out of 97 subjects) against a specificity of 73%
1997) and hippocampus (Elgh et al., 2002). For very early (30 out of 41 subjects) against other patients presenting with
AD, reductions in posterior cingulate tend to be greater symptoms of dementia. Azzn overall diagnostic accuracy of
than in ­parieto-temporal and frontal association cortices 89% was achieved for a subset of 55 patients who presented
(Minoshima et al., 1997). Entorhinal cortex and hippo- at the time of PET with questionable or mild dementia.
campal metabolic reductions have been successfully used A PET study in a group considered ‘diagnostically
as a classifier in the discrimination of cognitively normal challenging or difficult to characterize by clinical criteria’
controls from MCI (De Santi et al., 2001) with a diagnostic (Hoffman et al., 2000) produced sensitivity and specificity
accuracy of 81% and from AD using the temporal neocortex values of PET against a histological diagnosis at autopsy of
with a diagnostic accuracy of 100% (De Santi et al., 2001). AD of 93% and 63%, respectively. For comparison, clini-
In a prospective longitudinal analysis using the ratio of cal diagnosis of probable AD had sensitivity and specificity
deoxyglucose uptake in association cortex over primary values of 63% and 100%, respectively, concluding that the
Functional brain imaging and connectivity in dementia 111

overall diagnostic accuracy of PET at 82% was better than AD patients with 87% correctly classified (90% sensitiv-
clinical diagnosis at 73%. Two comparable SPECT studies ity, 83% specificity) (Du et al., 2006). Fluorodeoxyglucose-
with pathological verification of AD reported sensitivities of PET (FDG-PET) in behavioural variant FTD (bvFTD) was
86% and 63% (43 dementia patients versus 11 healthy elderly able to identify almost 50% of patients not diagnosed with
controls) against specificities of 73% and 93% (70 demen- structural MRI (Kerklaan et al., 2014).
tia patients versus 85 healthy elderly controls), respectively
(Bonte et al., 1997; Jagust et al., 2001). In a perfusion SPECT
study of 49 patients coming to autopsy, (Bonte et al., 2006) 11.5.4 DEMENTIA WITH LEWY BODIES
found specificity for a diagnosis of AD with and without AND DIFFERENTIAL DIAGNOSIS
Lewy bodies of 89.5%, against a mixed group of patients WITH DEPRESSION
with FTD, and sensitivity of 86.7% (Bonte et al., 2006).
Cerebrospinal fluid (CSF) biomarkers are equally found An autopsy-confirmed PET study of the differential diag-
to be associated with regional metabolic changes (Ceravolo nosis of DLB and AD subjects found occipital metabolic
et al., 2008; Arlt et al., 2009). reductions as a potential ante-mortem marker to distin-
guish DLB from AD, with sensitivity and specificity values
determined at 90% and 80% respectively (Minoshima et al.,
11.5.2 VASCULAR DEMENTIA 2001). SPECT studies have confirmed occipital reduction
Typical findings in VaD are multiple small areas of reduced in perfusion as an indicator of DLB (Lobotesis et al., 2001).
perfusion and metabolism extending over cortical and sub- Medial temporal and cingulate reductions in metabolism
cortical structures and a high diagnostic accuracy has been were significantly more pronounced in AD compared to
reported for the discrimination of probable AD and VaD DLB (Imamura et al., 1997), while the occipital deficit in
using this characteristic metabolic pattern (Mielke and DLB appears irrespective of clinical severity (Okamura et
Heiss, 1998). In addition, frontal lobes including cingulate al., 2001). There is, however, significant overlap between the
and superior frontal gyri have been reported to be more imaging phenotypes of AD and DLB (Chiba et al., 2015).
affected in VaD than in AD (Lee et al., 2001). On the other SPECT has been reported to be a useful tool for the differ-
hand, temporal-parietal brain regions have been said best ential diagnosis of AD and depression, with perfusion deficits
to discriminate AD from VaD with sensitivities reported as in depression lying between those of controls and AD and a
high as 90% and 82% against 80% and 82% specificity respec- reported sensitivity of 52% (against 94% with controls) using
tively (Butler et al., 1995; deFigueiredo et al., 1995). SPECT parieto-occipital perfusion as a marker (Stoppe et al., 1995).
has been reported to differentiate AD from multi-infarct
dementia (MID) in a study with a 77% correct classification 11.5.5 SPECT VERSUS PET
rate, compared with structural MRI which correctly classi-
fied only 50% in the same subject group (Butler et al., 1995). A direct comparison (Herholz et al., 2002) of SPECT and
PET produced an overall significant correlation of abnor-
mal tracer uptake between PET and SPECT across the entire
11.5.3 FRONTOTEMPORAL DEMENTIA brain (r = 0.43) with better correspondence achieved in the
temporo-parietal and posterior cingulate association cor-
Highly significant metabolic abnormalities have been tices. Using quantitative statistical parametric mapping
reported for PET in fronto-temporo-parietal association (SPM), the same study reported that PET discriminated
cortex, limbic area, basal ganglia and thalamus in FTD between healthy volunteers and AD with greater reliabil-
compared with normal volunteers. In particular, bilateral ity than SPECT, since PET was less sensitive to statistical
frontal hypoperfusion is a strong predictor of FTD versus threshold effects. Clinical utility, e.g. predictive validity, of
AD in SPECT, with sensitivity and specificity estimated at FDG-PET appears to be higher than SPECT in small pre-
88% and 79%, respectively (Sjogren et al., 2000). Medial liminary studies (Ishii and Minoshima, 2005), although
temporal lobe reduction has been suggested as a marker opinions differ (Yuan et al., 2009). In a small study directly
to separate AD from FTD (Sjogren et al., 2000). In a MRI comparing F-18-FDG-PET and IMP-SPECT in patients
study using ASL, AD patients had reduced CBF values with DLB, reductions of tracer uptake were found in poste-
relative to FTD patients in posterior brain regions, such rior parietal and to a lesser extend in occipital cortex, with
as posterior cingulate and parietal lobes, whereas FTD PET appearing to be more sensitive to changes (Ishii et al.,
patients had reduced CBF values in frontal regions relative 2004). A recent systematic review concluded that although
to AD patients. The combined use of grey matter atrophy studies suggest superiority of PET over SPECT, the evidence
and perfusion values improved the discrimination between base for this is limited (Davison and O’Brien, 2014). SPECT
FTD patients and healthy controls, yielding a correct clas- studies reported sensitivities of 65%–85% for diagnosing
sification value of 74% (71% sensitivity, 76% specificity). AD and specificities (for other dementias) of 72%–87%, PET
In addition, the combination of grey matter and CBF val- studies sensitivities of 75%–99% for AD and specificities of
ues improved the differential diagnosis between FTD and 71%–93% (Davison and O’Brien, 2014).
112 Dementia

11.5.6 METHODOLOGICAL ISSUES [EEG]) (Raichle et al., 2001; Goldman et al., 2002). The use
FOR ESTABLISHING of rs-fMRI data has been of great interest, particularly in
DIAGNOSTIC TOOLS those with or at risk of developing neurodegenerative disor-
ders (Greicius et al., 2004; Sorg et al., 2007).
PET and SPECT images can be analysed qualitatively using
visual inspection methods or quantitatively using a variety 11.6.1 COGNITIVE ACTIVATION STUDIES
of semi-automated methods (Morbelli et al., 2014; Perani IN DEMENTIA AND PRECLINICAL
et al., 2014). STAGES
For quantitative ROI analysis, the brain is divided into
areas often approximating underlying structural anatomy. Functional activation studies have shown that in mild-to-
Mean values of functional activity are then averaged within moderately severe AD, a cognitive or sensory challenge task
these regions and compared with a data set of controls can cause nearly normal levels of activation in areas that
(Defebvre et al., 1999). More objective methods have been are hypometabolic or hypoperfused at rest. However, with
developed recently, such as an automated multi-ROI pro- increased disease severity the degree of activation declines
gramme 3DSRT (Kobayashi et al., 2008), statistical para- (Devous, 2002). Furthermore, excessive activation may be
metric mapping (Friston et al., 1995), discriminant function required in AD patients who perform a task, or activation
analysis (O’Brien et al., 2001) and neural network analysis of additional brain areas, which are not activated by healthy
(Chan et al., 1994). Three-dimensional stereotactic surface controls (Cardebat et al., 1998). It has been hypothesized that
projection images improve the accuracy of visually detect- functional plasticity may be responsible for such additional
ing AD with PET (Burdette et al., 1996). Finally, a combina- recruitment of new brain regions in patients. Because the
tion of structural MRI or computed tomography (CT) and medial temporal lobe (MTL) complex, and the hippocampus
functional imaging improves the accuracy of diagnostic in particular, is the brain region to show earliest pathologi-
classification (O’Brien et al., 2001). cal signs in AD, and because of its involvement in memory
processes, most fMRI studies of AD patients have used mem-
ory paradigms to investigate disease-related effects on brain
11.6 THE CUTTING EDGE: BRAIN activity. fMRI studies using encoding and retrieval memory
tasks have consistently reported decreased activation in hip-
FUNCTION AND CONNECTIVITY
pocampus and parahippocampus of patients compared with
IN DEMENTIA healthy volunteers (Rombouts et al., 2000). More controver-
sial are reports for other neocortical brain regions, such as
The first cognitive deficit in AD is often associated with epi- prefrontal cortex, where both decreased (Small et al., 1999)
sodic memory loss, which corresponds to early reduction in and increased (Sperling et al., 2003) activation have been
perfusion of the posterior cingulate gyrus and precuneus observed in AD patients. Such increased activation in pre-
(Desgranges et al., 1998). The semantic memory impair- frontal cortex has been interpreted as compensatory reorga-
ment common in early AD and even MCI (Lonie et al., nization (plasticity) or as a dedifferentiation (mass activation)
2009) occurs when neurodegenerative changes extend to process (Prvulovic et al., 2005). Functional abnormalities
the adjacent temporal neocortex. Short-term memory per- have been also observed in brain regions not directly affected
formance is correlated with metabolism in the temporo- by AD in the early stages of the disease, such as sensory motor
parietal association cortex, with the left hemisphere for cortex (Buckner et al., 2000). Other neurodegenerative dis-
verbal and right for spatial memory respectively (Haxby et eases, such as FTD, DLB and VaD, have been less extensively
al., 1990b; Trollor and Valenzuela, 2001). If attention is the studied. An fMRI activation study compared patients with
next domain to be affected, the anterior cingulate is thought early AD and FTD during a working memory task in order
to be involved (Matsuda, 2001). This is consistent with the to identify group-related differences. During performance of
clinical picture – it is thought that attentional impairment the task, there was reduced activation in the frontal, tempo-
is responsible for those problems in activities of daily living ral and cingulate cortices in the FTD group compared with
that occur at an early stage. the AD group. There was, however, the opposite effect in the
Patients often have problems with task performance cerebellum, a region less consistently activated in functional
which makes the interpretation of group differences in working memory imaging studies. This cerebellar activation
imaging studies difficult. This does not apply to resting in FTD may reflect successful working memory specific com-
state fMRI (rs-fMRI) (Fox and Raichle, 2007). During an pensation, since test performance of FTD patients were not
rs-fMRI study, participants lie in the scanner without task different from the AD group (Rombouts et al., 2003).
or stimulation. Brain regions showing a strong connectivity FMRI has been advocated to detect early functional brain
(temporal coherence) are defined as ‘resting state networks’ changes in patients with MCI, where memory symptoms are
(RSNs) and reflect properties of functional brain organiza- still isolated, and before more wide-spread structural atro-
tion. RSNs have been consistently observed across subjects phy observed in AD patients has become manifest. fMRI
(Damoiseaux et al., 2006), sessions (Chen et al., 2008) and studies in MCI subjects have focused on detecting brain
imaging modalities (fMRI, PET, electroencephalography functional abnormalities within the MTL complex using
Functional brain imaging and connectivity in dementia 113

memory-based tasks. Results are less consistent than in AD that this may be due to a loss of myelinated fibres (Tang
studies, showing either increased (Dickerson et al., 2005) or et al., 1997). Studies demonstrating a negative correlation
decreased (Machulda et al., 2003) activation in MTL regions between FA and performance on cognitive tasks lend sup-
compared with healthy subjects. It has been suggested port to the hypothesis that white matter deterioration can
that MCI subjects may show increased MTL activation to underlie cognitive decline.
compensate for incipient structural damage. Accordingly, White matter hyperintensities are common in AD,
where MCI subjects were divided into two groups, those increased ADC and decreased FA have been reported
less impaired and those more impaired in an associative throughout the temporal, frontal and parietal lobes in
memory task, the less impaired group showed increased patients with AD (Chua et al., 2008), and several authors
activation, whereas the more impaired group showed hip- have related white matter deficits to neuropsychological
pocampal decreased activation, compared with a group of measures of cognitive decline (Bozzali et al., 2002). Reduced
healthy subjects (Celone et al., 2006). FA has also been reported in patients with early AD (Choi et
al., 2005), MCI (Rose et al., 2006) and in healthy carriers of
the APOE ε4 allele (Persson et al., 2006). Although reduc-
11.6.2 RESTING FMRI STUDIES IN tions in FA and increases in ADC are not specific to AD,
DEMENTIA AND PRECLINICAL these studies suggest that white matter degradation begins
STAGES early in the disease process.
Although white matter abnormalities have been
The value of rs-fMRI has been examined in several clinical
described in many areas of the brain in AD, there is some
studies (Greicius et al., 2004; Greicius et al., 2007; Sorg et al.,
suggestion that the regional pattern of impairment may
2007). The most commonly investigated RSN, the ‘default
distinguish AD from other dementias; for a recent review
mode network’ (DMN) (Raichle et al., 2001), includes brain
see Suri et al. (2014). Zarei et al. (2009) used TBSS to distin-
regions, such as prefrontal cortex, anterior and posterior
guish AD from VaD. Relative to controls, patients with AD
cingulate and lateral parietal extending to medial temporal
had reduced FA in the temporal lobe, whereas patients with
cortex, that overlap with amyloid plaques and grey matter
VaD had more widespread reductions in FA in frontal and
atrophy (Sperling et al., 2009). Reduced functional con-
parietal areas. They identified a small region in the genu of
nectivity in the DMN has been reported in AD relative to
the corpus callosum that was significantly more impaired
healthy controls, and is associated with faster disease pro-
in patients with VaD relative to those with AD. Zhang
gression (Raichle et al., 2001; Greicius et al., 2004; Zhang
et al. (2011) reported that patients with FTD have more
et al., 2010; Binnewijzend et al., 2012; Zamboni et al., 2013).
widespread abnormalities of white matter than patients
Reduced DMN connectivity has also been observed in
with AD. Like the patients with VaD in the previous study,
MCI (Sorg et al., 2007), unaffected relatives in familial AD
patients with FTD have significantly lower FA and higher
(Chhatwal et al., 2013) and volunteers with memory com-
plaints (Wang et al., 2013). Moreover, a disproportionate ADC in the genu of the corpus callosum (Zhang et al.,
reduction in DMN connectivity with advancing age has 2009), as well as in the uncinate fasciculus (Mahoney et al.,
been found in subjects carrying the apoprotein E (APOE) 2014). These studies suggest that there are more extensive
ε4 allele, reflecting the increased vulnerability of ε4 carriers impairments of white matter integrity in patients with VaD
to late-life pathology and cognitive decline (Sheline et al., and FTD relative to those with AD. There is also evidence
2010; Machulda et al., 2011). for regional differences in white matter microstructure
Reduced connectivity in the ‘salience’ network has been between AD and DLB patients. Using a tractography-based
consistently observed in FTD patients (Whitwell et al., 2011b; approach, Kiuchi et al. (2011) found that despite similar
Filippi et al., 2013). This network includes brain regions severity of dementia, DLB patients show greater reduc-
such as the dorsal anterior cingulate and the insular cortices tions in FA in temporo-occipital projection tracts relative
bilaterally and it is largely involved in guiding behaviourally to healthy controls than patients with AD. Damage to these
salient events (Seeley et al., 2007). Reduced connectivity in tracts may underlie the visual deficits commonly observed
a network including motor regions, basal ganglia nuclei and in DLB patients.
cerebellar areas has been observed in patients with pro-
gressive supranuclear palsy (PSP) (Whitwell et al., 2011a; 11.6.4 LONGITUDINAL STUDIES
Gardner et al., 2013), which is clinically characterized by
A number of functional imaging studies have investigated
motor symptoms and cognitive impairment.
cognitive functioning over time. These have shown that
patients with more severe perfusion or metabolic deficits
11.6.3 STRUCTURAL CONNECTIVITY IN in the temporo-parietal cortex at initial evaluation show
AD AND OTHER DEMENTIAS a more rapid cognitive decline over time (Devous, 2002).
With regard to MCI, significant bilateral regional CBF
Decreased white matter integrity (increases in MD/ (rCBF) decreases are seen in the posterior cingulate, pari-
ADC and decreases in FA) are a feature of normal ageing etal and precuneus regions of those who later meet criteria
(Charlton et al., 2006), and histopathological studies show for AD. Subsequently, at the stage of a clinical diagnosis of
114 Dementia

AD, additional rCBF abnormalities are seen in the hippo- cohorts. If the introduction of a new imaging method is to
campus and parahippocampus (Minoshima et al., 1997). be evidence-based in a National Health Service and good
For individual patients, the particular neuropsychologi- evidence is only available after the establishment of a ser-
cal pattern that develops during the progression of AD can vice, the creation of pilot services is necessary that are not
be predicted by early metabolic asymmetries in the asso- technology driven, i.e. that are centred in a normal clini-
ciation cortices (Haxby et al., 1990a). This would indicate cal set-up that will generate results transferable to the ser-
that there is a functional reserve in the brain, so that neu- vice in general (Elias et al., 2014). The theoretical appeal of
ropsychological dysfunction is likely to follow rather than imaging methods, after all we believe that the dementias
co-occur with rCBF changes. are brain diseases, holds out the promise of visualizing
More recently, longitudinal functional imaging studies brain changes that are relevant to illness outcome and
have been proposed to assess the response to treatments for treatment response.
AD (Alexander et al., 2002). Such activation studies are of
particular interest, as they can help in identifying poten-
tially reversible tissue damage. REFERENCES
Conversion from MCI to AD can be predicted with
functional scans (Drzezga et al., 2005; Borroni et al., 2006; Alexander, G.E., Chen, K., Pietrini, P. et al. (2002).
Nobili et al., 2009), and hypometabolism is even found in Longitudinal PET evaluation of cerebral metabolic
cognitively normal subjects at increased risk of AD (Reiman decline in dementia: A potential outcome measure
et al., 2004; Mosconi et al., 2007). in Alzheimer’s disease treatment studies. American
Journal of Psychiatry, 159: 738–745.
Alsop, D.C., Casement, M., de Bazelaire, C. et al. (2008).
11.7 CONCLUSIONS FOR THE Hippocampal hyperperfusion in Alzheimer’s disease.
CLINICIAN Neuroimage, 42: 1267–1274.
Arlt, S., Brassen, S., Jahn, H. et al. (2009). Association
One of the authors’ central tenets is that the ‘gold stan- between FDG uptake, CSF biomarkers and cognitive
dard’ of all imaging work has to be the patients’ clinical performance in patients with probable Alzheimer’s
state. By implication, ‘predictive validity’ is the concept of disease. European Journal of Nuclear Medicine and
greatest interest: imaging results should predict patients’ Molecular Imaging, 36: 1090–1100.
prognosis and in particular whether they are going to Arthurs, O.J. and Boniface, S. (2002). How well do we
respond to a treatment. This does not contradict the basic understand the neural origins of the fMRI BOLD sig-
notion that diagnosis has to be at the beginning of effec- nal? Trends in Neuroscience, 25: 27–31.
tive treatment, but it reflects the observation that brain Binnewijzend, M.A., Schoonheim, M.M., Sanz-Arigita,
pathological changes are not always specific to a particu- E. et al. (2012). Resting-state fMRI changes in
lar illness; that there is a substantial overlap of pathologi- Alzheimer’s disease and mild cognitive impairment.
cal changes even with health, and that pathological illness Neurobiology of Aging, 33: 2018–2028.
‘entities’ are likely to be caused by a varying admixture Bonte, F.J., Harris, T.S., Hynan, L.S. et al. (2006). Tc-99m
of genotype–environment interactions: AD is really a HMPAO SPECT in the differential diagnosis of the
collective term for a number of conditions resulting in dementias with histopathologic confirmation. Clinical
dementia, plaques and tangles. Histological diagnoses Nuclear Medicine, 31: 376–378.
merely reflect recognizable final common pathways for Bonte, F.J., Weiner, M.F., Bigio, E.H. and White, C.L., 3rd
a collection of pathological conditions and therefore do (1997). Brain blood flow in the dementias: SPECT with
not necessarily predict treatment response or prognosis. histopathologic correlation in 54 patients. Radiology,
In evaluating the usefulness of imaging methods, direct 202: 793–797.
comparisons should be made with clinical outcome mea- Borroni, B., Anchisi, D., Paghera, B. et al. (2006).
sures – histopathological follow-through studies are of Combined 99mTc-ECD SPECT and neuropsychological
interest, but will always be confounded by selection bias, studies in MCI for the assessment of conversion to AD.
intervening illness and other artefacts. The utility of any Neurobiology of Aging, 27: 24–31.
imaging biomarker depends on its specificity and sen- Bozzali, M., Falini, A., Franceschi, M. et al. (2002). White
sitivity for differential diagnosis, their ability to detect matter damage in Alzheimer’s disease assessed in vivo
disease early or their response to treatment (Dubois et using diffusion tensor magnetic resonance imaging.
al., 2014). Although most imaging modalities described Journal of Neurology, Neurosurgery and Psychiatry, 72:
here have clinical relevance, it is likely that the individual 742–746.
measurements will be most useful in combination. One Buckner, R.L., Snyder, A.Z., Sanders, A.L. et al. (2000).
of the binds we encounter is that useful data on sensitiv- Functional brain imaging of young, nondemented,
ity and specificity, and more particularly positive and and demented older adults. Journal of Cognitive
negative predictive value, require representative clinical Neuroscience, 12 (Suppl. 2): 24–34.
Functional brain imaging and connectivity in dementia 115

Burdette, J.H., Minoshima, S., Vander Borght, T. et al. Davison, C.M. and O’Brien, J.T. (2014). A comparison
(1996). Alzheimer disease: Improved visual interpreta- of FDG-PET and blood flow SPECT in the diagno-
tion of PET images by using three-dimensional stereo- sis of neurodegenerative dementias: A systematic
taxic surface projections. Radiology, 198: 837–843. review. International Journal of Geriatric Psychiatry,
Butler, R.E., Costa, D.C., Greco, A. et al. (1995). 29: 551–561.
Differentiation between Alzheimer’s disease and multi- De Santi, S., de Leon, M.J., Rusinek, H. et al. (2001).
infarct dementia: Spect vs. MR imaging. International Hippocampal formation glucose metabolism and
Journal of Geriatric Psychiatry, 10: 121–128. volume losses in MCI and AD. Neurobiology of Aging,
Callen, D.J., Black, S.E. and Caldwell, C.B. (2002). Limbic 22: 529–539.
system perfusion in Alzheimer’s disease measured by Defebvre, L.J., Leduc, V., Duhamel, A. et al. (1999).
MRI-coregistered HMPAO SPET. European Journal of Technetium HMPAO SPECT study in dementia with
Nuclear Medicine and Molecular Imaging, 29: 899–906. Lewy bodies, Alzheimer’s disease and idiopathic
Cardebat, D., Demonet, J.F., Puel, M. et al. (1998). Brain cor- Parkinson’s disease. Journal of Nuclear Medicine,
relates of memory processes in patients with dementia 40: 956–962.
of Alzheimer’s type: A SPECT activation study. Journal deFigueiredo, R.J., Shankle, W.R., Maccato, A. et al.
of Cerebral Blood Flow Metabolism, 18: 457–462. (1995). Neural-network-based classification of cog-
Celone, K.A., Calhoun, V.D., Dickerson, B.C. et al. (2006). nitively normal, demented, Alzheimer disease and
Alterations in memory networks in mild cognitive impair- vascular dementia from single photon emission
ment and Alzheimer’s disease: An independent compo- with computed tomography image data from brain.
nent analysis. Journal of Neuroscience, 26: 10222–10231. Proceedings of National Academy Sciences of the
Ceravolo, R., Borghetti, D., Kiferle, L. et al. (2008). CSF United States of America, 92: 5530–5534.
phosporylated TAU protein levels correlate with cerebral Desgranges, B., Baron, J.C. and Eustache, F. (1998). The
glucose metabolism assessed with PET in Alzheimer’s functional neuroanatomy of episodic memory: The role
disease. Brain Research Bulletin, 76: 80–84. of the frontal lobes, the hippocampal formation, and
Chan, K.H., Johnson, K.A., Becker, J.A. et al. (1994). other areas. Neuroimage, 8: 198–213.
A neural network classifier for cerebral perfusion Detre, J.A., Leigh, J.S., Williams, D.S. and Koretsky, A.P.
­imaging. Journal of Nuclear Medicine, 35: 771–774. (1992). Perfusion imaging. Magnetic Resonance in
Charlton, R.A., Barrick, T.R., McIntyre, D.J. et al. (2006). Medicine, 23: 37–45.
White matter damage on diffusion tensor imaging cor- Detre, J A. and Wang, J. (2002). Technical aspects
relates with age-related cognitive decline. Neurology, and utility of fMRI using BOLD and ASL. Clinical
66: 217–222. Neurophysiology, 113: 621–634.
Chen, S., Ross, T.J., Zhan, W. et al. (2008). Group inde- Devous, M.D., Sr. (2002). Functional brain imaging in
pendent component analysis reveals consistent the dementias: Role in early detection, differential
resting-state networks across multiple sessions. Brain diagnosis, and longitudinal studies. European Journal
Research, 1239: 141–151. of Nuclear Medicine and Molecular Imaging, 29:
Chhatwal, J.P., Schultz, A.P., Johnson, K. et al. (2013). 1685–1696.
Impaired default network functional connectivity in Dickerson, B.C., Salat, D.H., Greve, D.N. et al. (2005).
autosomal dominant Alzheimer disease. Neurology, Increased hippocampal activation in mild cogni-
81: 736–744. tive impairment compared to normal aging and AD.
Chiba, Y., Fujishiro, H., Ota, K. et al. (2015). Clinical pro- Neurology, 65: 404–411.
files of dementia with Lewy bodies with and without Dougall, N., Bruggink, S. and Ebmeier, K.P. (2003). Clinical
Alzheimer’s disease-like hypometabolism. International use of SPECT in dementia – A quantitative review.
Journal of Geriatric Psychiatry, 30: 316–323. In K.P. Ebmeier (ed.), SPECT in Dementia. Basel,
Choi, S.J., Lim, K.O., Monteiro, I. and Reisberg, B. (2005). Switzerland: Karger Verlag. pp. 4–37.
Diffusion tensor imaging of frontal white matter Drzezga, A., Grimmer, T., Riemenschneider, M. et al.
­microstructure in early Alzheimer’s disease: A prelimi- (2005). Prediction of individual clinical outcome in MCI
nary study. Journal of Geriatric Psychiatry Neurology, by means of genetic assessment and (18)F-FDG PET.
18: 12–19. Journal of Nuclear Medicine, 46: 1625–1632.
Chua, T.C., Wen, W., Slavin, M.J. and Sachdev, P.S. (2008). Du, A.T., Jahng, G.H., Hayasaka, S. et al. (2006).
Diffusion tensor imaging in mild cognitive impairment Hypoperfusion in frontotemporal dementia and
and Alzheimer’s disease: A review. Current Opinion in Alzheimer disease by arterial spin labeling MRI.
Neurology, 21: 83–92. Neurology, 67, 1215–1220.
Damoiseaux, J.S., Rombouts, S.A., Barkhof, F. et al. (2006). Dubois, B., Feldman, H.H., Jacova, C. et al. (2014).
Consistent resting-state networks across healthy sub- Advancing research diagnostic criteria for Alzheimer’s
jects. Proceedings of the National Academy of Science disease: The IWG-2 criteria. Lancet Neurology, 13:
of the United States of America, 103, 13848–13853. 614–629.
116 Dementia

Elgh, E., Sundstrom, T., Nasman, B., Ahlstrom, R. and Hoffman, J.M., Welsh-Bohmer, K.A., Hanson, M. et al.
Nyberg, L. (2002). Memory functions and rCBF (2000). FDG PET imaging in patients with pathologi-
(99m)Tc-HMPAO SPET: Developing diagnostics in cally verified dementia. Journal of Nuclear Medicine,
Alzheimer’s disease. European Journal of Nuclear 41: 1920–1928.
Medicine and Molecular Imaging, 29: 1140–1148. Holman, B.L., Johnson, K.A., Gerada, B. et al. (1992).
Elias, A., Woodward, M. and, Rowe, C.C. (2014). The scintigraphic appearance of Alzheimer’s disease:
Management impact of FDG-PET in dementia: Results A prospective study using technetium-99m-HMPAO
from a tertiary center memory clinic. Journal of SPECT. Journal of Nuclear Medicine, 33: 181–185.
Alzheimer’s Disease, 42: 885–892. Hu, W.T., Wang, Z., Lee, V.M. et al. (2010). Distinct cere-
Filippi, M., Agosta, F., Scola, E. et al. (2013). Functional bral perfusion patterns in FTLD and AD. Neurology,
network connectivity in the behavioral variant of fron- 75: 881–888.
totemporal dementia. Cortex, 49: 2389–2401. Imamura, T., Ishii, K., Sasaki, M. et al. (1997). Regional
Fox, M.D. and Raichle, M.E. (2007). Spontaneous fluc- cerebral glucose metabolism in dementia with Lewy
tuations in brain activity observed with functional bodies and Alzheimer’s disease: A comparative study
magnetic resonance imaging. Nature Reviews using positron emission tomography. Neuroscience
Neuroscience, 8: 700–711. Letters, 235: 49–52.
Friston, K.J., Holmes, A.P., Worsley, K.J. et al. (1995). Ishii, K., Hosaka, K., Mori, T. and Mori, E. (2004).
Statistical parametric maps in functional imaging: A gen- Comparison of FDG-PET and IMP-SPECT in patients
eral linea approach. Human Brain Mapping, 2: 189–210. with dementia with Lewy bodies. Annals of Nuclear
Gardner, R.C., Boxer, A.L., Trujillo, A. et al. (2013). Intrinsic Medicine, 18: 447–451.
connectivity network disruption in progressive supra- Ishii, K. and Minoshima, S. (2005). PET is better than
nuclear palsy. Annals of Neurology, 73: 603–616. perfusion SPECT for early diagnosis of Alzheimer’s
Goldman, R.I., Stern, J.M., Engel, J., Jr. and Cohen, M.S. disease. European Journal of Nuclear Medicine and
(2002). Simultaneous EEG and fMRI of the alpha Molecular Imaging, 32: 1463–1465.
rhythm. Neuroreport, 13: 2487–2492. Ishii, K., Yamaji, S., Kitagaki, H. et al. (1999). Regional
Greicius, M.D., Flores, B.H., Menon, V. et al. (2007). cerebral blood flow difference between dementia with
Resting-state functional connectivity in major depres- Lewy bodies and AD. Neurology, 53: 413–416.
sion: Abnormally increased contributions from sub- Jagust, W., Thisted, R., Devous, M.D., Sr. et al. (2001).
genual cingulate cortex and thalamus. Biological SPECT perfusion imaging in the diagnosis of
Psychiatry, 62: 429–437. Alzheimer’s disease: A clinical-pathologic study.
Greicius, M.D., Srivastava, G., Reiss, A.L. and Menon, Neurology, 56: 950–956.
V. (2004). Default-mode network activity distinguishes Johnson, N.A., Jahng, G.H., Weiner, M.W. et al. (2005).
Alzheimer’s disease from healthy aging: Evidence Pattern of cerebral hypoperfusion in Alzheimer dis-
from functional MRI. Proceedings of the National ease and mild cognitive impairment measured with
Academy Sciences of the United States of America, arterial spin-labeling MR imaging: Initial experience.
101: 4637–4642. Radiology, 234, 851–859.
Haxby, J.V., Grady, C.L., Koss, E. et al. (1990a). Kerklaan, B.J., van Berckel, B.N., Herholz, K. et al. (2014).
Longitudinal-study of cerebral metabolic asymme- The added value of 18-fluorodeoxyglucose-positron
tries and associated neuropsychological patterns in emission tomography in the diagnosis of the behavioral
early dementia of the Alzheimer type. Archives of variant of frontotemporal dementia. Amercian Journal of
Neurology, 47: 753–760. Alzheimers Disease and Other Dementias, 29: 607–613.
Haxby, J.V., Grady, C.L., Koss, E. et al. (1990b). Kiuchi, K., Morikawa, M., Taoka, T. et al. (2011). White
Longitudinal study of cerebral metabolic asymme- matter changes in dementia with Lewy bodies and
tries and associated neuropsychological patterns in Alzheimer’s disease: A tractography-based study.
early dementia of the Alzheimer type. Archives of Journal Psychiatric Research, 45: 1095–1100.
Neurology, 47: 753–760. Kobayashi, S., Tateno, M., Utsumi, K. et al. (2008).
Herholz, K. (2014). Guidance for reading FDG PET scans Quantitative analysis of brain perfusion SPECT in
in dementia patients. Quarterly Journal of Nuclear Alzheimer’s disease using a fully automated regional
Medicine and Molecular Imaging, 58 (4): 332–343. cerebral blood flow quantification software, 3DSRT.
Herholz, K., Nordberg, A., Salmon, E. et al. (1999). Journal of Neurological Sciecnes, 264, 27–33.
Impairment of neocortical metabolism predicts Lee, B.F., Liu, C.K., Tai, C.T. et al. (2001). Alzheimer’s
progression in Alzheimer’s disease. Dementia and disease: Scintigraphic appearance of Tc-99m HMPAO
Geriatric Cognitive Disorders, 10: 494–504. brain spect. Kaohsiung Journal of Medical Sciences,
Herholz, K., Schopphoff, H., Schmidt, M. et al. (2002). 17: 394–400.
Direct comparison of spatially normalized PET and Lobotesis, K., Fenwick, J.D., Phipps, A. et al. (2001).
SPECT scans in Alzheimer’s disease. Journal of Nuclear Occipital hypoperfusion on SPECT in dementia with
Medicine, 43: 21–26. Lewy bodies but not AD. Neurology, 56, 643–649.
Functional brain imaging and connectivity in dementia 117

Lonie, J.A., Herrmann, L.L., Tierney, K.M. et al. (2009). O’Brien, J.T., Ames, D., Desmond, P. et al. (2001).
Lexical and semantic fluency discrepancy scores Combined magnetic resonance imaging and
in aMCI and early Alzheimer’s disease. Journal of ­single-photon emission tomography scanning in
Neuropsychology, 3: 79–92. the d­ iscrimination of Alzheimer’s disease from
Machulda, M.M., Jones, D.T., Vemuri, P. et al. (2011). Effect ­age-matched controls. International Psychogeriatrics,
of APOE epsilon4 status on intrinsic network connec- 13: 149–161.
tivity in cognitively normal elderly subjects. Archives of Okamura, N., Arai, H., Higuchi, M. et al. (2001).
Neurology, 68: 1131–1136. [18F]FDG-PET study in dementia with Lewy
Machulda, M.M., Ward, H.A., Borowski, B. et al. (2003). bodies and Alzheimer’s disease. Progress
Comparison of memory fMRI response among nor- Neuropsychopharmacology Biological Psychiatry, 25:
mal, MCI, and Alzheimer’s patients. Neurology, 61, 447–456.
500–506. Perani, D., Della Rosa, P.A., Cerami, C. et al. (2014).
Mahoney, C.J., Ridgway, G.R., Malone, I.B. et al. (2014). Validation of an optimized SPM procedure for FDG-
Profiles of white matter tract pathology in frontotem- PET in dementia diagnosis in a clinical setting.
poral dementia. Human Brain Mapping, 35: 4163–4179. Neuroimage Clinical, 6: 445–454.
Matsuda, H. (2001). Cerebral blood flow and metabolic Persson, J., Lind, J., Larsson, A. et al. (2006). Altered
abnormalities in Alzheimer’s disease. Annals of brain white matter integrity in healthy carriers of the
Nuclear Medicine, 15: 85–92. APOE epsilon4 allele: A risk for AD? Neurology, 66:
Mielke, R. and Heiss, W.D. (1998). Positron emission 1029–1033.
tomography for diagnosis of Alzheimer’s disease and Prvulovic, D., Van de Ven, V., Sack, A.T. et al. (2005).
vascular dementia. Journal of Neural Transmission Functional activation imaging in aging and dementia.
Supplementa, 53: 237–250. Psychiatry Research, 140: 97–113.
Miller, K.L., Luh, W.M., Liu, T.T. et al. (2001). Nonlinear Raichle, M.E., MacLeod, A.M., Snyder, A.Z. et al. (2001).
temporal dynamics of the cerebral blood flow A default mode of brain function. Proceedings of the
response. Human Brain Mapping, 13: 1–12. National Academy Sciences of the United States of
Minoshima, S., Foster, N.L., Sima, A.A. et al. (2001). America, 98: 676–682.
Alzheimer’s disease versus dementia with Lewy bod- Reiman, E.M., Chen, K., Alexander, G.E. et al. (2004).
ies: Cerebral metabolic distinction with autopsy confir- Functional brain abnormalities in young adults at
mation. Annals of Neurology, 50: 358–365. genetic risk for late-onset Alzheimer’s dementia.
Minoshima, S., Giordani, B., Berent, S. et al. (1997). Proceedings of the National Academy Sciences of the
Metabolic reduction in the posterior cingulate cortex United States of America, 101: 284–289.
in very early Alzheimer’s disease. Annals of Neurology, Rombouts, S.A., Barkhof, F., Veltman, D.J. et al. (2000).
42: 85–94. Functional MR imaging in Alzheimer’s disease
Morbelli, S., Brugnolo, A., Bossert, I. et al. (2014). Visual during memory encoding. American Journal of
versus semi-quantitative analysis of 18F-FDG-PET Neuroradiology, 21: 1869–1875.
in amnestic MCI: An European Alzheimer’s Disease Rombouts, S.A., van Swieten, J.C., Pijnenburg, Y.A. et al.
Consortium (EADC) project. Journal of Alzheimers (2003). Loss of frontal fMRI activation in early fronto-
Disease, 44: 815–826. temporal dementia compared to early AD. Neurology,
Mosconi, L., Brys, M., Switalski, R. et al. (2007). Maternal 60: 1904–1908.
family history of Alzheimer’s disease predisposes to Rose, S.E., McMahon, K.L., Janke, A.L. et al. (2006).
reduced brain glucose metabolism. Proceedings of the Diffusion indices on magnetic resonance imaging and
National Academy of Sciences of the United States of neuropsychological performance in amnestic mild cog-
America, 104: 19067–19072. nitive impairment. Journal of Neurology, Neurosurgery
Neuropathology Group (2001). Pathological cor- and Psychiatry, 77: 1122–1128.
relates of late-onset dementia in a multicentre, Seeley, W.W., Menon, V., Schatzberg, A.F. et al. (2007).
community-based population in England and Wales. Dissociable intrinsic connectivity networks for
Neuropathology Group of the Medical Research salience processing and executive control. Journal of
Council Cognitive Function and Ageing Study (MRC Neuroscience, 27: 2349–2356.
CFAS). Lancet, 357: 169–175. Sheline, Y.I., Morris, J.C., Snyder, A.Z. et al. (2010).
Nitrini, R., Buchpiguel, C.A., Caramelli, P. et al. (2000). APOE4 allele disrupts resting state fMRI connectiv-
SPECT in Alzheimer’s disease: Features associated ity in the absence of amyloid plaques or decreased
with bilateral parietotemporal hypoperfusion. Acta CSF Abeta42. Journal of Neuroscience, 30:
Neurologica Scandinavica, 101: 172–176. 17035–17040.
Nobili, F., De Carli, F., Frisoni, G.B. et al. (2009). SPECT Silverman, D.H., Small, G.W., Chang, C.Y. et al. (2001).
predictors of cognitive decline and Alzheimer’s Positron emission tomography in evaluation of demen-
disease in mild cognitive impairment. Journal of tia: Regional brain metabolism and long-term out-
Alzheimers Disease, 17: 761–772. come. JAMA, 286: 2120–2127.
118 Dementia

Sjogren, M., Gustafson, L., Wikkelso, C. and Wallin, Valkanova, V. and Ebmeier, K.P. (2014). Neuroimaging in
A. (2000). Frontotemporal dementia can be distin- dementia. Maturitas, 79: 202–208.
guished from Alzheimer’s disease and subcortical Wang, J., Aguirre, G.K., Kimberg, D.Y. et al. (2003).
white matter dementia by an anterior-to-posterior Arterial spin labeling perfusion fMRI with very low
rCBF-SPET ratio. Dementia Geriatric Cognitive task frequency. Magnetic Resonance in Medicine,
Disorders, 11: 275–285. 49: 796–802.
Small, S.A., Perera, G.M., DeLaPaz, R. et al. (1999). Wang, Y., Risacher, S.L., West, J.D. et al. (2013). Altered
Differential regional dysfunction of the hippocampal default mode network connectivity in older adults
formation among elderly with memory decline and with cognitive complaints and amnestic mild cogni-
Alzheimer’s disease. Annals of Neurology, 45: 466–472. tive impairment. Journal of Alzheimers Disease, 35:
Smith, S.M., Jenkinson, M., Johansen-Berg, H. et al. (2006). 751–760.
Tract-based spatial statistics: Voxelwise analysis of multi- Whitwell, J.L., Avula, R., Master, A. et al. (2011a).
subject diffusion data. Neuroimage, 31: 1487–1505. Disrupted thalamocortical connectivity in PSP:
Sorg, C., Riedl, V., Muhlau, M. et al. (2007). Selective A ­resting-state fMRI, DTI, and VBM study.
changes of resting-state networks in individuals at risk Parkinsonism Related Disorders, 17: 599–605.
for Alzheimer’s disease. Proceedings of the National Whitwell, J.L., Josephs, K.A., Avula, R. et al. (2011b). Altered
Academy Sciences of the United States of America, functional connectivity in asymptomatic MAPT subjects:
104: 18760–18765. A comparison to bvFTD. Neurology, 77: 866–874.
Spehl, T.S., Hellwig, S., Amtage, F. et al. (2014). Syndrome- Wolf, H., Jelic, V., Gertz, H.J. et al. (2003). A critical
specific patterns of regional cerebral glucose metabo- discussion of the role of neuroimaging in mild cog-
lism in posterior cortical atrophy in comparison to nitive impairment. Acta Neurologica Scandinavica
dementia with Lewy bodies and Alzheimer’s disease-A Supplementum, 179: S52–S76.
[F-18]-Fdg pet study. Journal of Neuroimaging, 25(2): Wong, E.C., Buxton, R.B. and Frank, L.R. (1997).
281–288. Implementation of quantitative perfusion imaging
Sperling, R.A., Bates, J.F., Chua, E.F.et al. (2003). fMRI stud- techniques for functional brain mapping using pulsed
ies of associative encoding in young and elderly arterial spin labeling. NMR Biomedicine, 10: 237–249.
controls and mild Alzheimer’s disease. Journal of Xu, G., Antuono, P.G., Jones, J. et al. (2007). Perfusion
Neurology, Neurosurgery and Psychiatry, 74: 44–50. fMRI detects deficits in regional CBF during
Sperling, R.A., Laviolette, P.S., O’Keefe, K. et al. (2009). ­memory-encoding tasks in MCI subjects. Neurology,
Amyloid deposition is associated with impaired default 69: 1650–1656.
network function in older persons without dementia. Yuan, Y., Gu, Z.X. and Wei, W.S. (2009).
Neuron, 63: 178–188. Fluorodeoxyglucose-positron-emission tomography,
Stoppe, G., Staedt, J., Kogler, A. et al. (1995). 99mTc- single-photon emission tomography, and structural
HMPAO-SPECT in the diagnosis of senile dementia MR imaging for prediction of rapid conversion to
of Alzheimer’s type – A study under clinical routine Alzheimer disease in patients with mild cognitive
conditions. Journal of Neural Transmission General impairment: A meta-analysis. American Journal of
Section, 99: 195–211. Neuroradiology, 30: 404–410.
Suri, S., Topiwala, A., Mackay, C.E. et al. (2014). Using Zamboni, G., Wilcock, G.K., Douaud, G. et al. (2013).
structural and diffusion magnetic resonance imaging Resting functional connectivity reveals residual
to differentiate the dementias. Current Neurology and functional activity in Alzheimer’s disease. Biological
Neuroscience Reports, 14: 475. Psychiatry, 74: 375–383.
Takahashi, H., Ishii, K., Hosokawa, C. et al. (2014). Clinical Zarei, M., Damoiseaux, J.S., Morgese, C. et al. (2009).
application of 3D arterial spin-labeled brain perfusion Regional white matter integrity differentiates between
imaging for Alzheimer disease: Comparison with brain vascular dementia and Alzheimer disease. Stroke, 40:
perfusion SPECT. American Journal of Neuroradiology, 773–779.
35: 906–911. Zhang, Y., Schuff, N., Ching, C. et al. (2011). Joint assess-
Tang, Y., Nyengaard, J.R., Pakkenberg, B. and Gundersen, ment of structural, perfusion, and diffusion MRI in
H.J. (1997). Age-induced white matter changes Alzheimer’s disease and frontotemporal dementia.
in the human brain: A stereological investigation. International Journal of Alzheimers Disease, 2011:
Neurobiology of Aging, 18: 609–615. 546871.
Trollor, J.N. and Valenzuela, M.J. (2001). Brain ageing in Zhang, Y., Schuff, N., Du, A.T. et al. (2009). White matter
the new millennium. Australian New Zealand Journal of damage in frontotemporal dementia and Alzheimer’s
Psychiatry, 35: 788–805. disease measured by diffusion MRI. Brain, 132,
Uludag, K., Dubowitz, D.J., Yoder, E.J. et al. (2004). 2579–2592.
Coupling of cerebral blood flow and oxygen consump- Zhang, H.Y., Wang, S.J., Liu, B. et al. (2010). Resting
tion during physiological activation and deactivation brain connectivity: Changes during the progress of
measured with fMRI. Neuroimage, 23: 148–155. Alzheimer disease. Radiology, 256: 598–606.
12
Molecular brain imaging in dementia

VICTOR L. VILLEMAGNE AND CHRISTOPHER C. ROWE

applied early. So early detection of the underlying patho-


12.1 MOLECULAR IMAGING IN logical process is important. Since Aβ plaques and tau
DEMENTIA ­neurofibrillary tangles (NFT) are the hallmark brain lesions
at the centre of AD pathogenesis, several pharmacological
Molecular neuroimaging techniques such as positron agents aimed at reducing Aβ and tau levels in the brain
­emission tomography (PET) have been used for the in vivo are being developed and tested, many efforts have focused
assessment of molecular processes at their sites of action, on generating radiotracers that allow Aβ and tau imaging
permitting detection of subtle pathophysiological changes in vivo and are suited to widespread clinical application.
in the brain at asymptomatic stages, when there is no evi- For a radiotracer to be useful as a neuroimaging Aβ
dence of anatomic changes on computed tomography (CT) probe, a number of key general properties are desirable: they
or magnetic resonance imaging (MRI). The development should be lipophilic molecules that cross the blood–brain
of molecular imaging methods for non-invasively assess- barrier, preferably not to be metabolized, while reversibly
ing disease-specific traits such as beta-amyloid (Aβ) or tau binding to Aβ in a specific and selective fashion (Villemagne
in Alzheimer’s disease (AD) is allowing early diagnosis at et al., 2008a). Furthermore, low non-specific binding to
presymptomatic stages, more accurate differential diagnosis white matter is desirable as Aβ deposition usually starts at
as well as, when available, the evaluation and monitoring layers III and IV of the cortex (Thal et al., 2000) and spills
of disease-modifying therapy (Table 12.1) (Villemagne and over from high non-specific binding in white matter mask
Okamura, 2014; Rowe and Villemagne, 2011). or reduces the ability to detect this early deposition.
The last two decades have been focused on developing
Aβ radiotracers to detect cortical Aβ deposition in vivo
and some of them have already been approved for clinical
12.2 MOLECULAR IMAGING use. Aiming at mirroring the achievements of Aβ imaging,
RADIOTRACERS renewed efforts have been directed to the design of selec-
tive tau radiotracers (Klunk et al., 2004; Villemagne et al.,
Therapies, especially those targeting irreversible neuro- 2012a; Villemagne and Okamura, 2014). Stemming from
degenerative processes, have a better chance of success if the tracer’s capacity to bind a particular target over other

Table 12.1 Potential roles for Aβ and tau imaging in Alzheimer’s disease
• Accurate diagnosis of Alzheimer’s disease
• Prediction (Aβ) and monitoring (tau) of disease progression
• Early diagnosis of Alzheimer’s disease, allowing intervention when minimally impaired
• Investigate the spatial and temporal pattern of Aβ and tau deposition over a period of time, their interplay as well as
their relation to genetic and epigenetic factors, onset of phenotype, cognitive decline, brain volumetrics and other
disease biomarkers
• Subject selection for disease-specific therapeutic trials
• Monitor the effectiveness of anti-Aβ or anti-tau therapy
• Predict response to disease-specific therapy

119
120 Dementia

similar misfolded proteins has been used to classify them as Αβ-binding dye thioflavin-T that shows both high affinity
selective or non-selective (Villemagne and Okamura, 2014). and high specificity for fibrillary Aβ in plaques (Cohen et al.,
2012; Ye et al., 2005) as well as other Aβ-containing lesions
12.2.1 SELECTIVE Aβ IMAGING including cerebral amyloid angiopathy (CAA) (Lockhart
RADIOTRACERS et al., 2007). Furthermore, no specific binding was found in
subcortical white matter (Fodero-Tavoletti et al., 2009) and
Several compounds have been evaluated as potential Aβ at tracer concentrations, [11C]PiB does not bind at detectable
probes. Almost a decade after unsuccessful trials with levels to other misfolded proteins with a similar secondary
Aβ fragments, anti-Aβ antibodies and different dyes β-sheet structure such as α-synuclein (Fodero-Tavoletti et al.,
(Villemagne et al., 2014a) Αβ imaging came to fruition with 2007) or tau (Ikonomovic et al., 2012; Lockhart et al., 2007).
the first report of successful imaging in an AD patient with On visual inspection, cortical retention of [11C]PiB, regard-
2-(l-{6-[(2-[18F]fluoroethyl)(methyl)amino]-2-naph-thyl} less of disease severity, is markedly elevated in AD (Klunk
ethylidene)malononitrile (18F-FDDNP), a tracer character- et al., 2004; Mintun et al., 2006; Rowe et al., 2007), being high-
ized for binding both plaques and NFT (Shoghi-Jadid et al., est in frontal, cingulate, precuneus, striatum, parietal and
2002). Since then, human Αβ imaging studies have been con- lateral temporal cortices (Figure 12.2). Post-mortem studies
ducted in AD patients, normal controls and patients with using [11C]PiB revealed high correlations with regional Aβ
other dementias using C-11 labelled Pittsburgh Compound retention as measure at autopsy (Ikonomovic et al., 2012),
B (11C-PiB) (Klunk et al., 2004), 11C-SB13 (Verhoeff et al., as well as with Aβ1-42 in cerebrospinal fluid (CSF) (Zwan
2004), 11C-ST1859 (Bauer et al., 2006), 11C-BF227 (Kudo et al., et al., 2014a). A handful of other C-11 labelled radiotrac-
2007), 18F-florbetaben (Rowe et al., 2008a), [18F]flutemetamol ers have been tested in human studies, although low effect
(Vandenberghe et al., 2010), 11C-AZD2138 (Nyberg et al., sizes ([11C]SB13) (Verhoeff et al., 2004), small dynamic range
2009), 18F-florbetapir (Wong et al., 2010) and 18F-AZD4694 ([11C]ST1859) (Bauer et al., 2006) or lack of selectivity ([11C]
(Cselenyi et al., 2012) with PET and with 123I-clioquinol BF-227) (Okamura et al. 2004) made them less favourable
(CQ) (Opazo et al., 2006) and 123I-IMPY (Newberg et al., for detection of cortical Aβ burden compared with 11C-PiB.
2006) using single photon emission computed tomography On the other hand, a couple of C-11 tracers, [11C]AZD2184
(SPECT) (Figure 12.1). (Nyberg et al., 2009) and [11C]AZD2995 (Forsberg et al.,
The focus on radiotracers for Aβ imaging in vivo 2013) are detected projecting similar characteristics as [11C]
(Villemagne and Rowe, 2010) resulted in the development PiB. However, widespread clinical implementation of these
of multiple compounds, some of which were successfully C-11 tracers is hampered by the short radioactive half-life of
tested in human clinical trials, including the most success- C-11 (T½ = 20 min) requiring on-site production. To over-
ful and widely used radiotracer till date, 11C-PiB (Klunk come this limitation, various F-18 (T½ = 110 min) labelled
et al., 2004) (Figure 12.1). [11C]PiB is a derivative of the tracers were developed.

N H
N H
NC CN S N
OH 11CH
3
H O N 11
11C-PiB 18 CH3
F
CH3 18F-NAV4696
18F
N H
N
CH3 N N
18F-FDDNP
H S N 11CH
F O N 3
O
11CH
S CH3 11
C-AZD2184
3 11C-BF227
N 18F
N 11
CH3
N H
OH OH
H N
123I N S
11C-SB13 OH CH3
HN
18
F-f lutemetamol
CI
123I-CQ 18F
O
OH O O
11CH HN
2 18F-f lorbetaben
OH N
N
N 18F
123I O
18
F-f lorbetapir N O O
11C-ST1859 123I-IMPY

Figure 12.1 Chemical structure of Aβ ligands.


Molecular brain imaging in dementia 121

Figure 12.2 (See colour insert.) Typical pattern of 11C-PiB binding in AD. Representative sagittal and transaxial [11C]
PiB PET images overlayed on their respective MRI in a healthy elderly control subject (HC) (left) compared with an
Alzheimer’s disease (AD) patient (right). 11C-PiB retention is observed in frontal, temporal and parietal cortices as well as in
the posterior cingulate/precuneus areas, with relative sparing of occipital and sensorimotor cortex.

[18F]florbetapir ([18F]AV-45, Amyvid®), is a stilbene deriv- (Figure 12.1). [18F]florbetaben showed selective binding to
ative synthesized by Kung and colleagues at the University of Aβ plaques with lack of binding to Lewy bodies or NFT
Pennsylvania (Zhang et al., 2005a, 2005b) (Figure 12.1). [18F] in post-mortem tissue at low nanomolar concentrations
florbetapir showed a 96% agreement with neuropathology (Fodero-Tavoletti et al., 2012). Global 18F-florbetaben reten-
(Clark et al., 2011) with a sensitivity of 92% and a specificity tion showed excellent correlation with [11C]PiB retention
of 100% for detection of cortical Aβ in vivo against moder- (Villemagne et al., 2011a). Furthermore, it was effective in
ate to frequent Aβ plaque pathology at autopsy (Clark et al., discriminating AD from clinically normal controls and
2012). Furthermore, in direct comparison study, [18F]flor- frontotempolar lobar degeneration (FTLD) patients with
betapir was highly correlated with [11C]PiB (Landau et al., high sensitivity and specificity to detect high Aβ burden
2014) and found suitable to discriminate AD from controls against clinical diagnosis (Rowe et al., 2008; Barthel et al.,
(Wong et al., 2010). A multicentre study showed that, both 2011) and was able to detect the presence or absence of AD
in at-risk subjects and AD patients, high cortical Aβ burden pathology in a mixed population of cognitive impaired sub-
was associated with worse cognitive performance (Sperling jects (Villemagne et al., 2011a). In mild cognitive impair-
et al., 2013) and these patients were predicted to face a ment (MCI) patients, [18F]florbetaben was correlated with
greater cognitive deterioration over 3 years (Doraiswamy episodic memory and showed a predictive accuracy of 83%
et al., 2012; Doraiswamy et al., 2014). Furthermore, high for conversion to AD after 2 years and up to 94% after 4
cortical Aβ burden measure by 18F-florbetapir was associ- years (Ong et al., 2015). Furthermore, a multicentre phase 3
ated with lower memory performance in clinical normal trail confirmed that florbetaben PET was able to detect cor-
elderly (Sperling et al., 2013). [18F]Florbetapir was the first tical fibrillar Aβ plaques as assessed by visual reading with
radiotracer approved by the Food and Drug Administration 100% sensitivity and 92% specificity against post-mortem
(FDA; April 2012) and the European Medicines Agency silver-staining and immunohistochemistry (Sabbagh et al.,
(EMA; January 2013) for detection of Aβ in vivo and is being 2012). [18F]florbetaben received FDA and EMA approval for
used for both patient selection and evaluation of treatment clinical use in February and March 2014, respectively.
response in several anti-Αβ drug trails (Sperling et al., 2011). [18F]flutemetamol (GE-067, Vizamyl ®) is the [3]-fluoro-
To date, it is regarded as the most widely used Aβ radio- derivative of [11C]PiB developed by GE Healthcare for
tracer after [11C]PiB. detection of cortical Aβ deposition (Figure 12.1). A histo-
[18F]florbetaben ([18F]AV-1, [18F]BAY-94-9172, Neuraceq®) pathology study showed high concordance and strong cor-
was also synthesized by Kung and colleagues (Zhang et al., relation between [18F]flutemetamol PET retention and Aβ
2005a) and developed by Bayer Healthcare and Piramal burden measure by a monoclonal antibody in brain biopsy
122 Dementia

tissue and at autopsy (Ikonomovic et al., 2012). In addition, tracer retention than [11C]PiB is accompanied by higher
[18F]flutemetamol retention is highly correlated with [11C] white matter uptake, whereas in the cases of [18F]florbetapir
PiB across the spectrum of AD (Vandenberghe et al., 2010) and [18F]florbetaben (Landau et al., 2014; Villemagne et al.,
and phase II and III trails confirmed that [18F]flutemetamol 2011a), lower cortical retention than [11C]PiB is accompa-
can differentiate between AD and controls with a sensitivity nied by a similar white matter uptake. This results in a very
and specificity of 93% (Nelissen et al., 2009; Vandenberghe sharp contrast between grey and white matter in those cases
et al., 2010). Furthermore, [18F]flutemetamol PET has addi- with low or no detectable Aβ deposition in the brain and
tive value to MRI for diagnostic classification and prediction loss of this normal grey–white matter demarcation has been
of conversion in MCI subjects (Thurfjell et al., 2012; Duara proposed for the correct identification of high Aβ burden
et al., 2012). [18F]flutemetamol received FDA approval in with these F-18 tracers.
October 2013 and EMA approval in September 2014 under While all of the aforementioned tracers bind with vary-
the brand name of Vizamyl®. ing degrees of success to Aβ fibrils and brain homogenates
[18F]NAV4694 (also known as [18F]AZD4694) is a radio- of AD patients, Congo Red and thioflavin T – and some
tracer developed by Astra-Zeneca and Navidea (Figure of their derivatives – have been shown to also bind to the
12.1). Direct comparison with [11C]PiB revealed that [18F] soluble oligomeric forms of Aβ (Maezawa et al., 2008). On
AZD4694 showed similar binding kinetics, dynamic reten- the other hand, Aβ soluble species reportedly represent less
tion range and low non-specific binding to white matter than 1% of the total brain Aβ (McLean et al., 1999) and the
with an excellent correlation between [18F]AZD4694 and reported affinity of PiB for these soluble oligomers seems
[11C]PiB cortical retention (Rowe et al., 2013b). Furthermore, to be significantly lower than for Aβ fibrils (Maezawa et al.,
[18F]AZD4694 clearly identified AD patients from healthy 2008). Until highly selective radiotracers are developed to
controls both by quantitative measure and visual inspection bind the Aβ soluble species, the contribution of these oligo-
(Rowe et al., 2013b; Cselenyi et al., 2012). mers to the Αβ imaging PET signal in sporadic AD from
For visual inspection of Aβ imaging studies, high radio- tracers such as 11C-PiB is considered to be negligible (Mathis
tracer retention in the cortical and subcortical grey matter et al., 2007).
is synonymous of a high Aβ burden (Zwan et al., 2014b) Preliminary studies with SPECT Aβ radiotracers (Figure
(Figure 12.3). In contrast to [11C]PiB or [18F]NAV4694, [18F] 12.1) shows limited utility for the evaluation of Aβ burden
florbetapir, [18F]florbetaben and [18F]flutemetamol (Figure in AD (Newberg et al., 2006). New SPECT radiotracers
12.3) show the presence of relatively high white matter labelled with 123I or those that could be potentially labelled
uptake. In the case of [18F]flutemetamol, similar cortical with 99mTc are being evaluated (Lin et al., 2009).

Figure 12.3 (See colour insert.) Aβ imaging in Alzheimer’s disease. Representative parametric sagittal, transaxial and
coronal PET images of Alzheimer’s disease (AD) patients assessed with either [11C]PiB, [18F]florbetaben, [18F]flutemetamol,
[18F]florbetapir or [18F]NAV4694. All AD patients harboured high Aβ burdens in the brain, reflected in marked radiotracer
retention in cortical and subcortical grey matter areas. [18F]Florbetapir (Amyvid®); [18F]flutemetamol (Vizamyl®) and [18F]
florbetaben (Neuraceq®) have already been approved for clinical use by the FDA and EMA.
Molecular brain imaging in dementia 123

12.2.2 SELECTIVE TAU IMAGING deposits (Fodero-Tavoletti et al., 2014) nor other non-AD tau
RADIOTRACERS lesions. Initial PET studies showed significantly higher [18F]
THK523 retention in hippocampi, temporal, parietal and
In AD, the prevalent ultrastructural conformation of tau orbitofrontal regions of AD patients, but because cortical
aggregates is PHF, therefore, most of the focus has been retention was significantly lower than white matter reten-
directed at developing selective tau imaging radiotracers for tion it was impossible to discriminate tracer retention by
PHF-tau. Notwithstanding, some of these tracers recognize visual inspection of the images (Villemagne et al., 2014b).
other conformations of tau aggregates found in non-AD Notably, in healthy controls with high Aβ burden as assessed
tauopathies. The development of novel selective tau ligands by [11C]PiB, while cortical [18F]THK523 retention was low,
(Figure 12.4) has led to successful first-in-human tau imag- [18F] THK523 retention in hippocampus and insula was
ing PET studies. While several potential tracers have been similar to the one observed in AD (Villemagne et al., 2014b).
proposed by research groups working on anti-tau therapeu- Two improved derivatives, [18F]THK5105 and [18F]THK5117
tics (for review see Villemagne and Okamura, 2014), only a (Figure 12.4), were developed and tested (Okamura et al.,
few tau tracers amenable to radiolabeling made the transi- 2013). Both tracers displayed higher binding affinity to tau-
tion into human trials (Chien et al., 2013; Maruyama et al., rich AD brain homogenates than [18F]THK523 (Okamura
2013; Okamura et al., 2013). et al., 2013). Initial human PET studies with [18F]THK5105
For more than a decade, Kudo and colleagues at Tohoku and [18F]THK5117 demonstrated a substantially lower
University, Sendai, Japan, have been designing, developing retention in subcortical white matter and cortical reten-
and screening small binding molecules targeting misfolded tion in AD patients analogous to the pattern of tau depos-
proteins (Okamura et al., 2005, 2013). The first candidate, its described by Braak and Braak (1997) or Delacourte et al.
[18F]THK523 (Figure 12.4) showed low nanomolar affinity (1999), ­leading to a robust visual and quantitative separation
to tau fibrils, a 12-fold selectivity for tau over Aβ, and sig- between AD patients and healthy controls, while also corre-
nificant in vivo tracer retention in a tau transgenic mouse lating with dementia severity and brain atrophy (Okamura
model (Fodero-Tavoletti et al., 2011). In vitro studies dem- et al., 2014a) and showing a different pattern of retention
onstrated that while [18F]THK523 bound selectively to between tau and Aβ imaging with PiB (Okamura et al.,
PHF-tau (Harada et al., 2013), it did not bind to α-synuclein 2014b; Okamura et al., 2014a). [18F]THK5351, a new addition

N 18F O NC CN

N N CH3
H H 18F
N N
18F N
H
18
F-THK-523 CH3
18F-T807
18F-FDDNP
OH
18
F O
N

N N N N
CH3 N O
18F N S N
18F-THK-5105
CH3 N
18F-T808 11CH
3 O
OH 11 CF3
C-N-Methyl lansoprazole
F
18
O

H N N
H
OH S N
H F-THK-5117
18

CH3 N O
N
S N
N H311C N
11C-PBB3
OH CH3
18F O O
18 C18F3
N F-N-Methyl lansoprazole
H
N N
18F-THK-5351
CH3

Figure 12.4 Chemical structure of tau imaging ligands.


124 Dementia

to the THK series (Figure 12.4), has been recently evaluated binds tau at high specific activities (Maruyama et al., 2013).
in young and elderly volunteers as well as in AD patients A completely different [11C]PBB3 regional retention pattern
(Okamura et al., 2014c). The preliminary evaluation of [18F] was observed in a CBS patient, suggesting [11C]PBB3 might
THK5351 showed that it has faster tracer kinetics and higher also bind other non-PHF tau conformations. Development
signal-to-noise ratios than [18F]THK5105 and [18F]THK5117, of more selective and fluorinated derivatives will further
­suggesting [18F]THK5351 might be a superior tau tracer confirm the usefulness of the PBB scaffold.
(Okamura et al., 2014c).
Kolb and colleagues, first at Siemens and then at Avid/ 12.2.3 NON-SELECTIVE PAN-AMYLOID
Lilly have developed [18F]AV1451 ([18F]T807) and [18F]T808 IMAGING RADIOTRACERS
(Figure 12.4) two novel benzimidazole-pyrimidines deriva-
tives with nanomolar affinity and more than a 25-fold selec- [18F]FDDNP is a naphthol derivative synthesized and
tivity for PHF-tau over Aβ (Chien et al., 2013; Xia et al., characterized by Barrio and colleagues at UCLA (Barrio
2013; Chien et al., 2014). Initial human PET studies showed et al., 1999). [18F]FDDNP was initially reported to bind
very low [18F]AV1451 white matter retention and cortical non-selectively to both extracellular Aβ plaques and intra-
retention in AD that followed the known distribution of cellular NFT (Shoghi-Jadid et al., 2002; Smid et al., 2013)
tau deposits in the brain, retention that was also associated and has been applied to the evaluation of a wide spec-
with disease severity (Chien et al., 2013). Prof Johnson and trum of neurodegenerative conditions such as AD (Small
Dickerson at the Massachusets General Hospital in Boston et al., 2006), CTE (Small et al., 2013), Down syndrome
are conducting the largest series of head-to-head [18F]AV1451 (Nelson et al., 2011), Gerstmann–Straussler–Scheinker
and [11C]PiB PET studies in AD and non-AD tauopathies disease (Kepe et al., 2010), Creutzfeld–Jakob disease (CJD)
as well as controls (Johnson et al., 2013a; Dickerson et al., (Bresjanac et al., 2003), FTLD and progressive supranu-
2014). On preliminary evaluation, [18F]T808 displayed faster clear palsy (PSP) (Kepe et al., 2013). Human PET studies
tracer kinetics than [18F]AV1451, reaching steady state dur- showed [18F]FDDNP has a very narrow dynamic range
ing the scanning period (Chien et al., 2014). Unfortunately, (Small et al., 2006; Tolboom et al., 2009b), to correlate with
substantial defluorination was observed in some cases CSF-tau (Tolboom et al., 2009a), whereas in vitro assess-
(Chien et al., 2014). ments in concentrations similar to those achieved during a
Due to its in vitro binding affinity to PHF-tau, lansopra- PET scan showed limited binding to both Aβ plaques and
zole was assessed as a potential tau imaging tracer. A series NFT (Thompson et al., 2009). Given is a non-selective pan-
of lanzoprasole derivatives with subnanomolar affinity for amyloid tracer, [18F]FDDNP counterbalances its inability
tau fibrils were radiolabelled with 11C (Shao et al., 2012) to identify the misfolded protein responsible for a specific
and more recently, 18F (Fawaz et al., 2014) (Figure 12.4). phenotype by using the regional retention of [18F]FDDNP
Preclinical mice studies showed no entry of 11C-N-methyl- in the brain as to discriminate amid different neurodeagen-
lanzoprasole into the brain, effect that was reversed after erative conditions.
inhibition of the permeability-glycoprotein 1 transporter
with cyclosporine (Shao et al., 2012). In contrast, non-
human primate studies with either the 11C and 18F versions
of the tracer showed unencumbered entry into the brain 12.3 Aβ IMAGING
(Shao et al., 2012; Fawaz et al., 2014). No human studies
have been reported till date with these tracers. On visual inspection, as well as in quantitative and semi-
More recently, Maruyama and colleagues from the quantitative PET studies of different Aβ radiotracers, there
National Institute of Radiological Sciences, Chiba, Japan, is a robust difference in tracer retention between AD patients
described the preclinical and initial clinical characteriza- and age-matched controls (Rowe et al., 2007; Klunk et al.,
tion of a novel C-11 labelled selective tau tracer, [11C]PBB3 2004; Jack et al., 2008), with more than 90% of AD patients
(Maruyama et al., 2013; Hashimoto et al., 2014) (Figure (Figure 12.3) showing cortical tracer retention higher in
12.4). The comprehensive preclinical evaluation involved in frontal, cingulate, precuneus, striatum, parietal and lat-
vitro autoradiography, two-photon laser scanning fluores- eral temporal cortices, while occipital, sensorimotor and
cence microscopy and small animal PET studies, showing mesial temporal cortices are much less affected. These PET
higher PBB3 binding in the spinal cord of a tau transgenic images present a pattern of radiotracer retention that seems
mouse model (Maruyama et al., 2013). Although 70% of to replicate the sequence of Aβ deposition found at autopsy
unchanged [11C]PBB3 was found in mice brain homog- (Braak and Braak, 1997), but also similar to the anatomy of
enates, metabolite analysis showed that only 2% of the par- the ‘default network’ (Sperling et al., 2009b). The regional
ent compound remained in plasma at 5 min after injection distribution of these tracers varies with the characteristics
(Hashimoto et al., 2014). Preliminary clinical studies with of Aβ distribution among the different genotypes and phe-
both [11C]PBB3 and [11C]PiB in healthy controls and AD notypes. For example, carriers of mutations associated with
patients showed a distinct pattern of brain retention between familial AD (Klunk et al., 2007; Villemagne et al., 2009a),
the two tracers suggesting that and despite marked [11C] subjects with posterior cortical atrophy (Ng et al., 2007;
PBB3 retention in the venous sinuses, [11C]PBB3 selectively Tenovuo et al., 2008) or CAA (Dierksen et al., 2010; Johnson
Molecular brain imaging in dementia 125

et al., 2007) present a different pattern of tracer retention to (Wolk et al., 2009; Forsberg et al., 2008; Okello et al., 2009;
the one observed in sporadic AD (Klunk et al., 2004; Rowe Villemagne et al., 2011b). The fact that high Aβ burden
et al., 2007). relates to episodic memory impairment in non-demented
While the accuracy of clinical assessments for the diagno- individuals and that it is associated with a significant higher
sis of AD ranges between 70% and 90%, the regional reten- risk of cognitive impairment, emphasizes the non-benign
tion of Aβ radiotracers is highly correlated with regional nature of Aβ and supports the hypothesis that Aβ deposi-
Aβ plaques as reported at autopsy or biopsy (Ikonomovic tion occurs well before the onset of symptoms, further sug-
et al., 2008; Wolk et al., 2011; Sojkova et al., 2011; Clark et al., gesting that early disease-specific therapeutic intervention
2011), with higher Aβ burden in the frontal cortex than in at the presymptomatic stage might be the most promising
hippocampus, consistent with previous reports (Naslund approach to either delay onset or halt disease progression
et al., 2000). (Sperling et al., 2011). On the other hand, the prevalence
About 25%–35% of cognitively unimpaired elderly sub- of high Aβ deposition among non-demented individu-
jects present with high Aβ burden (Mintun et al., 2006; als, added to the absence of a strong association between
Villemagne et al., 2008b; Aizenstein et al., 2008; Rowe Aβ deposition and measure of cognition, synaptic activ-
et al., 2010; Rowe et al., 2007) (Figure 12.3), are in perfect ity and neurodegeneration in AD, suggesting that Aβ is an
agreement with post-mortem reports (Davies et al., 1988; early and necessary, though not sufficient cause for cogni-
Morris and Price, 2001) and likely to reflect preclinical AD tive decline in AD (Villemagne et al., 2008b; Sojkova and
(Morris and Price, 1999). While neuropsychological exami- Resnick, 2011), indicates that other – Aβ-dependent or inde-
nation show that individuals with high Aβ burden perform pendent – factors, such as acceleration and/or spreading of
within the normal limits, they do perform worse and have tau aggregation beyond the MTL, lead to synaptic failure
a higher risk for disease progression than those with low and eventually neuronal loss in cortical association areas
Aβ burden (Rowe et al., 2013a; Sperling et al., 2013). The which play a crucial role in cognitive decline and disease
detection of Aβ pathology at the presymptomatic stage is progression. Other factors such as age, years of education,
of crucial importance because if this group truly represents IQ, occupational level and brain volume among others
preclinical AD (Thal et al., 2004; Morris and Price, 1999), appear to play a modulatory role between Aβ deposition
it is precisely the group that may benefit the most from and cognition (Roe et al., 2008; Chetelat et al., 2010).
therapies aimed at reducing or eliminating Aβ from the With regard to some of these environmental variables,
brain (Sperling et al., 2011). Furthermore, one of the most age is the strongest risk factor in sporadic AD, with the
important conclusions to be drawn from all the Aβ imag- prevalence of the disease increasing exponentially with
ing studies conducted is that, the likelihood of a cognitively age. These risks are increased in the presence of the most
unimpaired individual with low Aβ burden in the brain consistent genetic risk factor associated with sporadic AD,
developing AD dementia is extremely small (Villemagne the ApoE ε4 allele (Farrer et al., 1997). The presence of the
et al., 2011b; Rowe et al., 2013a). Aβ imaging has proven ApoE ε4 allele has been associated with onset of the dis-
useful in identifying AD pathology in about 50%–70% of ease at an earlier age and a gene dose dependent on higher
individuals fulfilling criteria for MCI (Winblad et al., 2004; risk of developing AD (Farrer et al., 1997). Examination of
Petersen, 2000; Kemppainen et al., 2007; Pike et al., 2007; ApoE ε4 allele status revealed that, independent of clinical
Forsberg et al., 2008; Mormino et al., 2009). Most ­studies classification, ε4 carriers present with significantly higher
reported that subjects classified as non-amnestic MCI – Aβ burdens than non-ε4 carriers, further emphasizing the
and more specifically those classified as non-amnestic crucial role ApoE plays in Aβ metabolism (Reiman et al.,
single domain MCI – show low Aβ burden in the brain, 2009; Morris et al., 2010; Rowe et al., 2010). Although ApoE
consistent with a non-AD underlying pathological process ε4 carriage is associated with higher Aβ burdens and early
(Pike et al., 2007; Villemagne et al., 2011b). Although, at the disease onset, it has no effect on the rates of Aβ deposition
early MCI stages Aβ and hippocampal volume have inde- (Villemagne et al., 2013). Aβ imaging is also proving useful
pendent effects on cognition (Ong et al., 2013), at the late in the characterization of autosomal mutations in APP or
MCI stage, it is believed that the Aβ-related impairment is presenilin 1 or 2 genes that lead to the overproduction of Aβ,
mediated by hippocampal atrophy (Mormino et al., 2009; where mutation carriers present with abnormal Aβ deposi-
Ong et al., 2015) and modulated by cognitive reserve (Rentz tion up to 25 years before the onset of dementia (Benzinger
et al., 2010; Roe et al., 2008). et al., 2013; Bateman et al., 2012) has been noticed. These
Both post-mortem (Bennett et al., 2006; Price and studies are helping validate the relevance and change over
Morris, 1999) and Aβ imaging studies (Jack et al., 2013; a course of time of several biomarkers (Fagan et al., 2014;
Villemagne et al., 2013) indicate that Aβ deposition starts Benzinger et al., 2013), assessments that are being applied to
decades before dementia phenotype is manifested and non- better-targeted disease-specific therapeutic strategies (Mills
demented individuals with high Aβ burden in the brain are et al., 2013).
at a much greater risk of cognitive decline (Resnick et al., Aβ imaging has also been applied to a wide spectrum
2010; Villemagne et al., 2008b; Villemagne et al., 2011b; of neurodegenerative conditions. Although lower than
Rowe et al., 2013a), highlighting the clinical relevance and found in AD, similar patterns of Aβ deposition are usu-
potential impact in patient management of Aβ imaging ally observed in dementia with Lewy bodies (DLB) (Rowe
126 Dementia

et al., 2007; Gomperts et al., 2008; Maetzler et al., 2009) 2011; Rabinovici et al., 2008) (Figure 12.5). In contrast to
(Figure 12.5). There is usually no detectable Aβ burden FTLD and as confirmed by autopsy and Aβ imaging studies,
in patients with sporadic CJD (Villemagne et al., 2009b) more than half of DLB patients show signs of high Aβ depo-
(Figure 12.5) or those diagnosed with FTLD (Rowe et al., sition (McKeith et al., 2005; Rowe et al., 2007) (Figure 12.5).
2007; Drzezga et al., 2008; Rabinovici et al., 2007) with the Therefore, differential diagnosis from AD can be better
exception of patients presenting with logopenic aphasia that accomplished by assessing the integrity of the dopami-
do have high Aβ burden in the brain, and are thought to nergic nigrostriatal terminals (Villemagne et al., 2012c).
represent a language-onset variant of AD (Leyton et al., Aβ imaging has also facilitated differential diagnosis in

PiB- HC

Misfolded
proteins PiB- MCI

PD

α-syn

PiB- DLB

PiB+ HC

PiB+ MCI

Aβ PiB+ DLB

AD

tau

FTLD

PrPsc spCJD

Figure 12.5 (See colour insert.) Aβ imaging in ageing and dementia. Representative 40–70 min post-injection trans-
axial 11C-PiB images along the spectrum of some of the misfolded proteins associated with neurodegenerative diseases,
encompassing the different patterns of 11C-PiB retention in the brain, with – from top to bottom – a 73-year-old healthy
PiB-negative control (PiB-HC) subject (mini mental state examination [MMSE] 30), 83-year-old PiB-negative subject with
mild cognitive impairment (PiB-MCI) (MMSE 28), 61-year-old PiB-negative Parkinson’s disease (PD) patient (MMSE 27),
69-year-old PiB-negative patient with dementia with Lewy Bodies (DLB; MMSE 24), 77-year-old PiB-positive healthy
control (PiB+ HC) subject (MMSE 28), 82-year-old PiB-positive subject with mild cognitive impairment (PiB+ MCI) (MMSE
28), 78-year-old PiB-positive DLB patient (PiB+ DLB) (MMSE 19), 76-year-old Alzheimer’s disease (AD) patient (MMSE 21),
59-year-old patient with frontotemporal lobe degeneration (FTLD; MMSE 20) and 59-year-old PiB-negative patient with
confirmed sporadic Creutzfeldt–Jakob disease (spCJD). PET images show clear differences when comparing cortical PiB
retention in PiB-negative HC, MCI, PD, spCJD and FTLD with PiB-positive HC, MCI, DLB or AD patients. Only non-specific
PiB binding in white matter is observed in PiB-negative HC, MCI, PD, spCJD and FTLD compared with PiB binding in corti-
cal areas of AD and DLB patients. All images are scaled to the same standardized uptake value ratio (SUVR) maximum.
Molecular brain imaging in dementia 127

cases of inpatients with atypical presentations of dementia Aβ imaging is also assisting the evaluation of anti-Aβ
(Ng et al., 2007; Wolk et al., 2012). therapies in several ways, allowing better subject selec-
Another rapidly expanding area is the examination of tion, assessing target engagement and evaluating treat-
potential association between fluid and imaging biomark- ment response of patients for therapy trials and providing a
ers of pathology or neurodegeneration and how these bio- means to measure their impact on Aβ burden (Rinne et al.,
marker constructs are used to establish the criteria for the 2010; Ostrowitzki et al., 2011; Salloway et al., 2014).
different stages of the disease (Thal et al., 2006; Clark et al.,
2008; Albert et al., 2011; McKhann et al., 2011; Storandt
et al., 2012; Morris et al., 2005).
Clear criteria for appropriate use of Aβ imaging has been 12.4 TAU IMAGING
established emphasizing the need to integrate the result of
Aβ imaging with a comprehensive cognitive and clinical Tau imaging is a recent addition to the arsenal of tools for the
evaluation performed by a clinician experienced in the eval- non-invasive assessment of neurodegenerative conditions.
uation of dementia, to ensure a positive impact on patient While several international groups are implementing and
management (Johnson et al., 2013b). The criteria clearly applying tau imaging for the evaluation of AD (Figure 12.6)
stipulates the specific cases Aβ imaging should be used, such and non-AD tauopathies, little has been communicated or
as patients with persistent or progressive unexplained cog- published beyond the in vitro and initial human proof of
nitive impairment, progressive atypical or unclear clinical mechanism studies assessing novel tau tracers. However,
presentation of dementia, or dementia onset in i­ndividuals these initial studies have furnished enough information to
65 years old or younger (Johnson et al., 2013b). It also out- allow some clearer speculation on the potential applications
lines the inappropriate use of Aβ ­imaging, such as cases of this new technique. Although most of the potential appli-
of probable AD with typical age of onset or to determine cations are similar to those of Aβ imaging, there are some
dementia severity, asymptomatic i­ndividuals or uncon- that seem more appropriate or better suited for tau imaging.
firmed cognitive complaint, a family history of dementia or Given the critical and stereotypical pattern of tau deposi-
presence of the ApoE ε4 allele, as well as non-medical use tion and in contrast with CSF studies, tau imaging studies
found in litigation for insurance purposes (Johnson et al., can assess both the quantity and regional brain distribu-
2013b). tion of tau deposits, features that can be potentially used for

HC AD

18F-flutemetamol 18F-flutemetamol
L R R L L R R L

PET-SUVRpons PET-SUVRpons
1.2 1.2
0.9 0.9
RT.LAT LT.LAT SUP INF RT.LAT LT.LAT SUP INF
0.7 0.7
0.4 0.4

RT.MED LT.MED POST ANT RT.MED LT.MED POST ANT

18F-THK5117 18F-THK5117
L R R L L R R L

PET-SUVRCbCtx PET-SUVRCbCtx
2.0 2.0
1.6 1.6
RT.LAT LT.LAT SUP INF RT.LAT LT.LAT SUP INF
1.2 1.2
0.8 0.8

RT.MED LT.MED POST ANT RT.MED LT.MED POST ANT

Figure 12.6 (See colour insert.) Aβ and tau imaging in ageing and dementia. Surface display PET images of the respective
Aβ ([18F]flutemetamol, top row) and tau ([18F]THK5117, bottom row) burden in an age-matched healthy control (HC) and an
Alzheimer’s disease (AD) patient.
128 Dementia

staging the disease. Given the tight association between tau (Zetterberg and Blennow, 2006), a simple blood test assess-
deposition, cognition and neurodegeneration, tau imaging ing central features of the disease (Burnham et al., 2014) as
might prove essential for assessing disease progression. well as the use of near-infrared Αβ dyes (Raymond et al.,
The combination of tau with Aβ imaging studies will help 2008) should soon be available to permit widespread screen-
ascertain if Aβ accelerates and/or triggers the spread of tau ing of the population at-risk. On the other hand, molecu-
deposition outside the mesial temporal cortex and if this initial lar neuroimaging can provide highly accurate, reliable and
dissemination into cortical association areas is manifested as reproducible quantitative statements of Aβ and tau burden,
incipient and insidious objective cognitive impairment occurs essential for therapeutic trial recruitment and for the evalu-
known as MCI (Price and Morris, 1999; Delacourte et al., ation of disease-specific treatments directed at removing or
2002). Further spreading into the remaining cortical areas is slowing the brain deposition of either Aβ or tau.
usually accompanied by severe cognitive deterioration and
dementia (Price and Morris, 1999; Delacourte et al., 2002). This REFERENCES
sequence of events might help explain the preliminary results
from combined Aβ and tau imaging studies (Johnson et al., Aizenstein, H.J., Nebes, R.D., Saxton, J.A. et al. (2008).
2013a; Villemagne and Okamura, 2014) and is one of the essen- Frequent amyloid deposition without significant
tial issues to be tackled by the in vivo imaging of tau deposits. cognitive impairment among the elderly. Archives of
Aβ imaging studies have shown that the actual amount of Aβ Neurology, 65: 1509–1517.
in the brain is more relevant than the regional Aβ distribution Albert, M.S., Dekosky, S.T., Dickson, D. et al. (2011).
as an early driver of cognitive decline. Post-mortem and initial The diagnosis of mild cognitive impairment due to
tau imaging PET studies indicate that, not only the amount Alzheimer’s disease: Recommendations from the
of tau deposition, but also – or mainly – their topographical National Institute on Aging-Alzheimer’s Association
distribution in the brain (Royall, 2007; Delacourte et al., 1999) workgroups on diagnostic guidelines for Alzheimer’s
might be more relevant and robustly associated with neuro- disease. Alzheimer’s and Dementia, 7: 270–279.
degeneration and cognitive decline. Furthermore, this extra Barrio, J.R., Huang, S.C., Cole, G. et al. (1999). PET imag-
dimension is not limited to the AD spectrum but essentially to ing of tangles and plaques in Alzheimer disease with
all tauopathies where the regional distribution of tau deposits a highly hydrophobic probe. Journal of Labelled
is pathognomonic for each specific condition. Compounds and Radiopharmaceuticals, 42: S194–S195.
Barthel, H., Gertz, H.J., Dresel, S. et al. (2011). Cerebral
amyloid-beta PET with florbetaben ((18)F) in patients
with Alzheimer’s disease and healthy controls: A
12.5 SUMMARY multicentre phase 2 diagnostic study. The Lancet
Neurology, 10: 424–435.
The introduction of radiotracers for the non-invasive in Bateman, R.J., Xiong, C., Benzinger, T.L. et al. (2012).
vivo quantification of Aβ in the brain has revolutionized the Clinical and biomarker changes in dominantly inher-
approach in the evaluation of AD. Frank et al. (2003) proposed ited Alzheimer’s disease. New England Journal of
that an ideal biomarker for AD should be disease-specific, Medicine, 367: 795–804.
non-invasive, with sensitivity and specificity above 80%. Αβ Bauer, M., Langer, O., Dal-Bianco, P. et al. (2006). A
imaging already fulfills these criteria. Aβ burden as measure positron emission tomography microdosing study
by PET matches histopathological reports of Aβ distribution with a potential antiamyloid drug in healthy volun-
in ageing and dementia, it appears more accurate than FDG teers and patients with Alzheimer’s disease. Clinical
for the diagnosis of AD and is an excellent aid in the differen- Pharmacology and Therapeutics, 80: 216–227.
tial diagnosis of AD from FTLD. ApoE ε4 status is associated Bennett, D.A., Schneider, J.A., Arvanitakis, Z. et al. (2006).
with higher Aβ burden. As new treatments in clinical trials Neuropathology of older persons without cogni-
are aimed at preventing or slowing AD progression, either tive impairment from two community-based studies.
by preventing Aβ generation or deposition, or increasing the Neurology, 66: 1837–1844.
clearance of Aβ, the role of imaging and quantifying Aβ bur- Benzinger, T.L., Blazey, T., Jack, C.R. et al. (2013). Regional
den in vivo is becoming increasingly crucial. Although these variability of imaging biomarkers in autosomal domi-
treatments are aimed at AD, the available data suggest that nant Alzheimer’s disease. Proceedings of the National
they may have value in other dementias such as DLB, where Academy of Sciences, 110: E4502–E4509.
Aβ deposition is present. Braak, H. and Braak, E. (1997). Frequency of stages of
Despite the development of promising new and reliable Alzheimer-related lesions in different age categories.
Aβ and tau imaging ligands labelled with isotopes with Neurobiology of Aging, 18: 351–357.
longer radioactive half-lives (Rowe et al., 2008b; Serdons Bresjanac, M., Smid, L.M., Vovko, T.D. et al. (2003).
et al., 2009; Sundgren-Andersson et al., 2009; Sperling et al., Molecular-imaging probe 2-(1-[6-[(2-fluoroethyl)
2009a; Chien et al., 2014; Okamura et al., 2013), population (methyl) amino]-2-naphthyl]ethylidene) malononi-
screening is unlikely to involve neuroimaging approaches trile labels prion plaques in vitro. The Journal of
and despite the unreliability of plasma assays till date Neuroscience, 23: 8029–8033.
Molecular brain imaging in dementia 129

Burnham, S.C., Faux, N.G., Wilson, W. et al. (2014). A and 18-month cognitive decline: A multicenter study.
blood-based predictor for neocortical Abeta burden Neurology, 79: 1636–1644.
in Alzheimer’s disease: Results from the AIBL study. Doraiswamy, P.M., Sperling, R.A., Johnson, K. et al. (2014).
Molecular Psychiatry, 19: 519–526. Florbetapir F 18 amyloid PET and 36-month cognitive
Chetelat, G., Villemagne, V.L., Pike, K.E. et al. (2010). decline:a prospective multicenter study. Molecular
Larger temporal volume in elderly with high versus low Psychiatry, 19: 1044–1051.
beta-amyloid deposition. Brain, 133: 3349–3358. Drzezga, A., Grimmer, T., Henriksen, G. et al. (2008).
Chien, D.T., Bahri, S., Szardenings, A.K. et al. (2013). Early Imaging of amyloid plaques and cerebral glucose
clinical PET imaging results with the novel PHF-tau metabolism in semantic dementia and Alzheimer’s
radioligand [F-18]-T807. Journal of Alzheimer’s Disease, disease. Neuroimage, 39: 619–633.
34: 457–468. Duara, R., Loewenstein, D.A., Shen, Q. et al. (2012).
Chien, D.T., Szardenings, A.K., Bahri, S. et al. (2014). Early Amyloid positron emission tomography with (18)
clinical PET imaging results with the novel PHF-tau F-flutemetamol and structural magnetic resonance
radioligand [F18]-T808. Journal of Alzheimer’s Disease, imaging in the classification of mild cognitive impair-
38: 171–184. ment and Alzheimer’s disease. Alzheimer’s and
Clark, C.M., Davatzikos, C., Borthakur, A. et al. (2008). Dementia, 9 (3): 295–301.
Biomarkers for early detection of Alzheimer pathology. Fagan, A.M., Xiong, C., Jasielec, M.S. et al. (2014).
Neurosignals, 16: 11–18. Longitudinal change in CSF biomarkers in autosomal-
Clark, C.M., Pontecorvo, M.J., Beach, T.G. et al. (2012). dominant Alzheimer’s disease. Science Translational
Cerebral PET with florbetapir compared with neuropa- Medicine, 6: 226ra30.
thology at autopsy for detection of neuritic amyloid- Farrer, L.A., Cupples, L.A., Haines, J.L. et al. (1997).
beta plaques: A prospective cohort study. The Lancet Effects of age, sex and ethnicity on the association
Neurology, 11: 669–678. between apolipoprotein E genotype and Alzheimer
Clark, C.M., Schneider, J.A., Bedell, B.J. et al. (2011). Use disease. A meta-analysis. APOE and Alzheimer
of florbetapir-PET for imaging beta-amyloid pathol- Disease Meta Analysis Consortium. JAMA, 278 (16):
ogy. JAMA, 305: 275–283. 1349–1356.
Cohen, A.D., Rabinovici, G.D., Mathis, C.A. et al. (2012). Fawaz, M.V., Brooks, A.F., Rodnick, M.E. et al. (2014). High
Using Pittsburgh Compound B for in vivo PET imaging affinity radiopharmaceuticals based upon lansopra-
of fibrillar amyloid-beta. Advances in Pharmacology, zole for PET imaging of aggregated tau in Alzheimer’s
64: 27–81. disease and progressive supranuclear palsy: Synthesis,
Cselenyi, Z., Jonhagen, M.E., Forsberg, A. et al. (2012). preclinical evaluation, and lead selection. ACS
Clinical validation of 18F-AZD4694, an amyloid-beta- Chemical Neuroscience, 5 (8): 718–730.
specific PET radioligand. The Journal of Nuclear Fodero-Tavoletti, M.T., Brockschnieder, D., Villemagne,
Medicine, 53 (3): 415–424. V.L. et al. (2012). In vitro characterization of [(18)
Davies, L., Wolska, B., Hilbich, C. et al. (1988). A4 amyloid F]-florbetaben, an Abeta imaging radiotracer. Nuclear
protein deposition and the diagnosis of Alzheimer’s Medicine and Biology, 39: 1042–1048.
disease: Prevalence in aged brains determined Fodero-Tavoletti, M.T., Furumoto, S., Taylor, L. et al.
by immunocytochemistry compared with conven- (2014). Assessing THK523 selectivity for tau deposits
tional neuropathologic techniques. Neurology, 38: in Alzheimer’s disease and non-Alzheimer’s disease
1688–1693. tauopathies. Alzheimers Research Therapy, 6 (1): 11.
Delacourte, A., David, J.P., Sergeant, N., et al. (1999). Fodero-Tavoletti, M.T., Okamura, N., Furumoto, S. et al.
The biochemical pathway of neurofibrillary degenera- (2011). 18F-THK523: A novel in vivo tau imaging ligand
tion in aging and Alzheimer’s disease. Neurology, 52: for Alzheimer’s disease. Brain, 134: 1089–1100.
1158–1165. Fodero-Tavoletti, M.T., Rowe, C.C., McLean, C.A. et al.
Delacourte, A., Sergeant, N., Wattez, A. et al. (2002). Tau (2009). Characterization of PiB binding to white matter
aggregation in the hippocampal formation: An ageing in Alzheimer disease and other dementias. The Journal
or a pathological process? Experimental Gerontology, of Nuclear Medicine, 50: 198–204.
37: 1291–1296. Fodero-Tavoletti, M.T., Smith, D.P., McLean, C.A. et al.
Dickerson, B., Domoto-Reilly, K., Hochberg, D. et al. (2007). In vitro characterization of Pittsburgh com-
(2014). Imaging tau pathology in vivo in FTLD: Initial pound-B binding to Lewy bodies. The Journal of
experience with 18F T807 PET. Neurology, 82: S8.007. Neuroscience, 27: 10365–10371.
Dierksen, G.A., Skehan, M.E., Khan, M.A. et al. (2010). Forsberg, A., Engler, H., Almkvist, O. et al. (2008). PET
Spatial relation between microbleeds and amyloid imaging of amyloid deposition in patients with mild
deposits in amyloid angiopathy. Annals of Neurology, cognitive impairment. Neurobiology of Aging, 29:
68: 545–548. 1456–1465.
Doraiswamy, P.M., Sperling, R.A., Coleman, R.E. et al. Forsberg, A., Jureus, A., Cselenyi, Z. et al. (2013). Low back-
(2012). Amyloid-beta assessed by florbetapir F 18 PET ground and high contrast PET imaging of amyloid-beta
130 Dementia

with [11C]AZD2995 and [11C]AZD2184 in Alzheimer’s Kepe, V., Bordelon, Y., Boxer, A. et al. (2013). PET imag-
disease patients. European Journal of Nuclear Medicine ing of neuropathology in tauopathies: Progressive
and Molecular Imaging, 40: 580–593. supranuclear palsy. Journal of Alzheimer’s Disease,
Frank, R.A., Galasko, D., Hampel, H., Hardy, J., de Leon, 36: 145–153.
M.J., Mehta, P.D., et al. (2003). Biological markers for Kepe, V., Ghetti, B., Farlow, M.R. et al. (2010). PET of
therapeutic trials in Alzheimer’s disease. Proceedings of brain prion protein amyloid in Gerstmann-Straussler-
the biological markers working group; NIA initiative on Scheinker disease. Brain Pathology, 20: 419–430.
neuroimaging in Alzheimer’s disease. Neurobiology of Klunk, W.E., Engler, H., Nordberg, A. et al. (2004). Imaging
Aging, 24(4): 521–536. brain amyloid in Alzheimer’s disease with Pittsburgh
Gomperts, S.N., Rentz, D.M., Moran, E. et al. (2008). Compound-B. Annals of Neurology, 55: 306–319.
Imaging amyloid deposition in Lewy body diseases. Klunk, W.E., Price, J.C., Mathis, C.A. et al. (2007). Amyloid
Neurology, 71: 903–910. deposition begins in the striatum of presenilin-1
Harada, R., Okamura, N., Furumoto, S. et al. (2013). mutation carriers from two unrelated pedigrees. The
Comparison of the binding characteristics of [18F]THK- Journal of Neuroscience, 27: 6174–6184.
523 and other amyloid imaging tracers to Alzheimer’s Kudo, Y., Okamura, N., Furumoto, S. et al. (2007).
disease pathology. European Journal of Nuclear 2-(2-[2-Dimethylaminothiazol-5-yl]Ethenyl)-6-(2-[Fluoro]
Medicine and Molecular Imaging, 40: 125–132. Ethoxy)Benzoxazole: A novel PET agent for in vivo
Hashimoto, H., Kawamura, K., Igarashi, N. et al. (2014). detection of dense amyloid plaques in Alzheimer’s
Radiosynthesis, photoisomerization, biodistribution, disease patients. The Journal of Nuclear Medicine,
and metabolite analysis of 11C-PBB3 as a clinically 48: 553–561.
useful PET probe for imaging of tau pathology. The Landau, S.M., Thomas, B.A., Thurfjell, L. et al. and the
Journal of Nuclear Medicine, 55: 1532–1538. Alzheimer’s Disease Neuroimaging, Initiative. (2014).
Ikonomovic, M.D., Abrahamson, E.E., Price, J.C. et al. Amyloid PET imaging in Alzheimer’s disease: A
(2012). Early AD pathology in a [C-11]PiB-negative comparison of three radiotracers. European Journal
case: A PiB-amyloid imaging, biochemical, and of Nuclear Medicine and Molecular Imaging, 41 (7):
immunohistochemical study. Acta Neuropathologica, 1398–1407
123: 433–447. Leyton, C.E., Villemagne, V.L., Savage, S. et al. (2011).
Ikonomovic, M.D., Klunk, W.E., Abrahamson, E.E. et al. Subtypes of progressive aphasia: Application of the
(2008). Post-mortem correlates of in vivo PiB-PET amy- International Consensus Criteria and validation using
loid imaging in a typical case of Alzheimer’s disease. beta-amyloid imaging. Brain, 134: 3030–3043.
Brain, 131: 1630–1645. Lin, K.S., Debenath, M.L., Mathis, C.A. and Klunk, W.E.
Jack, C.R., Jr., Lowe, V.J., Senjem, M.L. et al. (2008). 11C (2009). Synthesis and beta-amyloid binding properties
PiB and structural MRI provide complementary infor- of rhenium 2-phenylbenzothiazoles. Bioorganic and
mation in imaging of Alzheimer’s disease and amnestic Medicinal Chemistry Letters, 19: 2258–2262.
mild cognitive impairment. Brain, 131: 665–680. Lockhart, A., Lamb, J.R., Osredkar, T. et al. (2007). PIB
Jack, C.R., Jr., Wiste, H.J., Lesnick, T.G. et al. (2013). Brain is a non-specific imaging marker of amyloid-beta
beta-amyloid load approaches a plateau. Neurology, (Abeta) peptide-related cerebral amyloidosis. Brain,
80: 890–896. 130: 2607–2615.
Johnson, K.A., Schultz, A., Betensky, R.A., Becker, J.A., Maetzler, W., Liepelt, I., Reimold, M. et al. (2009). Cortical
Sepulcre, J., Rentz, D., et al. (2016). Tau positron PIB binding in Lewy body disease is associated
emission tomographic imaging in aging and early with Alzheimer-like characteristics. Neurobiology of
Alzheimer disease. Annals of Neurology, 79(1): 110 Disease, 34: 107–112.
-119. Maezawa, I., Hong, H.S., Liu, R. et al. (2008). Congo red
Johnson, K.A., Minoshima, S., Bohnen, N.I. et al. and thioflavin-T analogs detect Abeta oligomers.
(2013b). Appropriate use criteria for amyloid PET: Journal of Neurochemistry, 104: 457–468.
A report of the Amyloid Imaging Task Force, the Maruyama, M., Shimada, H., Suhara, T. et al. (2013).
Society of Nuclear Medicine and Molecular Imaging, Imaging of tau pathology in a tauopathy mouse
and the Alzheimer’s Association. Alzheimer’s and model and in Alzheimer patients compared to normal
Dementia:The Journal of the Alzheimer’s Association, ­controls. Neuron, 79: 1094–1108.
9: e-1–e-16. Mathis, C.A., Lopresti, B.J. and Klunk, W.E. (2007).
Johnson, K.A., Gregas, M., Becker, J.A. et al. (2007). Impact of amyloid imaging on drug development in
Imaging of amyloid burden and distribution in Alzheimer’s disease. Nuclear Medicine and Biology,
cerebral amyloid angiopathy. Annals of Neurology, 34: 809–822.
62: 229–234. McKeith, I.G., Dickson, D.W., Lowe, J. et al. (2005).
Kemppainen, N.M., Aalto, S., Wilson, I.A. et al. (2007). Diagnosis and management of dementia with
PET amyloid ligand [11C]PIB uptake is increased in mild Lewy bodies: Third report of the DLB Consortium.
cognitive impairment. Neurology, 68: 1603–1606. Neurology, 65: 1863–72.
Molecular brain imaging in dementia 131

McKhann, G.M., Knopman, D.S., Chertkow, H. et al. (2011). Nyberg, S., Jonhagen, M.E., Cselenyi, Z. et al. (2009).
The diagnosis of dementia due to Alzheimer’s disease: Detection of amyloid in Alzheimer’s disease with
Recommendations from the National Institute on positron emission tomography using [11C]AZD2184.
Aging-Alzheimer’s Association workgroups on diag- European Journal of Nuclear Medicine and Molecular
nostic guidelines for Alzheimer’s disease. Alzheimer’s Imaging, 36: 1859–1863.
and Dementia:The Journal of the Alzheimer’s Okamura, N., Furumoto, S., Fodero-Tavoletti, M.T. et al.
Association, 7: 263–269. (2014a). Non-invasive assessment of Alzheimer’s dis-
McLean, C.A., Cherny, R.A., Fraser, F.W. et al. (1999). ease neurofibrillary pathology using 18F-THK5105 PET.
Soluble pool of Aß amyloid as a determinant of sever- Brain, 137: 1762–1771.
ity of neurodegeneration in Alzheimer’s disease. Okamura, N., Furumoto, S., Harada, R. et al. (2014b). In
Annals of Neurology, 46: 860–866. vivo selective imaging of tau pathology in Alzheimer’s
Mills, S.M., Mallmann, J., Santacruz, A.M. et al. disease with 18F-THK5117. The Journal of Nuclear
(2013). Preclinical trials in autosomal dominant Medicine, 55: 136.
AD: Implementation of the DIAN-TU trial. Revue Okamura, N., Furumoto, S., Harada, R. et al. (2014c).
Neurologique (Paris), 169: 737–743. Characterization of [18F]THK-5351, a novel PET tracer
Mintun, M.A., Larossa, G.N., Sheline, Y.I. et al. (2006). [11C] for imaging tau pathology in Alzheimer’s disease.
PIB in a nondemented population: Potential antecedent European Journal of Nuclear Medicine and Molecular
marker of Alzheimer disease. Neurology, 67: 446–452. Imaging, 41: S260.
Mormino, E.C., Kluth, J.T., Madison, C.M. et al. (2009). Okamura, N., Furumoto, S., Harada, R. et al. (2013).
Episodic memory loss is related to hippocampal- Novel 18F-labeled Arylquinoline derivatives for
mediated beta-amyloid deposition in elderly subjects. ­noninvasive imaging of tau pathology in Alzheimer
Brain, 132: 1310–1323. ­disease. The Journal of Nuclear Medicine,
Morris, J.C. and Price, A.L. (2001). Pathologic correlates of 54: 1420–1427.
nondemented aging, mild cognitive impairment, and Okamura, N., Suemoto, T., Furumoto, S. et al. (2005).
early-stage Alzheimer’s disease. Journal of Molecular Quinoline and benzimidazole derivatives: Candidate
Neuroscience, 17: 101–118. probes for in vivo imaging of tau pathology in
Morris, J.C., Kimberly, A., Quaid, K. et al. (2005). Role of Alzheimer’s disease. The Journal of Neuroscience,
biomarkers in studies of presymptomatic Alzheimer’s 25: 10857–10862.
disease. Alzheimer’s and Dementia:The Journal of the Okamura, N., Suemoto, T., Shiomitsu, T. et al. (2004). A
Alzheimer’s Association, 1: 145–151. novel imaging probe for in vivo detection of neuritic
Morris, J.C., Roe, C.M., Xiong, C. et al. (2010). APOE pre- and diffuse amyloid plaques in the brain. Journal of
dicts amyloid-beta but not tau Alzheimer pathology Molecular Neuroscience, 24: 247–255.
in cognitively normal aging. Annals of Neurology, 67: Okello, A., Koivunen, J., Edison, P. et al. (2009).
122–131. Conversion of amyloid positive and negative MCI to
Naslund, J., Haroutunian, V., Mohs, R. et al. (2000). AD over 3 years: An 11C-PIB PET study. Neurology,
Correlation between elevated levels of amyloid beta- 73: 754–760.
peptide in the brain and cognitive decline. JAMA, 283: Ong, K., Villemagne, V.L., Bahar-Fuchs, A. et al. (201). (18)
1571–1577. F-florbetaben Abeta imaging in mild cognitive impair-
Nelissen, N., Van Laere, K., Thurfjell, L. et al. (2009). Phase ment. Alzheimers Research Therpy, 5: 4.
1 study of the Pittsburgh compound B derivative Ong, K.T., Villemagne, V.L., Bahar-Fuchs, A. et al. (2015).
18F-flutemetamol in healthy volunteers and patients Abeta imaging with 18F-florbetaben in prodromal
with probable Alzheimer disease. The Journal of Alzheimer’s disease: A prospective outcome study.
Nuclear Medicine, 50: 1251–1259. Journal of Neurology, Neurosurgery and Psychiatry,
Nelson, L.D., Siddarth, P., Kepe, V. et al. (2011). Positron 86 (4): 431–436.
emission tomography of brain beta-amyloid and tau Opazo, C., Luza, S., Villemagne, V.L. et al. (2006).
levels in adults with Down syndrome. Archives of Radioiodinated clioquinol as a biomarker for beta-
Neurology, 68: 768–774. amyloid: Zn complexes in Alzheimer’s disease.
Newberg, A.B., Wintering, N.A., Plossl, K. et al. (2006). Aging Cell, 5: 69–79.
Safety, biodistribution, and dosimetry of 123I-IMPY: Ostrowitzki, S., Deptula, D., Thurfjell, L. et al. (2011).
A novel amyloid plaque-imaging agent for the diag- Mechanism of amyloid removal in patients with
nosis of Alzheimer’s disease. The Journal of Nuclear Alzheimer disease treated with gantenerumab.
Medicine, 47: 748–754. Archives of Neurology, 69 (2): 198–207.
Ng, S.Y., Villemagne, V.L., Masters, C.L. and Roew, C.C. Petersen, R.C. (2000). Mild cognitive impairment:
(2007). Evaluating atypical dementia syndromes using Transition between aging and Alzheimer’s disease.
positron emission tomography with carbon 11 labeled Neurologia, 15: 93–101.
Pittsburgh Compound B. Archives of Neurology, 64: Pike, K.E., Savage, G., Villemagne, V.L. et al. (2007).
1140–1144. Beta-amyloid imaging and memory in non-demented
132 Dementia

individuals: Evidence for preclinical Alzheimer’s dis- Rowe, C.C., Pejoska, S., Mulligan, R.S. et al. (2013b). Head-
ease. Brain, 130: 2837–2844. to-head comparison of 11C-PiB and 18F-AZD4694
Price, J.L. and Morris, J.C. (1999). Tangles and plaques (NAV4694) for beta-amyloid imaging in aging and
in nondemented aging and “preclinical” Alzheimer’s dementia. The Journal of Nuclear Medicine, 54:
disease. Annals of Neurology, 45: 358–368. 880–886.
Rabinovici, G.D., Furst, A.J., O’Neil, J.P. et al. (2007). 11C- Rowe, C.C., Ellis, K.A., Rimajova, M. et al. (2010).
PIB PET imaging in Alzheimer disease and frontotem- Amyloid imaging results from the Australian Imaging,
poral lobar degeneration. Neurology, 68: 1205–1212. Biomarkers and Lifestyle (AIBL) study of aging.
Rabinovici, G.D., Jagust, W.J., Furst, A.J. et al. (2008). Neurobiology of Aging, 31: 1275–1283.
Abeta amyloid and glucose metabolism in three Rowe, C.C., Ng, S., Ackermann, U. et al. (2007). Imaging
variants of primary progressive aphasia. Annals of beta-amyloid burden in aging and dementia.
Neurology, 64: 388–401. Neurology, 68: 1718–1725.
Raymond, S.B., Skoch, J., Hills, I.D. et al. (2008). Smart Royall, D.R. (2007). Location, location, location!
optical probes for near-infrared fluorescence imag- Neurobiology of Aging, 28: 1481–1482.
ing of Alzheimer’s disease pathology. European Sabbagh, M., Seibyl, J., Akatsu, H. et al. (2012).
Journal of Nuclear Medicine and Molecular Imaging, Multicentre phase 3 trial on florbetaben for
35 (Suppl. 1): S93–S98. ­beta-amyloid brain PET in Alzheimer’s disease.
Reiman, E.M., Chen, K., Liu, X. et al. (2009). Fibrillar Alzheimer’s and Dementia, 8: 90.
amyloid-beta burden in cognitively normal people at Salloway, S., Sperling, R., Fox, N.C. et al. (2014). Two
3 levels of genetic risk for Alzheimer’s disease. PNAS, phase 3 trials of bapineuzumab in mild-to-­moderate
106: 6820–6825. Alzheimer’s disease. New England Journal of
Rentz, D.M., Locascio, J.J., Becker, J.A. et al. (2010). Medicine, 370: 322–333.
Cognition, reserve, and amyloid deposition in normal Serdons, K., Verduyckt, T., Vanderghinste, D. et al. (2009).
aging. Annals of Neurology, 67: 353–364. Synthesis of 18F-labelled 2-(4’-fluorophenyl)-1,3-­
Resnick, S.M., Sojkova, J., Zhou, Y. et al. (2010). benzothiazole and evaluation as amyloid imaging
Longitudinal cognitive decline is associated with fibril- agent in comparison with [11C]PIB. Bioorganic and
lar amyloid-beta measured by [11C]PiB. Neurology, Medicinal Chemistry Letters, 19: 602–605.
74: 807–815. Shao, X.M., Carpenter, G.M., Desmond, T.J. et al. (2012).
Rinne, J.O., Brooks, D.J., Rossor, M.N. et al. (2010). 11C- Evaluation of [11C]N-Methyl lansoprazole as a radio-
PiB PET assessment of change in fibrillar amyloid-beta pharmaceutical for PET imaging of tau neurofibrillary
load in patients with Alzheimer’s disease treated tangles. ACS Medicinal Chemistry Letters, 3: 936–941.
with bapineuzumab: A phase 2, double-blind, pla- Shoghi-Jadid, K., Small, G.W., Agdeppa, E.D. et al. (2002).
cebo-controlled, ascending-dose study. The Lancet Localization of neurofibrillary tangles and beta-
Neurology, 9: 363–372. amyloid plaques in the brains of living patients with
Roe, C.M., Mintun, M.A., D’Angelo, G. et al. (2008). Alzheimer disease. The American Journal of Geriatric
Alzheimer disease and cognitive reserve: Variation of Psychiatry, 10: 24–35.
education effect with carbon 11-labeled Pittsburgh Small, G.W., Kepe, V., Ercoli, L.M. et al. (2006). PET of
Compound B uptake. Archives of Neurology, brain amyloid and tau in mild cognitive impairment.
65: 1467–1471. New England Journal of Medicine, 355: 2652–2663.
Rowe, C.C. and Villemagne, V.L. (2011). Brain amy- Small, G.W., Kepe, V., Siddarth, P. et al. (2013). PET scan-
loid imaging. The Journal of Nuclear Medicine, ning of brain tau in retired national football league
52: 1733–1740. players: Preliminary findings. The American Journal of
Rowe, C.C., Ackerman, U., Browne, W. et al. (2008a). Geriatric Psychiatry, 21: 138–144.
Imaging of amyloid beta in Alzheimer’s disease with Smid, L.M., Kepe, V., Vinters, H.V. et al. (2013).
18F-BAY94-9172, a novel PET tracer: Proof of mecha- Postmortem 3-D brain hemisphere cortical tau and
nism. The Lancet Neurology, 7: 129–135. amyloid-beta pathology mapping and quantification
Rowe, C.C., Ackerman, U., Browne, W. et al. (2008b). as a validation method of neuropathology imaging.
Imaging of amyloid beta in Alzheimer’s disease with Journal of Alzheimer’s Disease, 36: 261–274.
(18)F-BAY94-9172, a novel PET tracer: Proof of mecha- Sojkova, J. and Resnick, S.M. (2011). In vivo human amy-
nism. The Lancet Neurology, 7: 129–135. loid imaging. Current Alzheimer Research, 8: 366–372.
Rowe, C.C., Bourgeat, P., Ellis, K.A. et al. (2013a). Sojkova, J., Driscoll, I., Iacono, D. et al. (2011). In vivo
Predicting Alzheimer disease with beta-amyloid imag- fibrillar beta-amyloid detected using [11C]PiB positron
ing: Results from the Australian imaging, biomarkers, emission tomography and neuropathologic assess-
and lifestyle study of ageing. Annals of Neurology, ment in older adults. Archives of Neurology, 68:
74: 905–913. 232–240.
Molecular brain imaging in dementia 133

Sperling, R., Johnson, K., Pontecorvo, M.J. et al. (2009a). to 11C-PiB and 18F-FDDNP binding. The Journal of
PET imaging of Beta-amyloid with florpiramine F18 Nuclear Medicine, 50: 1464–1470.
(18F-AV-45): Preliminary results from a phase II study Tolboom, N., Yaqub, M., van der Flier, W.M. et al. (2009b).
of cognitively normal elderly subjects, individuals with Detection of Alzheimer pathology in vivo using both
mild cognitive impairment, and patients with a clini- 11C-PIB and 18F-FDDNP PET. The Journal of Nuclear
cal diagnosis of Alzheimer’s disease. Alzheimer’s and Medicine, 50: 191–197.
Dementia:The Journal of the Alzheimer’s Association, Vandenberghe, R., Van Laere, K., Ivanoiu, A. et al. (2010).
5: P197 [abstract]. 18F-flutemetamol amyloid imaging in Alzheimer dis-
Sperling, R.A., Laviolette, P.S., O’Keefe, K. et al. (2009b). ease and mild cognitive impairment: A phase 2 trial.
Amyloid deposition is associated with impaired default Annals of Neurology, 68: 319–329.
network function in older persons without dementia. Verhoeff, N.P., Wilson, A.A., Takeshita, S. et al. (2004).
Neuron, 63: 178–188. In-vivo imaging of Alzheimer disease beta-amyloid
Sperling, R.A., Jack, C.R., Jr. and Aisen, P.S. (2011). Testing with [11C]SB-13 PET. The American Journal of Geriatric
the right target and right drug at the right stage. Psychiatry, 12: 584–595.
Science Translational Medicine, 3: 111cm33. Villemagne, V.L. and Okamura, N. (2014). In vivo tau
Sperling, R.A., Johnson, K.A., Doraiswamy, P.M. et al. imaging: Obstacles and progress. Alzheimer’s and
(2013). Amyloid deposition detected with florbetapir Dementia:The Journal of the Alzheimer’s Association,
F 18 ((18)F-AV-45) is related to lower episodic memory 10: S254–S264.
performance in clinically normal older individuals. Villemagne, V.L. andRowe, C.C. (2010). Amyloid ligands
Neurobiology of Aging, 34: 822–831. for dementia. PET Clinics, 5: 33–53.
Storandt, M., Head, D., Fagan, A.M. et al. (2012). Villemagne, V.L., Ataka, S., Mizuno, T. et al. (2009a). High
Toward a multifactorial model of Alzheimer disease. striatal amyloid beta-peptide deposition across dif-
Neurobiology of Aging, 33 (10): 2262–2271. ferent autosomal Alzheimer disease mutation types.
Sundgren-Anderson, A.K., Svensson, S.P.S., Swahn, B.M. Archives of Neurology, 66: 1537–1544.
et al. (2009). AZD4694: Fluorinated positron emis- Villemagne, V.L., McLean, C.A., Reardon, K. et al. (2009b).
sion tomography (PET) radioligand for detection of 11C-PiB PET studies in typical sporadic Creutzfeldt-
beta-amyloid deposits. Alzheimer’s and Dementia: Jakob disease. Journal of Neurology, Neurosurgery
The Journal of the Alzheimer’s Association, 5: P267– and Psychiatry, 80: 998–1001.
P268 [abstract]. Villemagne, V.L., Burnham, S., Bourgeat, P. et al. (2013).
Tenovuo, O., Kemppainen, N., Aalto, S. et al. (2008). Amyloid beta deposition, neurodegeneration, and
Posterior cortical atrophy: A rare form of dementia cognitive decline in sporadic Alzheimer’s disease:
with in vivo evidence of amyloid-beta accumulation. A prospective cohort study. The Lancet Neurology,
Journal of Alzheimer’s Disease, 15: 351–355. 12: 357–367.
Thal, D.R., Del Tredici, K. and Braak, H. (2004). Villemagne, V.L., Fodero-Tavoletti, M., Yates, P. et al.
Neurodegeneration in normal brain aging and disease. (2014a). Aβ imaging in aging, Alzheimer’s disease and
Science of Aging Knowledge Environment, 2004 (23): other neurodegenerative conditions. In R.A. Dierckx,
pe26. A. Otte, E.F.J. De Vries and A. Van Waarde (eds.),
Thal, L.J., Kantarci, K., Reiman, E.M. et al. (2006). The PET and SPECT in Neurology. Berlin-Heidelberg:
role of biomarkers in clinical trials for Alzheimer ­disease. Springer-Verlag.
Alzheimer Disease and Associated Disorders, 20: 6–15. Villemagne, V.L., Furumoto, S., Fodero-Tavoletti, M.T.
Thal, D.R., Rub, U., Schultz, C. et al. (2000). Sequence of et al. (2014b). In vivo evaluation of a novel tau imag-
Abeta-protein deposition in the human medial tempo- ing tracer for Alzheimer’s disease. European Journal
ral lobe. Journal of Neuropathology and Experimental of Nuclear Medicine and Molecular Imaging, 41:
Neurology, 59: 733–748. 816–826.
Thompson, P.W., Ye, L., Morgenstern, J.L. et al. (2009). Villemagne, V.L., Fodero-Tavoletti, M.T., Pike, K.E. et al.
Interaction of the amyloid imaging tracer FDDNP with (2008a). The ART of loss: Aβ imaging in the evaluation
hallmark Alzheimer’s disease pathologies. Journal of of Alzheimer’s disease and other dementias. Molecular
Neurochemistry, 109: 623–630. Neurobiology, 38: 1–15.
Thurfjell, L., Lotjonen, J., Lundqvist, R. et al. (2012). Villemagne, V.L., Pike, K.E., Darby, D. et al. (2008b). Aβ
Combination of biomarkers: PET [F]Flutemetamol deposits in older non-demented individuals with cog-
imaging and structural MRI in dementia and mild nitive decline are indicative of preclinical Alzheimer’s
cognitive impairment. Neurodegenerative Diseases, disease. Neuropsychologia, 46: 1688–1697.
10 (1–4): 246–249. Villemagne, V.L., Furumoto, S., Fodero-Tavoletti, M.T.
Tolboom, N., van der Flier, W.M., Yaqub, M. et al. et al. (2012a). The challenges of tau imaging. Future
(2009a). Relationship of cerebrospinal fluid markers Neurology, 7: 409–421.
134 Dementia

Villemagne, V.L., Mulligan, R.S., Pejoska, S. et al. (2012b). Wong, D.F., Rosenberg, P.B., Zhou, Y. et al. (2010). In vivo
Comparison of 11C-PiB and 18F-florbetaben for Abeta imaging of amyloid deposition in Alzheimer disease
imaging in ageing and Alzheimer’s disease. European using the radioligand 18F-AV-45 (florbetapir [corrected]
Journal of Nuclear Medicine and Molecular Imaging, F 18). Journal of Internal Medicine, 51: 913–920.
39: 983–989. Xia, C.F., Arteaga, J., Chen, G. et al. (2013). [(18)F]T807,
Villemagne, V.L., Okamura, N., Pejoska, S. et al. (2012c). a novel tau positron emission tomography imag-
Differential diagnosis in Alzheimer’s disease and ing agent for Alzheimer’s disease. Alzheimer’s and
dementia with lewy bodies via VMAT2 and amyloid Dementia:The Journal of the Alzheimer’s Association,
imaging. Neurodegenerative Diseases, 10: 161–165. 9 (6): 666–676.
Villemagne, V.L., Ong, K., Mulligan, R.S. et al. (2011a). Ye, L., Morgenstern, J.L., Gee, A.D. et al. (2005).
Amyloid imaging with 18F-Florbetaben in Alzheimer Delineation of positron emission tomography
disease and other dementias. The Journal of Nuclear imaging agent binding sites on beta-amyloid pep-
Medicine, 52: 1210–1217. tide fibrils. Journal of Biological Chemistry, 280:
Villemagne, V.L., Pike, K.E., Chetelat, G. et al. (2011b). 23599–23604.
Longitudinal assessment of Aβ and cognition in aging Zetterberg, H. and Blennow, K. (2006). Plasma Abeta
and Alzheimer disease. Annals of Neurology, 69: in Alzheimer’s disease – Up or down? The Lancet
181–192. Neurology, 5: 638–639.
Winblad, B., Palmer, K., Kivipelto, M. et al. (2004). Mild Zhang, W., Oya, S., Kung, M.P. et al. (2005a). F-18
cognitive impairment – Beyond controversies, towards Polyethyleneglycol stilbenes as PET imaging agents
a consensus: Report of the International Working targeting Abeta aggregates in the brain. Nuclear
Group on Mild Cognitive Impairment. Journal of Medicine and Biology, 32: 799–809.
Internal Medicine, 256: 240–246. Zhang, W., Oya, S., Kung, M.P. et al. (2005b). F-18 stil-
Wolk, D.A., Grachev, I.D., Buckley, C. et al. (2011). Association benes as PET imaging agents for detecting beta-
between in vivo fluorine 18-labeled ­flutemetamol amy- amyloid plaques in the brain. Journal of Medicinal
loid positron emission ­tomography imaging and in vivo Chemistry, 48: 5980–5988.
cerebral ­cortical ­histopathology. Archives of Neurology, Zwan, M., Van Harten, A., Ossenkoppele, R. et al. (2014a).
68: 1398–1403. Concordance between cerebrospinal fluid biomarkers
Wolk, D.A., Price, J.C., Madeira, C. et al. (2012). Amyloid and [11C]PIB PET in a memory clinic cohort. Journal of
imaging in dementias with atypical presenta- Alzheimer’s Disease, 41: 801–807.
tion Alzheimer’s and Dementia:The Journal of the Zwan, M.D., Ossenkoppele, R., Tolboom, N. et al. (2014b).
Alzheimer’s Association, 8 (5): 389–398. Comparison of simplified parametric methods for
Wolk, D.A., Price, J.C., Saxton, J.A. et al. (2009). Amyloid visual interpretation of 11C-Pittsburgh compound-B
imaging in mild cognitive impairment subtypes. Annals PET images. The Journal of Nuclear Medicine, 55:
of Neurology, 65: 557–568. 1305–1307.
13
Services for people with dementia

SUBE BANERJEE

of the disease does to them, and between the state, private


13.1 INTRODUCTION and voluntary sectors.
Services for people with dementia are necessarily com-
We have come a long way in the past decade. From obscu- plex and include primary healthcare, specialist services in
rity in terms of policies, dementia is now almost universally mental health (e.g. old age psychiatry), care provided in gen-
acknowledged as one of the most common and serious dis- eral hospitals (e.g. geriatrics and neurology), as well as social
orders faced by the human population. It is the most feared care commissioned and provided by both local authorities,
illness in older people and the most expensive common the voluntary and independent sectors and business out-
condition. One-third of people aged over 65 live and die lets of home care and care homes. Most important of all is
with dementia (Brayne et al., 2006). The need for action to the unpaid care provided by families. There are examples
improve care for people with dementia has been acknowl- of excellent dementia care provided by all of these agencies
edged by international organizations like the World Health and also problematic issues while caring within each of the
Organization (WHO, 2012) and the G8 dementia summit sectors. Strategy and policy evolves and with the growth of
(Department of Health [DH], 2013). The ‘diagnosis gap’ in evidence base the last decade has seen a growing acknowl-
dementia where less than a half of those with dementia ever edgement and understanding of the challenge posed by
attract a diagnosis of dementia, is a global phenomenon dementia and the need for service improvement. This chap-
(Alzheimer’s Disease International [ADI], 2012). Dementia ter aims to look critically at some of the major issues in the
is a challenge at international, national, regional and local development of services for people with dementia through
levels as well as at the personal level. the prism of the provision of memory assessment services
In the last decade it has become clear that there are and the service challenge posed by multimorbidity and
multiple positive interventions that can promote indepen- complexity in dementia.
dence and give people with dementia and their family car-
ers a good quality of life. Things that can enable individuals
and their friends and families to adapt to the challenges
of dementia and steer a course in the 7–12 years that they
13.2 MEMORY ASSESSMENT SERVICES
might be living with dementia, avoiding crises and harm
and promoting wellbeing for all involved are proposed. In terms of service provisions for dementia, one major issue
However, it has also become clear that the large majority of is that, in current systems, less than a half of those suffer-
people with dementia and their family carers do not benefit ing from the illness are ever diagnosed with dementia. Such
from these positive interventions and supports. This makes diagnosis and contact only occurs in the later stages of the
dementia a real priority issue and challenge for service plan- illness or in crisis and very often not at all. In economically
ning and delivery. Services are built on how policies are developed countries, between 50–80% of dementia cases
delivered; services are the vehicles that enable practitioners are unrecognized (Alzheimer’s Disease International, 2011;
to deliver care. Developing and delivering services for those Alzheimer’s Society [AS], 2012). This ‘diagnosis gap’ and
with dementia is more complicated than they are in other the consequent ‘treatment gap’ is even greater in economi-
health conditions such as cancer and heart diseases, because cally developed and underdeveloped countries, with 90%
dementia does not respect the silos we erect between health unidentified cases in India. If these data are extrapolated
and social care, between what families do and what the state worldwide, 28 million (78%) of the 36 million people with

135
136 Dementia

dementia who have not received any diagnosis and there- construction. The assumption often made is that ‘igno-
fore, do not have access to treatment, care and organized rance is bliss’ or that ‘it is best for them not to know, it
support can be provided a formal diagnosis inclusive of all would be too upsetting’. These have major similarities with
these. the excuses made for not telling people they had cancer 40
Much is shared between countries about the challenges years ago. Alzheimer’s Disease International in their 2011
posed by dementia, and while different countries need solu- World Alzheimer Report reviewed in detail all the data
tions that fit and exploit local levers for change, it is strik- available and generated a useful categorization of the ratio-
ing that strategies have emerged in health systems as diverse nale for early diagnosis at an individual level into nine
as those in Japan, Australia, France, Denmark, Taiwan, the broad themes (Box 13.1).
United States and the United Kingdom who all share a com-
mitment to early diagnosis in dementia (http://www.alz.
co.uk/alzheimer-plans). The fourth objective of the French BOX 13.1: World Alzheimer Report 2011:
Plan for Alzheimer is ‘improving access to diagnosis and Nine reasons for early identification and
care pathways’. In the United States, early diagnosis of treatment of dementia
Alzheimer’s disease is one of the six main purposes of the
U.S. National Alzheimer’s Project Act (NAPA) which was 1 Optimizing current medical management
passed with bipartisan support and brought into force by 2 Relief gained from better understanding of
President Obama in January 2011. symptoms
The purpose of memory assessment services is to turn 3 Maximizing decision-making autonomy
people with worrying symptoms into people who know 4 Access to services
what is going on, turning toxic uncertainty into empow- 5 Risk reduction
ered knowledge and enabling access to effective care, sup- 6 Planning for the future
port and treatment (Banerjee, 2015a). For many illnesses 7 Improving clinical outcomes
there are clear and simple ways in which one becomes 8 Avoiding or reducing future costs
aware that there is a problem (e.g. central crushing chest 9 Diagnosis as part of human right
pain or a generalized seizure). This is not the case in
dementia where the onset is insidious, where there may
be a lack of insight and where there are misconceptions In a simpler world there would be an easy diagnostic
about the disease that result in inactivity. Public and test that could be done quickly and simply that would tell
professional misconceptions act to impair help seeking us if we had dementia and diagnose the particular ­subtype.
and help offering. In the National Dementia Strategy for Despite fantastic work in this area, it is still not the case
England (Department of Health, 2009; Banerjee, 2010), yet. In that desired world there would also be an efficient
three such barriers are identified: stigma, the misbelief screening test that would work across populations to iden-
that the symptoms of dementia as a normal part of ageing tify a group with a high likelihood of dementia who could
and the misbelief that there is nothing that can be done then undergo a diagnostic procedure to generate a defini-
for dementia patients. These factors act together to gener- tive diagnosis. Again, this is not the case in the complex
ate the systems that we have worldwide where a person world of dementia. In the absence of a technical fix, we
with dementia is more likely not to have had their condi- are left with an entirely clinical task of making a careful
tion diagnosed than it is for them to have had that diag- broad-based diagnosis based on multidisciplinary assess-
nosis made. ment and formulation of a synthesis of history, examination
A common element of health strategies to improve the and investigation. Tests can help us make decisions about
quality of care of people with dementia is the i­njunction absence or presence of the syndrome of dementia and the
that diagnosis rates should be increased and that d ­ iagnosis accurate diagnosis of the subtype or cause for dementia, but
should be made ‘early’, ‘earlier’ or in a ‘timely’ f­ashion the results cannot make those diagnoses for us. The chal-
(Department of Health, 2008;Nuffield Council on Bioethics, lenge then is how to generate a system that can achieve this.
2009; Burns and Buckman, 2013). In the last couple of Memory assessment services place this function as their
years, there has been debate and discussion about issues core service (Feldman et al., 2003; Banerjee et al., 2007,
surrounding the diagnosis of dementia with questions Brodaty et al., 2011, Banerjee, 2015a).
raised as to the balance of benefits and harms of a d­ iagnosis
or an early diagnosis in dementia, with even the ­suggestion
of a ‘curse of diagnosis’ (Le Couteur et al., 2013). There are
some who state that in the absence of definitive positive evi-
13.3 DEVELOPMENT OF MEMORY
dence as to the benefit of receiving an early and accurate CLINICS AND MEMORY SERVICES
diagnosis, services to deliver this should not be funded.
This statement is made on the basis of a lack of evidence Memory Clinics were first established in the United States,
rather than evidence of dis-benefit. There are strands of in the 1970s, in specialist centres interested in carrying out
both therapeutic nihilism and medical paternalism in this research in Alzheimer’s disease and other dementias. These
Services for people with dementia 137

clinics offered outpatient diagnostic treatment and advice This requires the service to do three things:
for people concerned about their memory. In addition to
general research into the types of dementia, Fraser (1992), 1. To provide a good quality diagnosis in the early stages
describing these early services, identified four core functions: of illness
2. To provide good quality early diagnosis and interven-
1. To forestall deterioration in dementia by early diagno- tion in dementia
sis and treatment 3. To provide this good quality service for all that might
2. To identify and treat disorders other than dementia need it in a given population
that might be contributing to the patient’s problems
3. To evaluate new therapeutic agents in the treatment of So a service without the skills to diagnose the cause or
dementia subtype of dementia and to have a high degree of clini-
4. To reassure people who are worried that they might cal certainty so much so that when a person diagnosed
be losing their memory, when no morbid deficits are with dementia is not misguided and vice versa, would not
found meet these criteria. Equally, the service needs to have the
skill to identify that small group where there is real clinical
The thing that set these memory clinics apart from nor- uncertainty, even after a full multifaceted multidisciplinary
mal neurological, neuropsychiatric or old age psychiatric assessment and where a longitudinal approach to diagnosis
clinics was their specific focus on dementia. Part of their is needed to be sure of the consequences. Using these tests,
rationale was the identification of ‘treatable’ forms of or a service that provides diagnosis but not treatment would
causes for dementia. None was a full-time operation; most also be seen as a failure.
were exactly what they said they were, a clinic or a room Finally, there is the stipulation that this service should
in a specific hospital where once or twice a week there was work for all in a given population. This is based on equity
a specific focus on dementia. The term ‘memory’ seems and access; these should not be services for the lucky few,
to have been used in order to avoid the negative connota- as many memory clinics have been in the past. They need to
tions of the word ‘dementia’ as well as to indicate a broad have the capacity to deal with all incident cases in the area
interest in cognition in a way that might be understood by they serve. An average UK Clinical Commissioning Group
referrers. The first memory clinic in the United Kingdom (CCG) population has 50,000 people over the age of 65 in its
was probably opened at St. Pancras Hospital in London in catchment area. To meet the needs of all, the memory ser-
1983. The numbers of such clinics grew with the passage of vice would need to see people of all ages not just those over
time so much so that it was possible to identify 20 clinics in the age of 65, but the number over 65 is the best population
1995 (Wright and Lindesay, 1995) and 102 clinics in 2002 denominator to make a broad assessment in terms of capac-
(Lindesay et al., 2002) with the authors noting that the ori- ity needed, given the exponential increase in the prevalence
entation of these clinics was moving from being research and incidence of dementia in this age group. If we take the
bases to ways of providing a diagnostic service for demen- incidence rate of dementia in this group at 2% per year, then
tia. The term ‘memory service’ was probably first used in this means that there will be 1000 new cases per year in that
naming the Croydon Memory Service (Banerjee et al., area. If a 70% diagnosis rate is aimed for (allowing for indi-
2007) when the term was coined to describe a new team viduals to opt out of referral and for those who die inciden-
working in south London. There were two reasons for the tally in the first year) and we apply the 64% rate of dementia
coinage of this term. Firstly, they wanted to indicate that in those referred by and found in the Croydon Memory
this should be a service dedicated to serving people with Service (Banerjee et al., 2007), then this means that the ser-
dementia and their carers rather than a clinic organized for vice needs be able to see around 1100 people a year. If the
the convenience of service providers to meet their interests threshold for referral is lowered or the target for diagnosis is
or needs (e.g. to recruit research participants or to pursue a increased then this number would also increase. This would
clinical interest). Secondly, these service providers wanted not be possible with the traditional once-a-week memory
to stress that such services were not about whether they clinic (Wright and Lindesay, 1995; Lindesay et al., 2002) that
represented ‘the clinic’ but were instead about what they sees only a couple of new cases per week.
did (for people with dementia and their carers) ‘the service’. Such services are potentially powerful local agents and
There is a lack of clarity of definition of what constitutes a function as engines for change. They are there to empower
memory assessment service, a memory service and a mem- people with dementia and their carers by providing them
ory clinic. What they all share in common is an interest and the benefits that come from good quality early diagnosis and
skill in the diagnosis of dementia and particularly, in the treatment. They are also there to enable the whole health
early diagnosis of dementia. A simple statement of what a and social care system to work well for dementia. Memory
memory service is for (i.e. its primary aim) is given in the services are, therefore, at the heart of a win-win scenario
National Dementia Strategy for England: where there is quality and value improvement for people
with dementia and carers for health services as well as social
Good-quality early diagnosis and intervention
services. While ambitious, the establishment of services
for all.
those are able to see 1000 to 1500 new patients a year pro
138 Dementia

rata for a catchment area including 50,000 over 65-year-old capacity and skills to provide the range of care needed for
patients, is not impossible. The Croydon Memory Service people with neuropsychiatric complexity in dementia. The
Model evaluation (Banerjee et al., 2007) is a proof of concept separation of ‘organic’ and ‘functional’ disorders is in many
and there are an increasing number of well-resourced mem- ways a false dichotomy for specialist mental health service
ory services that run on most days of the week in the United provision and one that is likely to disadvantage people with
Kingdom that have the ability to deliver quality assessment dementia and their family carers with complex needs. The
at the proposed rate (Doncaster et al., 2011). The paucity of natural history of dementia denotes a substantial propor-
current provision and counsels of despair with respect to tion of those affected will develop challenging behaviour,
funding should not limit our ambition. including symptoms, such as depression, hallucinations and
delusions. Where these are severe and complex, they may
benefit from specialist older people’s mental health services
care. For the system as a whole to work for people with
13.4 ROLE OF PRIMARY CARE IN dementia and their carers, these services need to be effective
DIAGNOSIS OF DEMENTIA and available. The focus of these teams could be conceptual-
ized as the ‘serious mental illness’ element of older people’s
There is an important set of questions concerning the role mental health dealing with urgent and complex disorders
of primary care in the diagnosis of dementia and in par- providing assessment and treatment in the community for
ticular, early dementia. There should not be a conflict about older people with functional and organic disorders. The role
professional protectionism between specialities or cost con- of memory services is a complementary one, of early inter-
tainment. Instead, there should be a discussion about the vention and harm prevention in dementia.
value brought by different approaches to the person with
dementia and their carers, primarily and secondarily to the
system as a whole. The equation will vary between countries 13.6 SERVICES FOR THOSE WITH
in terms of what can and should be done in primary care MULTIMORBIDITY IN DEMENTIA –
and what is best done in secondary care. Different systems DEMENTIA AS AN EXAMPLE
have different training available and different time packages OF INTEGRATIVE HEALTHCARE
that can be spent in assessment. What is vital is that those
SERVICES
who are charged with providing such services should have
the skills needed to do this and do it well. What is possible
in Canada may not be possible in France. In the case of older people with multimorbidity, their
healthcare needs and the extent to which they are met well
or met badly are sharp exemplars of the challenges faced by
13.5 SERVICES FOR THOSE WITH healthcare systems across the world in the twenty-first cen-
NEUROPSYCHIATRIC COMPLEXITY tury (Banerjee, 2015b). The increase in longevity in devel-
IN DEMENTIA – SPECIALIST oped and underdeveloped economies alike is a testament to
the success of twentieth century medicine as well as eco-
PEOPLE’S MENTAL HEALTH
nomic and social development. Research, policy and action
SERVICES has transformed our ability to prevent infant mortality, pre-
vent and treat infectious diseases and more recently, to pre-
One important element in good quality care for people with vent and treat the great killers of middle-aged people, such
dementia is specialist older people’s mental health services. as heart diseases and cancer. This is a fantastic success. It
The rationale for such services being separate from those shows we can make a profound positive difference towards
for adults of working age is based upon the need to have individual and world health. But, with this success comes
the skills in meeting the special needs of the elderly which consequences of the success. With increasing longevity
include the following: comes multimorbidity (people with two or more long-term
conditions) and with multimorbidity comes complexity.
●● Dementia While multimorbidity is not just a problem of older adults,
●● A pattern of multiple pathology its prevalence is much higher in older age groups with 65%
●● Complex co-morbidity of physical and mental health of those aged 65–84 and 82% of those aged 85 or more
with social care needs affected (Barnett et al., 2012).
●● Different patterns of social care and family support Dementia provides a concrete example of a disorder that
illustrates the complexities of the challenges of multimor-
If the system as a whole is to work well for dementia, bidity and the implications of not addressing them in a
this requires effective specialist expertise. There are strong systematic fashion. People with dementia have the highest
arguments for services that have a role that extends beyond level of multimorbidity with data from the Scottish School
dementia, to include responsibility for older adults with of Primary Care (Guthrie et al., 2012) suggesting, that
schizophrenia, depression and mania so that they have the only 5.3% of people diagnosed of dementia have no other
Services for people with dementia 139

long-term condition and that they have, on an average, 4.6 hospitals, the impression given to patients and families of
of the prior mentioned conditions. In an analysis of primary people with dementia is often, that, the patient has failed by
care data from Wishard Health Services in the United States not fitting the service.
(Schubert et al., 2006), people with dementia similarly had, In terms of treatment, the working assumption is that the
on an average, four chronic medical conditions and were optimal treatment of someone with conditions A, B and C
prescribed 5.1 medications; 82% had hypertension and 39% is treatment A + treatment B + treatment C. Clinical guide-
diabetes mellitus. lines for chronic illnesses almost always focus on single
As in other disorders, multimorbidity in dementia pre- disorders even though most with those disorders will have
dicts poor outcomes and poor quality service response. multimorbidity symptom (Guthrie et al., 2012). Questions
People with chronic illness and dementia report fewer can be raised as to whether treatments and services that are
symptoms (McCormick et al., 1994) and undiagnosed but developed on otherwise healthy people work for people with
treatable medical diseases have been reported in almost multiple problems as well. Therapeutic and adverse effects
half of those with dementia (Larson et al., 1984). So why may differ in those with multimorbidity. Dementia provides
do people with multimorbidity in general and those with good example in this regard. In those, with multimorbidity
dementia receive poor quality care? A large part of the of depression and dementia at the same time, antidepres-
answer would seem to lie in the fact that health systems sants that work well appear to have no effect on those with-
are providing twentieth century medicine to a twenty-first out dementia (Rosenberg et al., 2010; Banerjee et al., 2011).
century patient population. General hospitals are increas- In terms of serious adverse effects, the use of antipsychotic
ingly filled with older people with multimorbidity admit- medication in those with dementia has been shown to have
ted in the emergency ward; this is the twenty-first century a greater mortality rate than in those without dementia,
patient population. United Kingdom Hospital Episode with an estimated 1400 extra deaths attributable to their
Statistics show that people over 65 consist of 60% of the usage in the United Kingdom in 2009 (Banerjee, 2009).
admissions, 70% of occupied bed days, 85% of delayed What works for a single disorder does not necessarily work
transfers and 65% of emergency readmissions. Those over in multimorbidity subjects; what is safe for a single disorder
65 of age make up 17% of the UK population but more than is not necessarily safe in multimorbidity patients. We need
2 million unplanned admissions per year, of people over 65, to ensure that national data systems collect data in a way
account for 68% of hospital emergency bed days and the that enables an understanding of the burden, management
use of more than 51,000 acute beds at any one time (Imison and outcomes of those with multimorbidity. We need better
et al., 2012). Almost a quarter of the older people in general research to understand multimorbidity from the biological
hospitals may have dementia and people with dementia in perspective through applied health and social care research.
general hospitals have worse outcomes in terms of length Equally, we need to ensure that evaluative research includ-
of stay, mortality rate and institutionalization. The major- ing randomized controlled trials (RCTs) are conducted in
ity of these patients are not known to specialists providing real life clinical populations that include multimorbidity so
dementia services and are undiagnosed. General hospitals that the data generated can be generalized to those that will
are particularly challenging environments for people with be treated.
memory and communication problems, with cluttered ward The education that we provide our healthcare staff from
layouts, poor signage and other hazards. all professional backgrounds does not prepare them well for
These twentieth century services were generally designed the challenges of multimorbidity or long-term conditions of
for people, often young or middle aged, with one ailment dementia. Current clinical experience is delivered through
and often with a discreet episode of illness. Research, policy a series of discreet, time-limited clinical placements in acute
and health systems in the developed world, particularly, and primary care settings. This provides students with a
have evolved in the past 50 years to generate increasing spe- snapshot of different illnesses in different patients with an
cialization and increasingly deliver technical treatments for emphasis on the acute phase of illness. An example of what
individual conditions. This paradigm of uni-disciplinary might be done in this education is the Time for Dementia
technical super-specialism has grown to dominate policy, Programme (TFD). Funded by Health Education England,
research, practice and education. It casts a dense shadow, Kent, Surrey, Sussex, TFD is a longitudinal clerkship to help
acting to devalue and impair the growth of the generalized students to develop an understanding of the emerging chal-
and integrative focus of much of primary care and geriatric lenges presented by the ageing population, multimorbidity
medicine. The siloed services that it generates in hospitals and long-term conditions using dementia as an exemplar
are those that often fail the frail elderly populations with condition. TFD has been included in the core curricula for
multimorbidity who are the large majority of current patient the 2014 cohort of medical students starting their second
populations and who find services to be confusing, imper- year at Brighton and Sussex Medical School and nursing
sonal and challenging (Mason et al., 2014). Conditions other and paramedic students in their first year at the University
than that of the specialty, with dementia as a good example, of Surrey. All students visited an assigned patient and fam-
are often seen as a complication, ignored or managed by ily every 3–4 months for 24 months from January 2015. The
multiple specialist referral which may be both inefficient focus was on the health and care experience of the person
and ineffective (Wolff et al., 2002; Starfield et al., 2005). In with dementia and their family members. It was located in
140 Dementia

people’s own houses facilitated by the Alzheimer’s Society Banerjee, S. (2009). The Use of Antipsychotic Medication
rather than in a health service and running the programme for People with Dementia: Time for Action. London:
over a 2-year period helped students to gain a long-term Department of Health.
perspective. The programme’s aim was to support an Banerjee, S. (2010). Living well with dementia –
empathetic relationship between students and people with Development of the national dementia strategy for
dementia to enhance their understanding of dementia itself, England. International Journal of Geriatric Psychiatry,
the health, social and family care of people with dementia, 25 (9): 917–922.
what it is like to be ill and old in society, the progression of a Banerjee, S. (2015a). A narrative review of evidence
long-term condition and chronic disease management from for the provision of memory services. International
a patient/family viewpoint. Psychogeriatrics, 27 (10): 1583–1592.
So what would a twenty-first century healthcare that Banerjee, S. (2015b). Multimorbidity – Older adults need
works for those with dementia and physical multimorbid- health care that can count past one. The Lancet, 385:
ity look like? It should be multidisciplinary and integra- 587–589.
tive, valuing generalist skills as well as technical ones. It Banerjee, S., Hellier, J., Dewey, M. et al. (2011). Sertraline
would be patient-centred, focussed in what works for the or mirtazapine for depression in dementia (HTA-
patient not what works for the service. It would draw on the SADD): A randomised, multicentre, double-blind, pla-
insights and expertise of specialists but deliver them in a cebo-controlled trial. The Lancet, 378 (9789): 403–411.
balanced and seamless manner. The core of the challenge Banerjee, S., Willis, R., Matthews, D. et al. (2007).
is the increasing complexity of our clinical population with Improving the quality of care for mild to moderate
dementia as a marker. As Mencken (1982) reminds us, we dementia: An evaluation of the Croydon Memory
need to be wary of simple answers to complex problems that Service Model. International Journal of Geriatric
are wrong. Any solution to these complex set of problems is Psychiatry, 22 (8): 782–88.
likely to be complex in itself, it will require us to continue Barnett, K., Mercer, S.W., Norbury, M. et al. (2012).
with the technical innovation and scientific advancements Epidemiology of multimorbidity and implications for
that has characterized twentieth century medicine and use health care, research, and medical education: A cross-
this along with new ways of organizing to deliver services sectional study. The Lancet, 380 (9836): 37–43.
and make the system work for people with multiple disor- Brayne, C., Gao, L., Dewey, M. and Matthews, F.E. (2006).
ders as they do for those with single illnesses. Dementia before death in ageing societies – The
At a national level there is a need for political imagina- promise of prevention and the reality. PLOS Medicine,
tion, courage and will in re-engineering cherished current 3 (10): e397.
systems and services. The clinical and financial pressures Brodaty, H., Woodward, M., Boundy, K. et al. (2011).
imposed by our current system should, however, be the Patients in Australian memory clinics: Baseline char-
impetus to moving decisively and quickly on this mat- acteristics and predictors of decline at six months.
ter. The tendency to generate systems to fight the last war International Psychogeriatrics, 23 (07): 1086–1096.
rather than the current one is no less a characteristic of Burns, A. and Buckman, L. (2013). Timely Diagnosis
healthcare systems than it is of defence forces. We need of Dementia: Integrating Perspectives, Achieving
to develop our services to meet the actual needs of our Consensus. London: BMA and NHS England.
patients in the present century, not just those of the last Department of Health (2009). Living Well with Dementia:
century. This means a system that works for multimor- A National Dementia Strategy. London: TSO.
bidity. This means making multimorbidity everybody’s Department of Health (2013). Available at https://www.
business and creating policy, commissioning, services, gov.uk/government/publications/g8-dementia-
research and education tools to deliver good quality care summit-agreements. Accessed 28 June 2016.
to those suffering from more than one illness. One way Doncaster, E., McGeorge, M. and Orrell, M. (2011).
of testing if the methods are successful will be to observe Developing and implementing quality standards for
how well these services work for those suffering from memory services: The Memory Services National
dementia. Accreditation Programme (MSNAP). Aging and Mental
Health, 15 (1): 23–33.
REFERENCES Feldman, H., Levy, A.R., Hsiung, G.Y. et al. (2003). A
Canadian cohort study of cognitive impairment and
Alzheimer’s Disease International (2011). World related dementias (ACCORD): Study methods and
Alzheimer Report 2011. London: Alzheimer’s Disease baseline results. Neuroepidemiology, 22 (5): 265–274.
International. Fraser, M. (1992). Memory clinics and memory training. In
Alzheimer’s Disease International (2012). World T. Arie (ed.), Recent Advances in Psychogeriatrics, Vol.
Alzheimer Report 2012. London: Alzheimer’s Disease 2. Edinburgh: Churchill Livingstone, pp. 105–116.
International. Guthrie, B., Payne, K., Alderson, P. et al. (2012). Adapting
Alzheimer’s Society (2012). Mapping the Diagnosis Gap. clinical guidelines to take account of multimorbidity.
London: Alzheimer’s Society. British Medical Journal, 345: e6341–e6341.
Services for people with dementia 141

Imison, C., Poteliakhoff, E. and Thompson, J. (2012). Older Nuffield Council on Bioethics (2009). Dementia: Ethical
People and Emergency Bed Use – Exploring Variation. Issues. London: Nuffield Council on Bioethics.
London: The King’s Fund. Rosenberg, P.B., Martin, B.K., Frangakis, C. et al. (2010).
Larson, E.B., Reifler, B.V., Featherstone, H.J. and English, Sertraline for the treatment of depression in Alzheimer
D.R. (1984). Dementia in elderly outpatients: A prospec- disease. The American Journal of Geriatric Psychiatry,
tive study. Annals of Internal Medicine, 100 (3): 417–423. 18 (2): 136–145.
Le Couteur, D., Doust, J., Creasey, H. and Brayne, C. Schubert, C.C., Boustani, M., Callahan, C.M. et al. (2006).
(2013). Political drive to screen for pre-dementia: Not Comorbidity profile of dementia patients in pri-
evidence based and ignores the harms of diagnosis. mary care: Are they sicker? Journal of the American
BMJ, 347: f5125. Geriatrics Society, 54 (1): 104–109.
Lindesay, J., Marudkar, M., van Diepen, E. and Wilcock, G. Starfield, B., Shi, L. and Macinko, J. (2005). Contribution of
(2002). The second Leicester survey of memory clinics primary care to health systems and health. Milbank Q,
in the British Isles. International Journal of Geriatric 83: 457–502.
Psychiatry, 17 (1): 41–47. Wolff, J., Starfield, B. and Anderson, G. (2002).
Mason, B., Nanton, V., Epiphaniou, E. et al. (2014). ‘My Prevalence, expenditures, and complications of
body’s falling apart.’ Understanding the experiences multiple chronic conditions in the elderly. Archives of
of patients with advanced multimorbidity to improve Internal Medicine, 162: 2269–2276.
care: Serial interviews with patients and carers. BMJ World Health Organization (2012). Dementia: A Public
Supportive and Palliative Care, 6 (1): 60–65. Health Priority. Geneva, Switzerland: World Health
McCormick, W.C. (1994). Symptom patterns and comorbid- Organization.
ity in the early stages of Alzheimer’s disease. Journal of Wright, N. and Lindesay, J. (1995). A survey of memory
the American Geriatrics Society, 42 (5): 517–521. clinics in the British Isles. International Journal of
Mencken, H.L. (ed.) (1982). Mencken Chrestomathy. New Geriatric Psychiatry, 10: 379–385.
York, NY: Random House LLC.
14
Family carers of people with dementia

KATRIN SEEHER AND HENRY BRODATY

typically family members, also friends and neighbours


14.1 FAMILY CAREGIVING (World Health Organization and Alzheimer’s Disease
International, 2012; Alzheimer’s Association, 2014). At least
Family caregiving can be defined in many ways. Schulz and 15 million people in the United States were caring for a
Martire (2004) describe caregiving as: friend or family member with Alzheimer’s disease (AD) or
other dementia in 2013 (Alzheimer’s Association, 2014).
… the provision of extraordinary care, exceeding Family carers, also referred to as caregivers, are the ‘cor-
the bounds of what is normative or usual in family nerstone of support for people with dementia’ (Ferri et al.,
relationships. Caregiving typically involves a sig- 2005). In the developed world, up to 50% of care-associated
nificant expenditure of time, energy and money costs are attributable to informal care provided by family
over potentially long periods of time; it involves members (Hurd et al., 2013; Alzheimer’s Association, 2014),
tasks that may be unpleasant and uncomfortable rather than direct costs for medical or professional nurs-
and are psychologically stressful and physically ing care (Jelic et al., 2000). The role of family carers is even
exhausting (p. 240). more pivotal in low and middle income countries (LMIC),
where they account for 58%–65% of care costs due to low
According to the National Long-Term Care Survey rates of institutionalization and a lack of community ser-
(NLTCS), caregiving is the ‘assistance with at least one vices (World Health Organization and Alzheimer’s Disease
activity of daily living (ADL)’ (Stone et al., 1987; Kasper International, 2012).
et al., 1994), while the American Association of Retired In the United States, most dementia carers are spouses
Persons (AARP) defines caregiving as ‘any assistance pro- or adult children (72%) (Bouldin and Andresen, 2010).
vided for any physical ADL or two or more instrumental Women outnumber men, accounting for 60%–70% of
activities of daily living (IADLs)’ (Wagner, 1997). dementia carers (Bouldin and Andresen, 2010; Fisher et al.,
Other researchers have interpreted caregiving as ‘a con- 2011; Alzheimer’s Disease International, 2014), although
cern for, and taking responsibility for, the well-being of the number of male carers have has nearly doubled since
others’ (Resource Implications Study Group, 2000), or sim- 1996 (Alzheimer’s Association, 2008). The 10/66 Dementia
ply as ‘a continuum ranging from normal social exchanges Research Group (a collective of researchers from the devel-
between fully independent people through various mixes of oping and developed worlds) assessed the care arrange-
assistance’ (Lawton et al., 2000). ments of 706 people with dementia in South East Asia,
China, India, Latin America, the Caribbean and Nigeria
(Prince and the 10/66 Dementia Research Group, 2004). The
14.1.1 WHO ARE FAMILY CARERS? rates of female carers in LMIC ranged from 59% in China
and South East Asia to 95% in Nigeria. As in high-income
Alzheimer’s Disease International estimated that in 2013 countries, wives were most often the primary carers (21% in
there were 44.4 million people with dementia worldwide China and South East Asia to 45% in Nigeria), followed by
(Alzheimer’s Disease International, 2013). For approxi- daughters (15% in India to 40% in Nigeria) and daughters-
mately 75%–80% of people with dementia who live in the in-law (3% in Latin America and the Caribbean to 24% in
community, care is provided informally by unpaid carers India and South Asia). In India and South Asia daughters-
(Schulz and Martire 2004; Alzheimer’s Association, 2014), in-law were the second largest group of carers after wives.

142
Family carers of people with dementia 143

Persons with dementia in LMIC tend to live in larger mul- and they also care for a longer period of time (Alzheimer’s
tigenerational households than those in countries such as Disease International, 2014).
the United States and the United Kingdom (World Health In a study of 1181 UK and European carers (a ­random sam-
Organization and Alzheimer’s Disease International, 2012). ple from Alzheimer Europe’s member organizations fairly
evenly distributed between Germany, Poland, Spain, France
14.1.2 WHY DO THEY CARE? and Scotland), 44% reported spending at least 10 hours a day
assisting the person with dementia (Georges et al., 2008).
Informal carers are motivated to provide care for their The Australian Survey of Disability, Ageing and Carers
loved ones for several reasons, some of which are positive identified that 65% of ~12,000 primary dementia carers spent
and beneficial and others not. Positive reasons include a 40 hours or more per week in their caregiving role (Australian
sense of love or reciprocity, spiritual fulfilment and feel- Institute of Health and Welfare, 2007). Dementia severity
ings of mastery and accomplishment (Eisdorfer, 1991; is the strongest predictor of hours spent on providing care
Sanders, 2005). Carers who identify these reasons experi- (Alzheimer’s Disease International, 2009). Other variables
ence less burden, better health, more positive relationships include time since onset and the amount of instrumental
and more social support (Cohen et al., 2002). Less positive support available to the carer (Taylor et al., 2008).
reasons include, for instance, family responsibility, emo- Estimates of informal care time typically only reflect
tional obligation or ‘having no choice’ (Australian Institute the time spent by the primary carer. Including the input of
of Health and Welfare, 2012). Carers who are motivated by other carers increases the total estimate conservatively by
these reasons are more likely to suffer psychological dis- more than 10% (Neubauer et al., 2008) which may still be
tress, burden and anxiety and to institutionalize the person an underestimate.
with dementia (Pyke and Bengston, 1996; Camden et al., The demands of caregiving in LMIC are comparable to
2011). While the negative aspects of caregiving generally the developed world. On average, carers in LMIC spend
receive more attention in the literature, between 55% and between 2 and 7 hours assisting the person with dementia
90% of carers identified positive motivations for caregiv- (Alzheimer’s Disease International, 2009). Fifty per cent of
ing, including spiritual and personal growth, increased carers from LMIC spend between 3 (India, China, South
faith, accomplishment and mastery (Sanders, 2005). and South East Asia) and 9 (Nigeria) hours per day assisting
A number of variables influence the way carers view the person with dementia (Prince and the 10/66 Dementia
their role. More positive feelings towards caregiving have Research Group, 2004). Living in multigenerational house-
been associated with lower educational level, greater social holds is very common in these countries, but does not
resources, satisfaction with social participation, better decrease carer burden. While there may be a number of
physical health status, being non-Caucasian and being care managers involved, typically only one care provider
older (Haley et al., 1996; Kramer, 1997; Rapp and Chao, performs most of the hands-on care. Additionally, if the
2000). African Americans identified more traditional, col- primary carer does not live with the care recipient, hav-
lectivist reasons for providing care than White Americans ing more people involved appears to create additional ten-
(Dilworth-Anderson et al., 2004, 2005). With the increasing sion and conflict (Prince and the 10/66 Dementia Research
prevalence of male carers, gender differences may be of par- Group, 2004). In some developing countries, particularly
ticular interest, but research to date is lacking and requires African countries, there is a weakening of traditional kin-
further refinement (Baker and Robertson, 2008). ship and support systems. Economic hardship, migration
of younger generations from rural areas and shifting val-
14.1.3 HOW DO THEY CARE? ues and priorities have meant that families are less able or
Family carers can be differentiated not only by their rela- willing to provide care in traditional ways for older people
tionship or living arrangements with the person with (Aboderin, 2004; Okojie, 2004).
dementia (i.e. co-residing or not) but also by their level of
care input. They can be care providers, who facilitate the
14.2 EFFECTS OF DEMENTIA ON
day-to-day hands-on care, or care managers, who organize
the care delivered by others (World Health Organization
CARERS
and Alzheimer’s Disease International, 2012).
Providing care for a person with dementia is very The consequences of dementia caregiving are diverse,
demanding. Primary carers typically spend large propor- ranging from adverse effects on carers’ mental and physi-
tions of their day assisting and supervising care recipients cal health, compromised immune function, as well as
with ADLs (e.g. bathing, toileting) and IADLs (e.g. shop- financial and social disadvantages (Schulz and Martire
ping, food preparation). According to a systematic lit- 2004; Alzheimer’s Association, 2014) to positive effects
erature review, carers spend on average 1.6 hours helping such as uplifts, personal gain, role satisfaction and rewards
with ADLs, 2.1 hours on IADLs and 3.7 hours on general (Kramer, 1997).
supervision (Wimo et al., 2007). Female carers are more The most widely studied negative outcomes for demen-
than twice as likely to provide more hours of care than men, tia carers are carer burden (Pinquart and Sörensen, 2003a;
144 Dementia

Torti et al., 2004; Etters et al., 2008; van der Lee et al., 2014) and and a 23% higher level of stress hormones and a 15% lower
increased levels of depressive symptoms or distress (Pinquart level of antibody responses than non-carers (Vitaliano et al.,
and Sörensen 2003a, 2003b; van der Lee et al., 2014). 2003). The rates of higher numbers of chronic conditions,
greater health service utilization, compromised immunity,
14.2.1 CARER BURDEN cardiovascular and metabolic diseases were small and did
not reach statistical significance but given the large num-
Carer burden refers to the subjective burden specific to the ber of caregivers in the population have important implica-
experiences of carers. The Zarit Burden Interview (ZBI) tions (Vitaliano et al., 2003). The reasons for poorer health
(Zarit et al., 1980) is the most widely used measure of carer may be physiological or because carers are less likely to
burden in dementia carers (Knight et al., 2000; O’Rourke engage in preventative health behaviours such as exercise,
and Tuokko, 2003; Bachner and O’Rourke, 2007). Scores and are more likely to smoke, drink alcohol and soft drinks,
range from 0 to 88 with higher scores reflecting greater bur- eat fast food, and have poor sleep patterns, thereby placing
den and statistically derived cut-points of 24/26 out of 88 themselves at risk of further health problems (Schulz and
indicating high burden (Schreiner et al., 2006). Williamson, 1997; Hoffman et al., 2012).
Bachner and O’Rourke (2007) pooled ZBI data across
138 studies and reported a mean ZBI score of 33.6 (stan- 14.2.4 SOCIAL ISOLATION
dard deviation = 12.1). The 10/66 Dementia Research Group
also found high levels of carer burden in population-based About one-third of dementia carers report feeling socially iso-
samples from LMIC, with mean ZBI scores ranging from lated (Alzheimer’s Association, 2014) as they tend to sacrifice
17 to 28 (Prince et al., 2012). their leisure pursuits and hobbies, restrict time with friends
and family and give up or reduce employment (Brodaty and
14.2.2 PSYCHOLOGICAL MORBIDITY Hadzi-Pavlovic, 1990; LoGiudice et al., 1999; Leong et al.,
2001). Distance from family and friends can compound carer
The strain of caregiving can manifest as psychological mor- isolation. In one study, half the carers had contact with some-
bidity, particularly depression and anxiety (Cooper et al., one outside their household once per week or less and 13%
2006; Campbell et al., 2008; Gaugler et al., 2008). In devel- had no personal contacts outside the home in the previous 2
oped countries, rates of depression among dementia carers weeks (Brodaty and Hadzi-Pavlovic, 1990). Social isolation,
range from 23% to 85% (Adkins, 1999; Clare et al., 2002; on the other hand, is a risk factor for carer burden (Adelman
Schoenmakers et al., 2010a). According to a systematic litera- et al., 2014) and depression, particularly for female carers
ture review, 22% of dementia carers meet diagnostic criteria (Alzheimer’s Association, 2014). Interventions may assist in
for major depressive disorder, with rates for individual studies decreasing/preventing social isolation in carers by increasing
ranging from 15% to 32% (Cuijpers, 2005). The rates of anxi- the number of support persons for carers, their satisfaction
ety vary between 16% and 45% (Schulz et al., 1995; Livingston with their support network, and the assistance they receive
et al., 2005; Cooper et al., 2006, 2007). In developing coun- with caregiving (Roth et al., 2005).
tries rates of psychiatric morbidity range from 40% to 75%
(Prince and the 10/66 Dementia Research Group, 2004). 14.2.5 FINANCIAL IMPACT
Compared to other carers, dementia carers experience
higher levels of psychological distress and lower levels of In 2013, the monetary contribution of informal carers
self-efficacy and subjective well-being (Schulz and Martire to the overall costs of dementia in the United States was
2004). These differences are even larger when compared to $220 billion (Alzheimer’s Association, 2014). Almost 60%
non-carers (Pinquart and Sörensen, 2003b; Schoenmakers of U.S. dementia carers are also employed of whom two-
et al., 2010a) with effect sizes on meta-analysis ranging from thirds reported that they missed work and 8% reported
0.40 (95% confidence interval [CI]: 0.32–0.42) for subjective that they turned down promotion opportunities. Up to
well-being to 0.66 (95% CI: 0.49–0.84) for clinician-rated 31% had given up work to attend to caregiving responsi-
depression (Pinquart and Sörensen, 2003b). bilities (Alzheimer’s Association and National Alliance
for Caregiving, 2004; Alzheimer’s Association, 2007).
14.2.3 PHYSICAL MORBIDITY In Australia, 56% of all dementia carers are employed,
although rates are lower among primary dementia car-
Carers report more health problems and worse perceived ers (39%) (Australian Institute of Health and Welfare,
overall health compared to controls, with meta-analytic 2012). The most common reason for leaving the work-
effects ranging from 0.15 (95% CI: 0.09–0.22) for objec- force, stated by 74%, is a lack of alternative care arrange-
tive to 0.20 (95% CI: 0.12–0.28) for subjective health ments (Australian Institute of Health and Welfare, 2012).
measures. Dementia carers compared to other carers or Female carers are almost seven times more likely than
non-carers reported particularly poor physical health men to reduce their employment from full-time to part-
(B = 0.15, p < 0.001) (Pinquart and Sörensen, 2003b). time; and more than twice as many female carers give up
Dementia caregiving has also been associated with nega- work entirely or lose job benefits compared to male carers
tive objective health measures such as greater medication use (Alzheimer’s Association, 2014).
Family carers of people with dementia 145

In the developing world, the costs of dementia in com- literature review (van der Lee et al., 2014) identified four
parative terms represent a greater burden than in the devel- theoretical frameworks that are used most consistently in
oped world, and the economic disadvantage associated with the literature to explain the development of carers’ stress:
caregiving is significant (Prince and the 10/66 Dementia the Transactional Model of Stress and Coping (Lazarus
Research Group, 2004). On average, 32% of carers in LMIC and Folkman, 1984); the Stress Process and Coping Model
cut back on paid work to care for a family member with (Haley et al., 1987, 1996); the Two-Dimensional Model of
dementia and very few were able to access compensatory Psychosocial Morbidity (Poulshock and Deimling, 1984); and
financial support such as government pensions. While the Stress Process Model of Alzheimer’s Disease Caregiver’s
healthcare services are cheaper in absolute dollars than in Stress (Pearlin et al., 1990). These frameworks have in com-
developed countries, in relative terms these families spend mon that they incorporate both patient characteristics and
a greater proportion of their income on healthcare for the dementia-specific measures on one side and carer charac-
person with dementia. Paradoxically, despite their eco- teristics and carer resources on the other.
nomic disadvantage, carers from poorer countries tend to As an example, Pearlin’s Stress Process Model (Pearlin
use the more expensive services of private doctors as public et al., 1990) is depicted schematically in Figure 14.1. The
services are unsatisfactory (Prince and the 10/66 Dementia model proclaims six distinct groups of stressors: (1) contex-
Research Group, 2004). tual or background variables (i.e. patient and carer sociode-
mographic details, relationship characteristics; caregiving
context); (2) objective primary stressors (i.e. impairment
indicators such as patient cognition, function and behav-
14.3 PREDICTORS OF AND
iour); (3) subjective primary stressors (i.e. carer’s subjective
PROTECTORS FROM CARER
appraisals of objective stressors); (4) secondary role strains
DISTRESS (i.e. family conflict, job-caregiving conflicts, economic
problems, constrictions of social life. Secondary stressors
Several characteristics of the carer and/or care-recipient are no less potent than primary stressors, but merely result
have been linked to increased levels of depression, carer from trying to ­balance primary stressors and other non-
burden or other negative outcomes in those caring for dementia-related conflicts/conditions); (5) intrapsychic/
family members with dementia (Torti et al., 2004; Etters intrapersonal strains (i.e. low self-esteem, poor mastery,
et al., 2008) or mild cognitive impairment (Seeher et al., role captivity, poor health); and (6) mediators of stress/pro-
2013). tective factors (i.e. coping, social support).
Negative carer outcomes are best analyzed in the context The scientific evidence (mainly based on meta-analyses
of multidimensional stress development models (Brodaty and systematic reviews) for each of these areas of stress is
and Donkin, 2009; Noyes et al., 2010). A recent systematic summarized next.

Context / background of stress

PWD: Demographic details, disease duration


Carer: Demographic/relationship characteristics
Direct effects
Primary objective stressors Indirect/mediated effects
Moderated effects
PWD: Cognition, function, behaviour

Primary subjective stressors

Carer: Appraisal of objective stress, role


Protective factors

overload, relational deprivation

Secondary role strain Coping


Social support
Carer: Family conflict, job-caregiving
conf licts

Intrapersonal strains
Carer: Self-esteem, mastery, loss of self, role captivity,
competence, gain

Outcome of stress

Carer: Depression, anxiety, yielding of role, physical health etc.

Figure 14.1 The stress process model. PWD: person with dementia (Adapted from Pearlin, L.I. et al., The Gerontologist,
30, 583–594, 1990.)
146 Dementia

14.3.1 CONTEXT OF BACKGROUND OF (Kinney and Stephens, 1989; Goode et al., 1998; Lévesque
STRESS et al., 1998).

Meta-analytic and pooled evidence consistently suggest 14.3.3 SECONDARY STRESSORS


that female carers report worse carer outcomes than men,
including but not limited to higher levels of depression, Secondary carer strains include conflicts within the fam-
anxiety and carer burden (Pinquart and Sörensen, 2003a, ily; struggling with balancing work, family and caregiving
2006a; Schoenmakers et al., 2010a). Likewise, older carer demands; economic problems; constrictions to one’s social
age and being a spousal carer have been linked to more life, but also carer characteristics such as low self-esteem,
negative carer outcomes (Pinquart and Sörensen, 2003a; poor mastery or role captivity.
Schoenmakers et al., 2010a; Pinquart and Sörensen, 2011). Carers who have multiple roles (e.g. being a working
Mixed evidence exists for living arrangements, with some mother also looking after a parent with dementia) are more
studies reporting positive associations between living with likely to experience negative carer outcomes (e.g. Stephens
the person with dementia and more negative carer outcomes et al., 2001; Edwards et al., 2002).
while others do not find such a link (Schulz et al., 1995). Likewise, carers who report high levels of neuroticism,
Providing care for a male person with dementia has been low optimism and negative expressed emotion, and who
linked to greater burden (Torti et al., 2004; Rosdinom et al., have insecure attachment styles experience higher levels of
2013), but not consistently (Abdollahpour et al., 2012). At distress (Schulz et al., 1995; Nomura et al., 2005; Campbell
the same time, younger patient age is associated with greater et al., 2008; Cooper et al., 2008) and more burden (van der
carer depression (Covinsky et al., 2003) and higher levels of Lee et al., 2014). Recently, carer neuroticism, particularly in
burden (Torti et al., 2004). This is most likely due to higher adult children, has been linked to greater cognitive decline
levels of burden reported by carers of people with young- in the PWD (Norton et al., 2013). At the same time, better
onset dementia (YOD), although the evidence is not conclu- the person with dementia and secure attachment style are
sive (van Vliet et al., 2010). Similarly, carers of people with protective against distress and burden (Cooper et al., 2008;
frontotemporal dementia (FTD) experience significantly Lopez and Crespo, 2008). Similarly, high levels of mastery
higher levels of burden than AD carers (Reidijk et al., 2006), are linked to lower levels of depression (Schulz et al., 1995)
while carers of people with AD, vascular or Lewy body and reduced stress reactions (Roepke et al., 2008). However,
dementia report similar levels of stress (Draper et al., 1992; mastery has been shown to deteriorate for some AD car-
Lowery et al., 2000). However, these generalizations are not ers (Aneshensel et al., 1995; Skaff et al., 1996; Pearlin et al.,
undisputed and effects may in fact be mediated by other 2007) and a lack of mastery or confidence in one’s ability in
important stressors such as patient problem difficult. the caregiving role is associated with increased strain and
burden for carers (Campbell et al., 2008). Role captivity, or
14.3.2 PRIMARY OBJECTIVE AND feeling ‘trapped’ in one’s role as a carer predicts burden levels
SUBJECTIVE STRESSORS and psychological ill health as it erodes the carer’s sense of
self and subsequently causes feelings of resentment towards
Negative carer outcomes are most consistently and strongly the patient (Pearlin et al., 1990; Campbell et al., 2008).
linked to the presence of behavioural and psychological
symptoms of dementia (BPSD). Moderately strong associa- 14.3.4 MEDIATORS OF STRESS
tions with carer burden and depression have been reported
in meta-analyses and systematic reviews (Schulz et al., 1995; Dysfunctional and immature coping strategies (largely
Pinquart and Sörensen, 2003a; Black and Almeida, 2004; emotion-based strategies such as escape avoidance) are
Etters et al., 2008; Schoenmakers et al., 2010a; Ornstein and mildly to moderately associated with increased anxiety
Gaugler, 2012; van der Lee et al., 2014). and greater distress for carers (Cooper et al., 2006, 2007;
The evidence is less conclusive for patients’ cogni- del-Pino-Casado et al., 2011; van der Lee et al., 2014). The
tive symptoms. Links are generally weaker than for BPSD pooled weighted mean correlations derived from a recent
(Schulz et al., 1995; Pinquart and Sörensen, 2003a; Etters meta-analysis are 0.39 (95% CI: 0.28–0.50) and 0.46 (95%
et al., 2008; Schoenmakers et al., 2010a) or may not be pres- CI: 0.36–0.56), respectively (Li et al., 2012). Greater levels
ent at all (van der Lee et al., 2014). of emotional support and acceptance-based coping were
The same is true for patients’ functional dependency. mildly protective, with the weighted mean correlation rang-
Only 36% of carer burden studies and 21% of carer ing from −0.22 (95% CI: −0.26 to −0.18) for carer anxiety to
depression studies reported a significant association with −0.20 (95% CI: −0.28 to −0.11) for depression (Li et al., 2012).
patients’ functional dependence (van der Lee et al., 2014). Problem/solution-focused coping strategies (largely cogni-
If associations are found, they are generally weaker than for tively based strategies such as positive reframing) can mitigate
BPSD (Schulz et al., 1995; Pinquart and Sörensen, 2003a; distress for carers (Lazarus and Folkman, 1984; Kneebone
Schoenmakers et al., 2010a). In addition, carers’ nega- and Martin, 2003; Cooper et al., 2007; del-Pino-Casado et al.,
tive appraisal of primary stressors has strong detrimental 2011; van der Lee et al., 2014). However, this effect did not
effects on their mental health and psychological well-being withstand meta-analytic scrutiny. The pooled effects on carer
Family carers of people with dementia 147

anxiety and depression were 0.10 (95% CI: −0.02–0.21) and be institutionalized, if their carers are employed and balanc-
−0.04 (95% CI: −0.11–0.04), respectively (Li et al., 2012). Some ing too many demands, unemployed and financially unsta-
cognitive coping styles, such as cognitive confronting were ble, feel burdened by the carer role, have difficulty dealing
even associated with higher levels of anxiety, while others, with behavioural manifestations of dementia, are in a poorer
such as ‘planful problem-solving’ and ‘cognitive confronting’ relationship with the person with dementia or have poorer
showed inconsistent results (Cooper et al., 2007). physical and psychological health (Brodaty et al., 1993; Tun
Carer support can be formal or informal and can come et al., 2007; te Boekhorst et al., 2008; Gaugler et al., 2009).
in the form of instrumental, emotional or informational
support. Formal support is provided by organizations such
as community services or aged care facilities while informal
support is provided by family and friends. Instrumental 14.5 CARER INTERVENTIONS
support includes help with daily living needs and house-
work; emotional support provides a ‘shoulder to cry on’; and A plethora of interventions, therapies and services have been
informational support includes information and knowledge devised to buffer family carers from the negative effects that
from both health professionals and other carers who have dementia caregiving has on their health and well-being (Zarit
experienced similar situations. and Femia, 2008). Interventions can be broadly categorized
The evidence for receiving support is mixed, but support into psychological interventions including psychoeduca-
is generally regarded as being protective (Schulz et al., 1995; tion and psychotherapy such as skills training, provision of
Shrout and Bolger, 2002; Kneebone and Martin, 2003; van structured information, cognitive behavioural therapy (CBT)
der Lee et al., 2014) as it provides a buffer against burden and and/or counselling; educational/information interventions
stress for carers by increasing the perception that resources which purely involve information provision and demen-
are available to handle stress (Cohen, 2004). However, not all tia education; or support interventions including support
support is beneficial; inappropriate or unwelcome support groups, home visits and social support. Often interventions
can be more stressful than helpful (Edwards and Cooper, involve psychological and support components (Parker et al.,
1988). Satisfaction with social support requires the percep- 2008; Zarit and Femia, 2008). Traditionally, interventions
tion by the carer that the right type of support is available at have been delivered face-to-face in individual or group set-
the desired level (Drentea et al., 2006). Carers who were more tings. However, Internet-based interventions have become
satisfied with their social interactions showed fewer nega- more common and may be beneficial for some but not all
tive psychological ­symptoms (Serrano-Aguilar et al., 2006). carers (Powell et al., 2008). Alzheimer Associations are
another important form of help and support for dementia
carers. Support organisations for people with dementia and
their carers exist in over 85 countries worldwide (www.alz.
14.4 NURSING HOME ADMISSION AND co.uk). These are discussed further in Chapter 36. Referral of
CARERS families to Alzheimer Associations is part of the clinician’s
therapeutic armamentarium. Respite care and community
Carers may choose to place the person with dementia in a services also offer support to carers. These are discussed in
nursing home as the illness progresses, usually when the care- Chapter 15.
giving demands become overwhelming (Gaugler et al., 2009). Table 14.1 lists meta-analyses and systematic literature
While institutionalization reduces the direct obligations on reviews that have analyzed the efficacy of carer interven-
the carer and can provide relief and reduced stress (Brodaty tions in improving measures of carer mental health and
and Hadzi-Pavlovic, 1990; Aneshensel et al., 1995), it does not psychological well-being. Intervention effects on patient
necessarily remove distress. Following nursing home admis- symptoms and the ability to delay nursing home placement
sion, carers may experience increased guilt, anger, anxiety, are summarized in Table 14.2. Generally, there is no ‘one
depression, loss of self and financial difficulties (Tornatore size fits all’ solution in improving carer mental health or
and Grant, 2002; Schulz et al., 2004). These can persist for up patient symptoms. The efficacy of interventions depends
to several years following institutionalization (Gaugler et al., highly on the outcome one seeks to improve.
2007). It was hypothesized that distress would be less for car- Depressive symptoms of dementia carers respond best
ers if the person with dementia was placed in group homes to CBT (Pinquart and Sörensen, 2006b; Vernooij-Dassen
that had smaller numbers, more informal daily routines and et al., 2011) and psychoeducational group interventions
more decision-making involvement of residents and informal (Thompson et al., 2007), both showing moderately strong
carers, compared with traditional nursing homes. However, effects (Table 14.1). The effects of decreased carer anxiety
there were no significant differences between carers of 67 are more modest (effect size around −0.21) with the best
care recipients in group homes and of 97 in nursing homes evidence currently existing for CBT (Vernooij-Dassen
on measures of competence, psychopathology and burden (te et al., 2011). Carer burden is more difficult to improve,
Boekhorst et al., 2008). with effects generally being small or not statistically sig-
Demographic and psychosocial factors predict nursing nificant possibly because studies do not always distinguish
home admission. Persons with dementia are more likely to between objective and subjective burden (Brodaty et al.,
148 Dementia

Table 14.1 Overview of the meta-analytic and systematic review evidence for the effectiveness of interventions on carer
outcomes

Psych. Subj.
Reference morb. Depression Anxiety Burden stress Know­ledge Coping SE SWB/QoL Other

Any psychosocial (between 5 and 98 interventions)


Pinquart and −0.24z −0.12 0.46 0.31
Sörensen (2006b)a
Brodaty et al. (2003)a 0.31 0.09 0.51 Any: 0.32

Psychoeducation (between 4 and 34 interventions)


Pinquart and −0.27 −0.15 0.46 0.21
Sörensen (2006b)a
Thompson et al. −0.71 −2.15
(2007)a
Selwood et al. (2007)b n/e n/e n/e

CBT (between 1 and 15 interventions)


Pinquart and −0.70 −0.36 1.12 0.37
Sörensen (2006b)a
Vernooij-Dassen et al. −0.66 −0.21 0.14 −0.24 0.64
(2011)c
Schoenmakers et al. 0.03 −2.94
(2010b)a

Counselling/case management (between 2 and 7 interventions)


Pinquart and −0.20 −0.50 0.43 0.42
Sörensen (2006b)a
Thompson et al. −0.21 0.37
(2007)a
Schoenmakers et al. −0.32
(2010b)a

Coping skills training (between 2 and 5 interventions) DFC PC


Li et al. (2012)ad 0.39d 0.28
Selwood et al. (2007)b m.d.n.

Meditation (between 5 and 8 interventions)


Hurley et al. (2014)b 5/7e+1fU 3/5eg 3/4ef

Technology based (Internet, computer, telephone) (between 2 and 6 interventions)


Thompson et al. 0.62
(2007)a
Schoenmakers et al. 0.07
(2010b)a
Boots et al. (2014)b 1/2e 2/3e 2/4eh 2/4i n.s. 5/6e n.s.
Powell et al. (2008)b 1/5e 0/1e 0/4e 1/1e Social: 0/3e

Information provision services (e.g. dementia advisory services, AA) (between 1 and 4 interventions)
Corbett et al. (2012)a −0.87 0.23–0.43

Support groups (between 2 and 19 interventions)


Pinquart and 0.05 0.01 0.29 2.03
Sörensen (2006b)a
Chien et al. (2011)a −0.44 −0.40 −0.23 −0.44 Social: 0.40
Thompson et al. −0.40
(2007)a
Selwood et al. (2007)b n/e n/e n/e
(Continued )
Family carers of people with dementia 149

Table 14.1 (Continued ) Overview of the meta-analytic and systematic review evidence for the effectiveness of interventions
on carer outcomes

Psych. Subj.
Reference morb. Dep­ression Anxiety Burden stress Know­ledge Coping SE SWB/QoL Other

Physical activity (between 2 and 4 interventions) Obj


burden:
Orgeta and Miranda- −0.17 −0.22 −0.22−0.43j –0.18 −0.22
Castillo (2014)a

Behaviour management interventions for carers (between 3 and 17 interventions)


Brodaty and 0.15
Arasaratnam
(2012)a
Selwood et al. (2007)b ++k n/e
ind 6+
Selwood et al. n/e n/e
(2007)bind <6/G

Dyadic interventions (up to 20 interventions)


Van’t Leven et al. 3/9e(0 to 13/17e(−1.19 7/10e
(2013) b −0.98) to 0.31) (−0.15 to
0.92)

Respite (between 2 and 12 interventions)


Pinquart and Sörensen −0.12 −0.26 −0.06 0.27
(2006b)a
Schoenmakers et al. 0.30l
(2010b)a
Maayan et al. (2014)c n/d n/d 1m

Training of the care recipient (between 2 and 8 studies)


Pinquart and 0.01 −0.17 −0.12 0.42
Sörensen (2006b)a

Pharmacological treatment of the care recipient (between 4 and 8 interventions) Carer time:
Schoenmakers et al. −0.27 −42 min/d
(2009)a
Lingler et al. (2008)a −0.18 0.15

Notes: Bold values are statistically significant pooled effects; shaded effects are the strongest significant effect for the respective carer outcome;
effects are pooled standardized mean differences; effect size 0.2: small effect; effect size 0.5: moderate effect; effect size ≥0.8: strong effect.
Abbreviations: AA, Alzheimer’s Associations; CBT, Cognitive behavioural therapy; DFC, dysfunctional coping; ind 6+, therapies for individuals
with six or more sessions; ind<6/G, therapies for individuals with less than six sessions or group therapy; m.d.n, possibly beneficial,
more data needed; n/e, not effective; n/d, no data; NH, nursing home; n.s., not significant; Obj, objective; PC, positive coping; Psych.
Morb., psychiatric morbidity; QoL, quality of life; SWB, subjective well-being, including quality of life; SE, self-efficacy; Sx, symptoms.
a Meta-analysis.

b Systematic literature review.

c Cochrane review.

d Meta-analysis of interventions that significantly improved carer depression to analyze the extent to which they also changed coping styles.

Counterintuitively, these interventions increased dysfunctional coping.


e x/y: Ratio of significant effects to the total number of studies that investigated such effect.

f Follow-up.

g Post-treatment.

h One additional study found significant positive effects for high-frequent users of the programme.

i x/y: Reported increase in x of y studies.

j Based on studies using the same measure (i.e. SCB, Screen for Caregiver Burden).

k Short term/long term.

l Favouring controls.

m Negative.

++,effective.
150 Dementia

Table 14.2 Overview of the meta-analytic evidence for the effectiveness of interventions on patient outcomes

Reference BPSD Mood ADL/IADL QoL NH placement

Any psychosocial carer intervention (between 5 and 98 interventions)


Pinquart and Sörensen (2006b)a −0.17 0.72
Brodaty et al. (2003)a 0.68

Psychoeducation of carer (between 4 and 34 interventions)


Pinquart and Sörensen (2006b)a −0.15 1.09

CBT for carer (between 1 and 15 interventions)


Pinquart and Sörensen (2006b)a −0.29 n/d
Vernooij-Dassen et al. (2011)b n.s.

Carer counselling/case management (between 2 and 7 interventions)


Pinquart and Sörensen (2006b)a −0.33 n/d

Behaviour management interventions for carers (between 3 and 17 interventions)


[Brodaty and Arasaratnam 0.34
(2012)]a

Information provision services (e.g. dementia advisory services, AA) (1 to 4 interventions)


Corbett et al. (2012)a −1.48 n.s.

Carer support groups (between 2 and 19 interventions)


Pinquart and Sörensen (2006b)a 0.07 0.89

Dyadic interventions (up to 20 interventions)


Van’t Leven et al. (2013)c d3/8 3/6
d 5/10
d 4/8
d

(−0.66 to 0.27) (−0.47 to 0.34) (−1.46 to 0.23) (−0.26 to 1.27)

Training of the care-recipient (between 2 and 8 interventions)


Pinquart and Sörensen (2006b)a −0.35

Respite (between 2 and 12 interventions)


Pinquart and Sörensen (2006b)a −0.08 0.76
Orgeta and Miranda-Castillo n/d
(2014)b
Notes: Bold values indicate significant pooled effects; effects are pooled standardized mean differences, except effects on nursing home
placement which are odds ratios; effect size 0.2: small effect; effect size 0.5: moderate effect; effect size ≥0.8: strong effect.
Abbreviations: ADL, activities of daily living; BPSD, behavioural and psychological symptoms of dementia; BPSD dis, carer distress associated
with BPSD; IADL, instrumental activities of daily living; n/e, not effective; NH, nursing home; QoL, quality of life; Sx, symptoms.
a Meta-analysis.

b Cochrane review.

c Systematic literature review.

d x/y, ratio of significant effects to the total number of studies that investigated such effect

2003). However, one meta-analysis yielded moder- distress (ES = 0.15, 95% CI: 0.04–0.26; Table 14.1) (Brodaty
ately strong effects for counselling or case management and Arasaratnam, 2012).
(Pinquart and Sörensen, 2006b). Physical activity interven- Past carer intervention studies were often limited in their
tions may also provide relief from carer burden, although design and methodology, contributing to the modest effects
these results are based on two interventions only (Orgeta traditionally obtained (Zarit and Femia, 2008). More recent
and Miranda-Castillo, 2014). Carer knowledge is best studies have addressed these shortcomings by ensuring that:
improved by psychoeducational interventions (Pinquart carers’ needs are clarified, the treatment is relevant to car-
and Sörensen, 2006b), while carers’ subjective well-being ers’ needs, the methodology meets the treatment goal and
benefits most from support group interventions (Chien et the heterogeneity of carers is taken into account (Zarit and
al., 2011). Non-pharmacological interventions for patients Femia, 2008). A selection of carer interventions is briefly
with BPSD which are delivered through the carer at home described below.
significantly decrease patient BPSD (ES (effect size) = 0.34, The Seattle Protocols (Reducing Disability in Alzheimer’s
95% CI: 0.20–0.48; see Table 14.2) and the associated carer disease, RDAD) are a systematic and structured, yet
Family carers of people with dementia 151

individualized approach to training family carers to reduce reasons for varying effects on nursing home admission across
behavioural and psychiatric disturbances in persons with the three countries were differences in aged care systems and
dementia. The protocols teach carers to monitor problems, financial disincentives to institutionalization and differences
identify possible events that trigger disturbances and develop in the amount of counselling provided (more ad hoc counsel-
effective responses (Teri et al., 2005). They have been effective ling was provided in Sydney) (Brodaty et al., 2009).
at improving carer quality of life, reducing reactive responses In general, multidimensional, flexible carer interventions
to problem behaviours, improving burden levels (Teri et al., that involve follow-up and an ongoing relationship between
2005) and depression, agitation and sleep disturbance (Teri helper and carer are more likely to delay nursing home
et al., 1997, 2000; McCurry et al., 2005). The programme placement (Brodaty et al., 2003; Pinquart and Sörensen,
was successfully rolled out by chapters of the Alzheimer’s 2006b). A 10-day structured carer intervention programme
Association in the United States, showing a significant reduc- delayed institutionalization of care recipients over 7 years
tion in carers’ unmet needs and care-efficacy strain in the with concurrent improvement in the psychological health of
67% of completers (n = 219) (Menne et al., 2014). carers, without increasing use of health services or demen-
The Resources for Enhancing Alzheimer’s Carer Health tia drugs, and with significant cost savings over the first
(REACH) multisite, multicomponent project using a variety 3 years (Brodaty and Gresham, 1989; Brodaty and Peters,
of interventions is a 6-year study conducted over six loca- 1991; Brodaty et al., 1997).
tions in the United States (Belle et al., 2006). The follow-up Developing countries rely primarily on families to pro-
study, REACH II, was conducted over five sites with 692 vide support to people with dementia as health services
Latino, Caucasian and African American carers divided into are often ill-equipped to meet their needs (Dias et al.,
intervention and control groups (Stevens et al., 2009). The 2008). Family carers in LMIC face additional challenges as
intervention group received a comprehensive programme awareness and understanding about dementia are lacking;
including provision of information, didactic instruction, symptoms may be perceived to be part of normal ageing, or
role playing, problem solving, skills training, stress manage- denied because of the stigma attached to the illness (Patel
ment and telephone support (Belle et al., 2006). Compared and Prince, 2001; Senanarong et al., 2004; Dias et al., 2008).
to controls, significant improvements in depression, bur- Effective interventions addressing issues and needs relevant
den, care recipient problem behaviours, self-care and social to these carers are required and have been trialled as part of
support were found for Caucasian and Hispanic carers, but the 10/66 Dementia Research Group network.
not for African Americans (Belle et al., 2006). Since then, An RCT conducted in Goa, India, evaluated a home-
REACH II has been implemented by community agencies based intervention consisting of basic education about
(Lykens et al., 2014; Cheung et al., 2015; Altpeter et al., 2015) dementia and common behaviour problems, strategies for
showing significant reductions in depression and carer bur- managing problem behaviours, support to carers in ADLs,
den in completers of the intervention (Lykens et al., 2014; referral to psychiatrists or other medical professionals for
Cheung et al., 2015). However, high rates of non-completion assistance with BPSD, networking to assist the carers to
(up to 64%) due to institutionalization or death of the per- form support groups and advice on government provisions
sons with dementia or other reasons (Lykens et al., 2014) for the elderly (Dias et al., 2008). The intervention led to sig-
abate the enthusiasm for the programme somewhat. nificant improvements in carer mental health and perceived
The New York University Caregiver Intervention (NYUCI) burden for the 41 carers in the treatment group (compared to
is a carer programme involving individual and family coun- 40 controls). There were also reductions in the behavioural
selling sessions focusing on carer skills training, behaviour disturbances and improvements in the functional abilities
management techniques and carers’ communication strate- of the PWD, but these were not statistically significant (Dias
gies with the PWD; weekly support group participation; and et al., 2008). The programme used local health and human
availability of ad hoc telephone counselling. The intervention resources, making it affordable and easily accessible.
has been proven to be successful in improving carer well- A 6-month group counselling intervention was tri-
being by reducing depressive symptoms; improving reactions alled with 50 carers (half each assigned to the treatment
to memory and behaviour problems; and increasing the size and control groups) in Thailand (Senanarong et al., 2004).
and satisfaction with support networks (e.g. Roth et al., 2005; Counselling, support, education and strategies for manag-
Mittelman et al., 2006). The NYUCI also significantly delayed ing problem behaviours formed part of the intervention. At
nursing home placement (Mittelman et al., 2006; Gaugler 6 months, the neuropsychiatric symptoms of the PWD had
et al., 2013) and reduced the level of carer burden following the significantly declined. Carer distress associated with these
institutionalization (Gaugler et al., 2010). Community imple- symptoms declined over time compared with the control
mentations (Mittelman and Bartels, 2014) and multinational group, but not significantly.
adaptations of the NYUCI also yielded promising results An RCT in Moscow, Russia, involving a 5-week course
(Burns et al., 2010). A randomized controlled trial (RCT) of covering basic education about dementia and specific train-
five NYUCI sessions conducted in Manchester, New York and ing on managing problem behaviours, resulted in statisti-
Sydney significantly reduced carer depression (Mittelman cally significant reductions in carer burden and increases in
et al., 2008) and reduced time to nursing home placement quality of life that were close to significant (Gavrilova et al.,
but only at the Australian site (Brodaty et al., 2009). Possible 2008). The effect sizes for these were in the moderate range
152 Dementia

(0.64 and 0.52, respectively), and larger than those typically et al., 2011). As with late-onset dementia, female carers report
seen in interventions undertaken in high-income coun- higher levels of psychological morbidity than their male
tries (Brodaty et al., 2003; Gavrilova et al., 2008). Thus, an counterparts (van Vliet et al., 2010; Svanberg et al., 2011).
intervention focusing on education and training might be Systematic research focusing on the impact of dementia
particularly beneficial in a setting where community aware- and the needs of homosexual men and women and their car-
ness of dementia and appropriate formal care services are ers remains largely inadequate (Price, 2008). Homosexual
lacking (Gavrilova et al., 2008). Carer psychological mor- partners of PWD may feel that the existing interventions
bidity and the severity of, and the distress caused by BPSD and support services do not meet their needs (Birch, 2009)
did not improve significantly as a result of the intervention and that the issues they face (for instance next-of-kin rights)
(Gavrilova et al., 2008). are not well understood or addressed especially in residential
For all three studies small sample sizes limited the statis- and acute care settings. Homosexual carers also experience
tical power and possibly explained the lack of significance prejudice and insensitivity in their interactions with health
in some of the results. Additionally, the follow-up periods services; lack social and emotional support due to efforts to
may have been too short to demonstrate an effect, or to show maintain privacy in their nontraditional relationship; were
whether the intervention had a long-term impact on carer unable to use employee benefits to assist their partner with
and care-recipient well-being. dementia; faced opposition from employers when attempting
to take compassionate leave; and faced legal difficulties with
estate planning (Moore, 2002). Country-specific legislations
such as the Mental Capacity Act 2005 for England and Wales
14.6 SPECIAL CATEGORIES OF CARERS (Department for Constitutional Affairs, 2007) promise
important legal changes for homosexual patients and their
There are certain groups of carers who may experience carers concerning advance care planning and power of attor-
additional challenges beyond those directly related to ney (Price, 2008; Birch, 2009) (see below for more details).
caregiving. These include (but are not limited to) carers of Increasingly, people with Down syndrome and other
younger people with dementia; carers of people who iden- intellectual disabilities (ID) are living to an age where they
tify as gay, lesbian, bisexual, transsexual or transgender; and become susceptible to developing dementia (Evans et al.,
carers of people with intellectual disabilities (World Health 2013). Persons with Down syndrome are likely to develop
Organization and Alzheimer’s Disease International, 2012). Alzheimer pathology by their fourth decade, and clinical
People with YOD and their carers face additional prob- symptoms by their fifth or sixth decades (Margallo-Lana
lems as they are more likely to be working and have depen- et al., 2007). Carers of people with ID and dementia, mostly
dent children (World Health Organization and Alzheimer’s parents who have been the primary carer for decades, face
Disease International, 2012). Sometimes, these carers are unique challenges (Haley and Perkins, 2004). Aside from
the affected person’s parents; their challenges are poignant. the prolonged caregiving experience, parental carers are
Carers are often unprepared for the task and have fewer now facing age-related diseases themselves and anxieties
appropriate services available to them (Chaston et al., 2004; arise about who will care for their dependents if these par-
Arai et al., 2007; van Vliet et al., 2010). Formal services such ents become disabled or die.
as respite are usually designed for older people and are con- When a care recipient is in a second (or later) marriage,
sidered inappropriate for people with YOD (Withall 2013; particularly when there are children from a previous mar-
Withall et al., 2014). Informal social support may be fewer as riage, disputes are more likely to arise about legal and
family and friends are likely to be working and have depen- guardianship issues. When people marry close to the time
dents themselves, and are unable to empathize with the that one begins to dement, further issues can arise regard-
carer’s experience. Two systematic literature reviews have ing capacity to marry, the partner’s motivation (Peisah
summarized the evidence for negative mental health effects and Bridger, 2008) and possible issues to do with less well-­
of YOD carers (van Vliet et al., 2010; Svanberg et al., 2011). developed feelings of reciprocity and obligation.
There is strong evidence that YOD carers experience high lev-
els of burden, depression, distress and psychiatric symptoms
(van Vliet et al., 2010; Svanberg et al., 2011), with 50% or more
meeting cut-off scores for psychiatric caseness on global dis- 14.7 LEGAL ASPECTS
tress measures (Svanberg et al., 2011). When systematically
compared to carers of people with late-onset dementia, YOD Legal aspects concerning advance care planning (ACP), liv-
carers experience potentially higher levels of burden but the ing wills, enduring guardianship or power of attorney (POA)
evidence is less conclusive (van Vliet et al., 2010). The impact are particularly relevant to family carers (for more details see
of FTD on carers appears greater than for carers of people Chapters 30 and 32 in this book). Living wills or advance
with other types of dementia; with FTD carers reporting care directives specify personal preferences for future medi-
higher levels of burden, depression and distress associated cal care. Enduring guardians are appointed to make lifestyle,
with behavioural symptoms despite similar levels of behav- health or personal welfare decisions on a person’s behalf once
ioural symptoms (Kaiser and Panegyres, 2007; Svanberg the person is deemed no longer competent to do so. A POA
Family carers of people with dementia 153

authorizes a person to make financial or property decisions particularly effective in promoting ACP (Jezewski et al., 2007;
on behalf of an individual if this person no longer has capac- Bravo et al., 2008). General practitioners are often viewed to
ity to do so (Mullick et al., 2013). In some jurisdictions prior be best suited to initiate first discussions about ACP and POA
POA becomes invalid when the appointer becomes incom- (Samsi and Manthorpe, 2011; Mitchell et al., 2014).
petent. This can be circumvented, if the PWD declares that Even if advance care plans are in place, the different con-
these proxy powers should endure even if he or she loses cepts are often poorly understood by carers (Ruggieri and
competency (i.e. enduring, durable or lasting POA). Piccoli, 2003; Astell et al., 2013). For instance, family mem-
There is considerable variation in laws between countries bers often assume a POA takes immediate effect, while in
and even between jurisdictions within countries (Peisah some countries (e.g. United Kingdom and New Zealand)
et al., 2009; Stoppe, 2009). In any case, the person’s right to it only becomes effective once the person loses capacity.
self-determination and to have his or her wishes respected (Astell et al., 2013; Mullick et al., 2013). In Australia, the
should be protected. In England, the Mental Capacity Act person taking out an enduring POA can specify under what
provides a legal framework for people who lack capacity or circumstances it becomes active (Mitchell et al., 2014).
who want to make provisions for a time when they no longer Carers also have an important legal role in assisting
have capacity (Department for Constitutional Affairs, 2007). care recipients to make decisions about treatment and par-
Among healthy older community-dwellers, the hypo- ticipation in research. Informed consent is required before
thetical onset of a dementing illness was stated as a trig- prescribing medication, which requires the patient to
ger for putting in place ACP arrangements (Samsi and understand what the medication is for, the likely benefits,
Manthorpe, 2011; Mitchell et al., 2014). They certainly possible adverse events and any alternatives (Brodaty and
should be discussed with anyone who has dementia and Green, 2002). If the person with dementia lacks capacity the
their family (Bisson et al., 2009). The sooner after a diag- carer may be called on to give proxy consent to initiate phar-
nosis ACP is considered the better, so that the person can macotherapy, or to participate in a drug trial or other rel-
participate as fully as possible in the process (Alzheimer’s evant research. Jurisdictions vary regarding the legislation
Association, 2007; Raivio et al., 2008). The potential ben- and requirements for proxy consent. In some jurisdictions
efits of advance care plans for patients and carers have in the United States, uncertainty has led to institutions dis-
been demonstrated in an Australian RCT involving 309 allowing proxy consent for research (Kim et al., 2009). This
elderly legally competent hospital inpatients, of whom 40% is despite broad public support for a policy of proxy (or sur-
received an ACP intervention. Patients with advance care rogate) consent for AD research. A 2006 survey of 1515 older
directives in place were more likely to have their end-of-life Americans (aged 51 and older) showed between 68% and
wishes known and acted upon (86% compared to 30% in 83% support depending on the scenario (Kim et al., 2009).
the control group) (Detering et al., 2010). Having advanced
care directives in place was also associated with signifi-
cantly lower psychological morbidity in family members
and greater satisfaction with the end-of-life care after the
14.8 CONCLUSIONS
loved one’s death (Detering et al., 2010).
Despite these benefits, the rates at which advance care Dementia seldom afflicts just one person. Carers (or care-
directives, legal guardianships and financial POA are in givers) are an integral part of the dementia management
place vary widely from 0 to 71% depending on the coun- equation from symptom onset through diagnosis and
try, the setting and demographic and clinical aspects of the management to death. Carers, the ‘second patient’, deserve
person with dementia and the carer (Ruggieri and Piccoli, attention in their own right. Those vulnerable to stress can
2003; Lingler et al., 2008; Raivio et al., 2008; Garand et al., be identified and interventions can reduce carers’ distress
2011; Astell et al., 2013). The main factor associated with and improve their quality of life. This, in turn, can lead to a
initiating an advance care directive or durable POA was better quality of life for persons with dementia and increase
younger patient age (i.e. <65 years), moderated by disease their length of time living in the community.
severity, male sex, having a spousal carer, a family history of
AD and the absence of major depressive disorder at baseline REFERENCES
(Garand et al., 2011). Other studies have identified older age
(Mitchell et al., 2014) or disease severity, behavioural prob- Abdollahpour, I., Noroozian, M., Nedjat, S. and
lems and carer strain but not demographic factors as pre- Majdzadeh, R. (2012). Caregiver burden and its deter-
dictors of initiating advance care plans (Raivio et al., 2008). minants among the family members of patients with
Reasons for not having advance care plans in place are dementia in Iran. International Journal of Preventive
lack of awareness and knowledge. Raising the awareness for Medicine, 3 (8): 544–551.
the importance of ACP and POA in particular has a posi- Aboderin, I. (2004). Decline in material family support
tive impact on a person’s willingness to put a POA in place for older people in urban Ghana, Africa: understand-
(Mitchell et al., 2014). Meta-analytic evidence suggests that ing processes and causes of change. Journals of
one-on-one interactive interventions which encourage ques- Gerontology Series B – Psychological Sciences and
tions and offer practical help with completing forms are Social Sciences, 59 (3): S128–S137.
154 Dementia

Adelman, R.D., Tmanova, L.L., Delgado, D. et al. (2014). Bachner, Y.G. and O’Rourke, N. (2007). Reliability gener-
Caregiver burden: A clinical review. JAMA – Journal of alization of responses by care providers to the Zarit
the American Medical Association, 311 (10): 1052–1059. Burden Interview. Aging & Mental Health, 11 (6):
Adkins, V.K. (1999). Treatment of depressive disorders 678–685.
of spousal caregivers of persons with Alzheimer’s Baker, K.L. and Robertson, N. (2008). Coping with
disease: A review. American Journal of Alzheimer’s caring for someone with dementia: Reviewing the
Disease, 14: 289–293. literature about men. Aging & Mental Health, 12 (4):
Altpeter, M., Gwyther, L.P., Kennedy, S.R. et al. (2015). 413–422.
From evidence to practice: Using the RE-AIM frame- Belle, S., Burgio, L., Burns, R. et al. (2006). Enhancing the
work to adapt the REACHII caregiver intervention to quality of life of dementia caregivers from different
the community. Dementia, 14 (1): 104–113. ethnic or racial groups. Annals of Internal Medicine,
Alzheimer’s Association (2007). Alzheimer’s Disease Facts 145: 727–738.
and Figures. Chicago, IL: Alzhemier’s Association. Birch H. (2009). Dementia, Lesbians and Gay Men.
Alzheimer’s Association (2008). More Men Take the Lead Canberra, Australia: Alzheimer’s Australia.
Role in Caring for Elderly Parents. Retrieved December Bisson, J.I., Hampton, V., Rosser, A. and Holm, S. (2009).
17, 2008; Available at http://www.alz.org/news_and_ Developing a care pathway for advance decisions and
events_in_the_news.asp. powers of attorney: Qualitative study. British Journal
Alzheimer’s Association (2014). 2014 Alzheimer’s disease of Psychiatry, 194 (1): 55–61.
facts and figures. Alzheimer’s & Dementia, 10 (2): Black, W. and Almeida, O.P. (2004). A systematic review
e47–e92. of the association between the Behavioral and
Alzheimer’s Association and National Alliance for Psychological Symptoms of Dementia and burden of
Caregiving (2004). Families Care: Alzheimer’s care. International Psychogeriatrics, 16 (3): 295–315.
Caregiving in the United States. Chicago, IL: Boots, L.M.M., De Vugt, M.E., Van Knippenberg, R.J.M.
Alzheimer’s Association and National Alliance for et al. (2014). A systematic review of Internet-based
Caregiving. supportive interventions for caregivers of patients with
Alzheimer’s Disease International (2009). World Alzheimer dementia. International Journal of Geriatric Psychiatry,
Report 2009. Prince, M. and Jackson, J. (eds.). London, 29 (4): 331–344.
United Kingdom: Alzheimer’s Disease International. Bouldin, E. and Andresen, E. (2010). Caregiving across the
Alzheimer’s Disease International (2013). Policy Brief United States: Caregivers of Persons with Alzheimer’s
for Heads of Government – The Global Impact of Disease or Dementia in 8 States and the District of
Dementia 2013–2050. London, United Kingdom: Columbia. Data from the 2010 Behavioral Risk Factor
Alzheimer’s Disease International. Surveillance System. Available at http://www.alz.org/
Alzheimer’s Disease International (2014). World documents_custom/public-health/brfss_caregiver_
Alzheimer Report 2014 – Dementia and Risk report_2010.pdf. Accessed 5 June 2016.
Reduction: An Analysis of Protective and Modifiable Bravo, G., Dubois, M.-F. and Wagneur, B. (2008).
Factors. Prince, M., Albanese, E., Guerchet, M. and Assessing the effectiveness of interventions to pro-
Prina, M. London, United Kingdom, Alzheimer’s mote advance directives among older adults: A sys-
Disease International. tematic review and multi-level analysis. Social Science
Aneshensel, C., Pearlin, L.I., Mullan, J.T. et al. (1995). and Medicine, 67 (7): 1122–1132.
Profiles in Caregiving: The Unexpected Career. San Brodaty, H. and Arasaratnam, C. (2012). Meta-analysis
Diego, CA: Academic Press. of nonpharmacological interventions for neuropsy-
Arai, A., Matsumoto, T., Ikeda, M. and Arai, Y. (2007). Do chiatric symptoms of dementia. American Journal of
family caregivers perceive more difficulty when they Psychiatry, 169 (9): 946–953.
look after patients with early onset dementia com- Brodaty, H. and Donkin, M. (2009). Family caregiv-
pared to those with late onset dementia. International ers of people with dementia. Dialogues in Clinical
Journal of Geriatric Psychiatry, 22: 1255–1261. Neuroscience, 11 (2): 217–228.
Astell, H., Lee, J.H. and Sankaran, S. (2013). Review of Brodaty, H. and Green, A. (2002). Defining the role of the
capacity assessments and recommendations for caregiver in Alzheimer’s disease treatment. Drugs and
examining capacity. New Zealand Medical Journal, 126 Aging, 19 (12): 891–898.
(1383): 38–48. Brodaty, H., Green, A. and Koschera, A. (2003). Meta-
Australian Institute of Health and Welfare (2007). analysis of psychosocial interventions for caregivers
Dementia in Australia: National Data Analysis and of people with dementia. Journal of the American
Development. Canberra, Australia: Australian Institute Geriatric Society, 51: 657–664.
of Health and Welfare. Brodaty, H. and Gresham, M. (1989). Effects of a
Australian Institute of Health and Welfare (2012). training programme to reduce stress in carers of
Dementia in Australia. Canberra, Australia: Australian patients with dementia. British Medical Journal, 299:
Institute of Health and Welfare. 1375–1379.
Family carers of people with dementia 155

Brodaty, H., Gresham, M. and Luscombe, G. (1997). The Cooper, C., Balamurali, T.B.S. and Livingston, G. (2007).
Prince Henry Hospital dementia caregivers’ train- A systematic review of the prevalence and covari-
ing programme. International Journal of Geriatric ates of anxiety in caregivers of people with dementia.
Psychiatry, 12: 183–192. International Psychogeriatrics, 19 (2): 175–195.
Brodaty, H. and Hadzi-Pavlovic, D. (1990). Psychosocial Cooper, C., Katona, C., Orrell, M. and Livingston, G.
effects on carers of living with persons with dementia. (2006). Coping strategies and anxiety in caregivers of
Australian and New Zealand Journal of Psychiatry, 24: people with Alzheimer’s disease: The LASER-AD study.
351–361. Journal of Affective Disorders, 90: 15–20.
Brodaty, H., McGilchrist, C., Harris, L. and Peters, K. Cooper, C., Owens, C., Katona, C. and Livingston, G. (2008).
(1993). Time until institutionalization and death in Attachment style and anxiety in carers of people with
patients with dementia: Role of caregiver training and Alzheimer’s disease: Results from the LASER-AD study.
risk factors. Archives of Neurology, 50: 643–650. International Psychogeriatrics, 20 (3): 494–507.
Brodaty, H., Mittelman, M., Gibson, L. et al. (2009). The Corbett, A., Stevens, J., Aarsland, D. et al. (2012).
effects of counseling spouse caregivers of people with Systematic review of services providing information
Alzheimer’s disease taking donepezil and of country of and/or advice to people with dementia and/or their
residence on rates of admission to nursing homes and caregivers. International Journal of Geriatric Psychiatry,
mortality. American Journal of Geriatric Psychiatry, 17: 27 (6): 628–636.
734–743. Covinsky, K.E., Newcomer, R., Fox, P. et al. (2003). Patient
Brodaty, H. and Peters, K. (1991). Cost effectiveness of and caregiver characteristics associated with depres-
a training program for dementia carers. International sion in caregivers of patients with dementia. Journal of
Psychogeriatrics, 3 (1): 11–22. General Internal Medicine, 18 (12): 1006–1014.
Burns, A., Mittelman, M., Cole, C. et al. (2010). Transcultural Cuijpers, P. (2005). Depressive disorders in caregivers
influences in dementia care: Observations from a psy- of dementia patients: A systematic review. Aging &
chosocial intervention study. Dementia and Geriatric Mental Health, 9 (4): 325–330.
Cognitive Disorders, 30 (5): 417–423. del-Pino-Casado, R., Frías-Osuna, A., Palomino-Moral,
Camden, A., Livingston, G. and Cooper, C. (2011). Reasons P.A. and Pancorbo-Hidalgo, P.L. (2011). Coping and
why family members become carers and the outcome subjective burden in caregivers of older relatives: A
for the person with dementia: Results from the CARD quantitative systematic review. Journal of Advanced
study. International Psychogeriatrics, 23 (9): 1442–1450. Nursing, 67 (11): 2311–2322.
Campbell, P., Wright, J., Oyebode, J. et al. (2008). Department for Constitutional Affairs (2007). Mental
Determinants of burden in those who care for some- Capacity Act 2005 – Code of Practice. London, United
one with dementia. International Journal of Geriatric Kingdom: The Stationery Office (TSO).
Psychiatry, 23: 1078–1085. Detering, K.M., Hancock, A.D., Reade, M.C. and Silvester,
Chaston, D., Pollard, N. and Jubb, D. (2004). Young onset W. (2010). The impact of advance care planning on end
dementia: A case for real empowerment. Journal of of life care in elderly patients: Randomised controlled
Dementia Care, 12: 24–26. trial. BMJ, 340: c1345.
Cheung, K.S.L., Lau, B.H.P., Wong, P.W.C. et al. (2015). Dias, A., Dewey, M.E., D’Souza, J. et al. (2008). The
Multicomponent intervention on enhancing dementia effectiveness of a home care program for supporting
caregiver well-being and reducing behavioral prob- caregivers of persons with dementia in developing
lems among Hong Kong Chinese: A translational study countries: A randomised controlled trial from Goa,
based on REACH II. International Journal of Geriatric India. PLos One, 3 (6): 1–7.
Psychiatry, 30 (5): 460–469. Dilworth-Anderson, P., Brummett, B.H., Goodwin, P. et al.
Chien, L.-Y., Chu, H., Guo, J.-L. et al. (2011). Caregiver (2005). Effect of race on cultural justifications for care-
support groups in patients with dementia: A meta- giving. Journal of Gerontology, 60B (5): S257–S262.
analysis. International Journal of Geriatric Psychiatry, Dilworth-Anderson, P., Goodwin, P. and Williams, S.
26 (10): 1089–1098. (2004). Can culture help explain the physical health
Clare, L., Wilson, B.A., Carter, G. et al. (2002). Depression effects of caregiving over time among African
and anxiety in memory clinic attenders and their car- American caregivers? Journal of Gerontology: Social
ers: Implications for evaluating the effectiveness of Sciences, 59B: S138–S145.
cognitive rehabilitation interventions. International Draper, B., Poulos, C.J., Cole, A.M.D. et al. (1992). A compari-
Journal of Geriatric Psychiatry, 17: 962–967. son of caregivers for elderly stroke and dementia victims.
Cohen, S. (2004). Social relationships and health. Journal of the American Geriatric Society, 40: 896–901.
American Psychologist, 59: 676–684. Drentea, P., Clay, O.J., Roth, D.L. and Mittelman, M.S.
Cohen, C.A., Colantonio, A. and Vernich, L. (2002). (2006). Predictors of improvement in social support:
Positive aspects of caregiving: Rounding out the care- Five-year effects of a structured intervention for
giving experience. International Journal of Geriatric caregivers of spouses with Alzheimer’s disease. Social
Psychiatry, 12: 331–336. Science and Medicine, 63: 957–967.
156 Dementia

Edwards, J. and Cooper, C. (1988). Research in stress, Georges, J., Jansen, S., Jackson, J. et al. (2008).
coping and health: Theoretical and methodological Alzheimer’s disease in real life – The dementia carer’s
issues. Psychological Medicine, 18: 15–20. survey. International Journal of Geriatric Psychiatry, 23
Edwards, A.B., Zarit, S., Stephens, M. and Townsend, (5): 546–551.
A. (2002). Employed family caregivers of cognitively Goode, K.T., Haley, W.E., Roth, D.L. and Ford, G.R. (1998).
impaired elderly: An examination of role strain and Predicting longitudinal changes in caregiver physical
depressive symptoms. Aging & Mental Health, 6 (1): and mental health: A stress process model. Health
55–61. Psychology, 17 (2): 190–198.
Eisdorfer, C. (1991). Caregiving: An emerging risk factor Haley, W.E., Levine, E.G., Brown, S.L. et al. (1987). Stress,
for emotional and physical pathology. Bulletin of the appraisal, coping and social support as predictors of
Menninger Clinic, 55: 238–247. adaptational outcome among dementia caregivers.
Etters, L., Goodall, D. and Harrison, B.E. (2008). Caregiver Psychological Ageing, 2: 223–230.
burden among dementia patient caregivers: A review Haley, W.E. and Perkins, E.A. (2004). Current status and
of the literature. Journal of the American Academy of future directions in family caregiving and aging people
Nurse Practitioners, 20 (8): 423–428. with intellectual disabilities. Journal of Policy and
Evans, E., Bhardwaj, A., Brodaty, H.l et al. (2013). Practice in Intellectual Disabilities, 1 (1): 24–30.
Dementia in people with intellectual disability: Insights Haley, W.E., Roth, D.L., Coleton, M.I. et al. (1996).
and challenges in epidemiological research with an Appraisal, coping and social support as mediators of
at-risk population. International Review of Psychiatry, well-being in black and white Alzheimer’s family care-
25 (6): 755–763. givers. Journal of Consulting and Clinical Psychology,
Ferri, C.P., Prince, M., Brodaty, H. et al. (2005). Global 64: 121–129.
prevalence of dementia: A Delphi consensus study. Hoffman, G.J., Lee, J. and Mendez-Luck, C.A. (2012).
Lancet, 366: 2112–2117. Health behaviors among Baby Boomer informal care-
Fisher, G.G., Franks, M.M., Plassman, B.L. et al. (2011). givers. Gerontologist, 52 (2): 219–230.
Caring for individuals with dementia and cognitive Hurd, M.D., Martorell, P., Delavande, A. et al. (2013).
impairment, not dementia: Findings from the aging, Monetary costs of dementia in the United States. New
demographics, and memory study. Journal of the England Journal of Medicine, 368 (14): 1326–1334.
American Geriatrics Society, 59 (3): 488–494. Hurley, R.V.C., Patterson, T.G. and Cooley, S.J. (2014).
Garand, L., Dew, M.A., Lingler, J.H. and DeKosky, S.T. (2011). Meditation-based interventions for family caregivers
Incidence and predictors of advance care planning of people with dementia: A review of the empirical
among persons with cognitive impairment. American literature. Aging & Mental Health 18(3): 281–288.
Journal of Geriatric Psychiatry, 19 (8): 712–720. Jelic, V., Johansson, S., Almkvist, O. et al. (2000).
Gaugler, J.E., Mittelman, M.S., Hepburn, K. and Newcomer, Quantitative electroencephalography in mild cognitive
R. (2010). Clinically significant changes in burden and impairment: Longitudinal changes and possible pre-
depression among dementia caregivers following nurs- diction of Alzheimer’s disease. Neurobiology of Aging,
ing home admission. BMC Medicine, 8: 85. 21 (4): 533–540.
Gaugler, J.E., Pot, A.M. and Zarit, S.H. (2007). Long-term Jezewski, M.A., Meeker, M.A., Sessanna, L. and Finnell,
adaptation to institutionalization. The Gerontologist, D.S. (2007). The effectiveness of interventions to
47: 730–740. increase advance directive completion rates. Journal
Gaugler, J.E., Reese, M. and Mittelman, M.S. (2013). Effects of Aging and Health, 19 (3): 519–536.
of the NYU caregiver intervention-adult child on residen- Kaiser, S. and Panegyres, P.K. (2007). The psychoso-
tial care placement. Gerontologist, 53 (6): 985–997. cial impact of young onset dementia on spouses.
Gaugler, J.E., Roth, D.L., Haley, W.E. and Mittelman, M.S. American Journal of Alzheimer’s Disease and Other
(2008). Can counseling and support reduce burden Dementias, 21 (6): 398–402.
and depressive symptoms in caregivers of people with Kasper, J.D., Steinbach, U. and Andrews, J. (1994).
Alzheimer’s disease during the transition to institu- Caregiver role appraisal and caregiver tasks as factors
tionalization? Results from the New York University in ending caregiving. Journal of Aging and Health, 6
caregiver intervention study. Journal of the American (3): 397–414.
Geriatrics Society, 56 (3): 421–428. Kim, S.Y., Kim, H.M., Langa, K.M. et al. (2009). Surrogate
Gaugler, J.E., Yu, F., Krichbaum, K. and Wyman, J.F. consent for dementia research: A national survey of
(2009). Predictors of nursing home admission for per- older Americans. Neurology, 72 (2): 149–155.
sons with dementia. Medical Care, 47 (2): 191–198. Kinney, J.M. and Stephens, M.P. (1989). Hassles and uplifts
Gavrilova, S.I., Ferri, C.P., Mikhaylova, N. et al. (2008). of giving care to a family member with dementia.
Helping carers to care: The 10/66 Dementia Research Psychology and Aging, 4 (4): 402–408.
Group’s randomized control trial of a caregiver inter- Kneebone, I.I. and Martin, P.R. (2003). Coping and care-
vention in Russia. International Journal of Geriatric givers of people with dementia. British Journal of
Psychiatry, 24 (4): 347–354. Health Psychology, 8 (1): 1–17.
Family carers of people with dementia 157

Knight, B., Fox, L. and Chou, C.-P. (2000). Factor struc- Margallo-Lana, M., Moore, P., Kay, D. et al. (2007). Fifteen-
ture of the burden interview. Journal of Clinical year follow-up of 92 hospitalized adults with Down’s
Geropsychology, 6 (4): 249–258. syndrome: Incidence of cognitive decline, its relation-
Kramer, B.J. (1997). Gain in the caregiving experience: ship to age and neuropathology. Journal of Intellectual
Where are we? What next? The Gerontologist, 2: Disability Research, 51 (6): 463–477.
218–232. McCurry, S., Gibbons, L., Logsdon, R. et al. (2005).
Lawton, M.P., Moss, M., Hoffman, C. and Perkinson, Nighttime insomnia treatment and education for
M. (2000). Two transitions in daughters’ caregiving Alzheimer’s disease: A randomized controlled trial.
careers. The Gerontologist, 40(4): 437–448. Journal of the American Geriatric Society, 53: 793–802.
Lazarus, R.S. and Folkman, S. (1984). Stress, Appraisal and Menne, H.L., Bass, D.M., Johnson, J.D. et al. (2014).
Coping. New York, NY: Springer. Statewide implementation of “Reducing Disability
Leong, J., Madjar, I. and Fiveash, B. (2001). Needs of fam- in Alzheimer’s Disease”: Impact on family caregiver
ily carers of elderly people with dementia living in the outcomes. Journal of Gerontological Social Work, 57
community. Australasian Journal on Ageing, 20 (3): (6–7): 626–639.
133–138. Mitchell, L.K., Pachana, N.A., Wilson, J. et al. (2014).
Lévesque, L., Cossette, S. and Lachance, L. (1998). Promoting the use of enduring powers of attorney in
Predictors of the psychological well-being of primary older adults: A literature review. Australasian Journal
caregivers living with a demented relative: A 1-year on Ageing, 33 (1): 2–7.
follow-up study. Journal of Applied Gerontology 17 (2): Mittelman, M.S. and Bartels, S.J. (2014). Translating
240–258. research into practice: Case study of a community-
Li, R., Cooper, C., Bradley, J., Shulman, A. and Livingston, based dementia caregiver intervention. Health Affairs,
G. (2012). Coping strategies and psychological mor- 33 (4): 587–595.
bidity in family carers of people with dementia: A sys- Mittelman, M.S., Brodaty, H., Wallen, A.S. and Burns, A.
tematic review and meta-analysis. Journal of Affective (2008). A three-country randomized controlled trial of a
Disorders, 139 (1): 1–11. psychosocial intervention for caregivers combined with
Lingler, J.H., Hirschman, K.B., Garand, L. et al. (2008). pharmacological treatment for patients with Alzheimer
Frequency and correlates of advance planning disease: Effects on caregiver depression. The American
among cognitively impaired older adults. The Journal of Geriatric Psychiatry, 16 (11): 893–904.
American Journal of Geriatric Psychiatry, 16 (8): Mittelman, M.S., Haley, W.E., Clay, O.J. and Roth, D.L.
643–649. (2006). Improving caregiver well-being delays nursing
Livingston, G., Mahoney, R., Regan, C. and Katona, home placement of patients with Alzheimer disease.
C. (2005). The caregivers for Alzheimer’s disease Neurology, 67 (1): 1592–1599.
Problems Scale (CAPS): Development of a new scale Moore, W. (2002). Lesbian and gay elders: Connecting
within the LASER-AD study. Age and Ageing, 34: care providers through a telephone support group.
287–290. Journal of Gay and Lesbian Social Services 14 (3):
LoGiudice, D., Waltrowicz, W., Brown, K. et al. (1999). Do 23–41.
memory clinics improve the quality of life of carers? A Mullick, A., Martin, J. and Sallnow, L. (2013). An intro-
randomized pilot trial. International Journal of Geriatric duction to advance care planning in practice. BMJ
Psychiatry, 14: 626–632. (Online), 347 (7930): f6064.
Lopez, J. and Crespo, M. (2008). Analysis of the efficacy of Neubauer, S., Holle, R., Menn, P., Grossfeld-Schmitz, M.
a psychotherapeutic program to improve the emo- and Graesel, E. (2008). Measurement of informal care
tional status of caregivers of elderly dependent rela- time in a study of patients with dementia. International
tives. Aging & Mental Health, 12 (4): 451–461. Psychogeriatrics, 20 (6): 1160–1176.
Lowery, K., Mynt, P., Aisbett, J. et al. (2000). Depression Nomura, H., Inoue, S., Kamimura, N. et al. (2005).
in the carers of dementia sufferers: A comparison A cross-cultural study on expressed emotion in
of the carers of patients suffering from dementia carers of people with dementia and schizophrenia:
with Lewy Bodies and the carers of patients with Japan and England. Social Psychiatry and Psychiatric
Alzheimer’s disease. Journal of Affective Disorders, Epidemiology, 40: 564–570.
59: 61–65. Norton, M.C., Clark, C., Fauth, E.B. et al. (2013). Caregiver
Lykens, K., Moayad, N., Biswas, S. et al. (2014). Impact of personality predicts rate of cognitive decline in a com-
a community based implementation of REACH II pro- munity sample of persons with Alzheimer’s disease.
gram for caregivers of Alzheimer’s patients. PLos One, The Cache County Dementia Progression Study.
9 (2): e89290. International Psychogeriatrics, 25 (10): 1629–1637.
Maayan, N., Soares-Weiser, K. and Lee, H. (2014). Respite Noyes, B.B., Hill, R.D., Hicken, B.L. et al. (2010). The
care for people with dementia and their carers. role of grief in dementia caregiving. American
The Cochrane Database of Systematic Reviews, 1: Journal of Alzheimer’s Disease and Other Dementias,
CD004396. 25 (1): 9–17.
158 Dementia

O’Rourke, N. and Tuokko, H.A. (2003). Psychometric Pinquart, M. and Sörensen, S. (2006b). Helping caregiv-
properties of an abridged version of the Zarit Burden ers of persons with dementia: Which interventions
Interview within a representative Canadian caregiver work and how large are their effects? International
sample. The Gerontologist, 43 (1): 121–127. Psychogeriatrics, 18 (4): 577–595.
Okojie, F.A. (2004). Ageing in sub-Saharan Africa: Toward Pinquart, M. and Sörensen, S. (2011). Spouses, adult chil-
a redefinition of needs, research, and policy directions. dren, and children-in-law as caregivers of older adults:
Journal of Cross Cultural Gerontology, 3: 3–19. A meta-analytic comparison. Psychology and Aging,
Orgeta, V. and Miranda-Castillo, C. (2014). Does physi- 26 (1): 1–14.
cal activity reduce burden in carers of people with Poulshock, S.W. and Deimling, G.T. (1984). Families caring
dementia? A literature review. International Journal of for elders in residence: Issues in the measurement of
Geriatric Psychiatry, 29 (8): 771–783. burden. Journal of Gerontology, 39 (2): 230–239.
Ornstein, K. and Gaugler, J.E. (2012). The problem with Powell, J., Chiu, T. and Eysenbach, G. (2008). A systematic
“problem behaviors”: A systematic review of the review of networked technologies supporting carers
association between individual patient behavioral of people with dementia. Journal of Telemedicine and
and psychological symptoms and caregiver depres- Telecare, 14: 154–156.
sion and burden within the dementia patient-care- Price, E. (2008). Pride or prejudice? Gay men, lesbians
giver dyad. International Psychogeriatrics, 24 (10): and dementia. British Journal of Social Work, 38 (7):
1536–1552. 1337–1352.
Parker, D., Mills, S. and Abbey, J. (2008). Effectiveness of Prince, M., Brodaty, H., Uwakwe, R. et al. (2012). Strain
interventions that assist caregivers to support people and its correlates among carers of people with
with dementia living in the community: A system- dementia in low-income and middle-income countries.
atic review. International Journal of Evidence-Based A 10/66 Dementia Research Group population-based
Healthcare, 6 (2): 137–172. survey. International Journal of Geriatric Psychiatry, 27
Patel, V. and Prince, M. (2001). Ageing and mental health (7): 670–682.
in a developing country: Who cares? Qualitative stud- Prince, M. and the 10/66 Dementia Research Group
ies from Goa, India. Psychological Medicine, 31 (1): (2004). Care arrangements for people with demen-
29–38. tia in developing countries. International Journal of
Pearlin, L.I., Mullan, J.T., Semple, S.J. and Skaff, M.M. Geriatric Psychiatry, 19 (2): 170–177.
(1990). Caregiving and the stress process: An overview Pyke, K.D. and Bengston, V.L. (1996). Caring more or
of concepts and their measures. The Gerontologist, less: individualistic and collectivist systems of family
30: 583–594. eldercare. Journal of Marriage and the Family, 58:
Pearlin, L.I., Nguyen, K.B., Schieman, S. and Milkie, M.A. 379–392.
(2007). The life-course origins of mastery among older Raivio, M.M., Maki-Petaja-Leinonen, A.P., Laakkonen, M.L.
people. Journal of Health and Social Behavior, 48 (2): et al. (2008). The use of legal guardians and finan-
164–179. cial powers of attorney among home-dwellers with
Peisah, C. and Bridger, M. (2008). Abuse by marriage: The Alzheimer’s disease living with their spousal caregiv-
exploitation of mentally ill older people. International ers. Law, Ethics and Medicine, 34: 882–886.
Journal of Geriatric Psychiatry, 23: 883–888. Rapp, S.R. and Chao, D. (2000). Appraisals of strain and
Peisah, C., Forlenza, O. and Chiu, E. (2009). Ethics, capac- of gain: Effects on psychological wellbeing of caregiv-
ity, and decision making in the practice of old age ers of dementia patients. Aging & Mental Health, 4:
psychiatry: an emerging dialogue. Current Opinion in 142–148.
Psychiatry, 22: 519–521. Reidijk, S.R., De Vugt, M.E., Duivenvoorden, H.J. et al.
Pinquart, M. and Sörensen, S. (2003a). Associations of (2006). Caregiver burden, health-related quality of life
stressors and uplifts of caregiving with caregiver bur- and coping in dementia caregivers: A comparison of
den and depressive mood: A meta-analysis. Journals Frontotemporal Dementia and Alzheimer’s Disease.
of Gerontology – Series B Psychological Sciences and Dementia and Geriatric Cognitive Disorders, 22:
Social Sciences, 58 (2): P112–P128. 405–412.
Pinquart, M. and Sörensen, S. (2003b). Differences Resource Implications Study Group (2000). Psychological
between caregivers and noncaregivers in psycho- morbidity among informal caregivers of older people:
logical health and physical health: A meta-analysis. A 2-year follow-up study. Psychological Medicine, 30
Psychology and Aging, 18: 250–267. (04): 943–955.
Pinquart, M. and Sörensen, S. (2006a). Gender differences Roepke, S.K., Mausbach, B.T., Aschbacher, K. et al.
in caregiver stressors, social resources, and health: (2008). Personal mastery is associated with reduced
An updated meta-analysis. Journals of Gerontology – sympathetic arousal in stressed alzheimer caregiv-
Series B Psychological Sciences and Social Sciences, 61 ers. American Journal of Geriatric Psychiatry 16 (4):
(1): P33–P45. 310–317.
Family carers of people with dementia 159

Rosdinom, R., Zarina, M., Zanariah, M. et al. (2013). Seeher, K., Low, L.-F., Reppermund, S. and Brodaty,
Behavioural and psychological symptoms of demen- H. (2013). Predictors and outcomes for caregiv-
tia, cognitive impairment and caregiver burden in ers of people with mild cognitive impairment – A
patients with dementia. Preventive Medicine, 57: ­systematic literature review. Alzheimer’s & Dementia,
S67–S69. 9 (3): 346–355.
Roth, D.L., Mittelman, M.S., Clay, O.J. et al. (2005). Selwood, A., Johnston, K., Katona, C. et al. (2007). A sys-
Changes in social support as mediators of the impact tematic review of the effect of psychological interven-
of a psychosocial intervention for spouse caregivers tions on family caregivers of people with dementia.
of persons with Alzheimer’s Disease. Psychology and Journal of Affective Disorders, 101: 75–89.
Aging, 20 (4): 634–644. Senanarong, V., Jamjumras, P., Harmphadungkit, K. et al.
Ruggieri, R.M. and Piccoli, F. (2003). Legal and assistance (2004). A counseling intervention for caregivers: Effect
aspects of Alzheimer’s disease: Analysis of 100 cases. on neuropsychiatric symptoms. International Journal of
Neurological Science, 24: 125–129. Geriatric Psychiatry, 19: 781–788.
Samsi, K. and Manthorpe, J. (2011). ‘I live for today’: Serrano-Aguilar, P.G., Lopez-Bastida, J. and Yanes-Lopez,
A qualitative study investigating older people’s V. (2006). Impact on health-related quality of life and
­attitudes to advance planning. Health and Social Care perceived burden of informal caregivers of individu-
in the Community, 19 (1): 52–59. als with Alzheimer’s Disease. Neuroepidemiology, 27:
Sanders, S. (2005). Is the glass hald empty or half full? 136–142.
Reflections on strain and gain in caregivers of individu- Shrout, P.E. and Bolger, N. (2002). Mediation in experi-
als with Alzheimer’s Disease. Social Work in Health mental and nonexperimental studies: New procedures
Care, 40 (3): 57–73. and recommendations. Psychological Methods, 7(4):
Schoenmakers, B., Buntinx, F. and De Lepeleire, J. (2009). 422–445.
Can pharmacological treatment of behavioural dis- Skaff, M.M., Pearlin, L.I. and Mullan, J.T. (1996). Transitions
turbances in elderly patients with dementia lower the in the caregiving career: Effects on sense of mastery.
burden of their family caregiver? Family Practice, 26 Psychology and Aging, 11 (2): 247–257.
(4): 279–286. Stephens, M.A.P., Townsend, A.L., Martire, L.M. and
Schoenmakers, B., Buntinx, F. and Delepeleire, J. (2010a). Druley, J.A. (2001). Balancing parent care with other
Factors determining the impact of care-giving roles: Interrole conflict of adult daughter caregiv-
on caregivers of elderly patients with dementia. ers. Journals of Gerontology – Series B Psychological
A ­systematic literature review. Maturitas, 66 (2): Sciences and Social Sciences, 56 (1): P24–P34.
191–200. Stevens, A.R., Lancer, K., Smith, E.R. et al. (2009).
Schoenmakers, B., Buntinx, F. and DeLepeleire, J. (2010b). Engaging communities in evidence-based interven-
Supporting the dementia family caregiver: The effect tions for dementia caregivers. Family and Community
of home care intervention on general well-being. Health, 32 (Suppl. 1): s83–s92.
Aging & Mental Health, 14 (1): 44–56. Stone, R., Cafferata, G.L. and Sangl, J. (1987).
Schreiner, A.S., Morimoto, T., Arai, Y. and Zarit, S. Caregivers of the frail elderly: A national profile. The
(2006). Assessing family caregiver’s mental health Gerontologist, 27 (5): 616–626.
using a statistically derived cut-off score for the Zarit Stoppe, G. (2009). Competence assessment in dementia.
Burden Interview. Aging & Mental Health, 10 (2): Retrieved 5 August, 2009; Available at http://edcon-
107–111. dementia.net/en/consensus.php?id=4
Schulz, R., Belle, S., Czaja, S, et al. (2004). Long-term care Svanberg, E., Spector, A. and Stott, J. (2011). The impact
placement of dementia patients and caregiver health of young onset dementia on the family: A litera-
and well-being. Journal of the American Medical ture review. International Psychogeriatrics, 23 (03):
Asssociation, 292: 961–967. 356–371.
Schulz, R. and Martire, L.M. (2004). Family caregiving of Taylor, D.H., Jr., Kuchibhatla, M. and Ostbye, T. (2008).
persons with dementia: Prevalence, health effects, Trajectories of caregiving time provided by wives to
and support strategies. American Journal of Geriatric their husbands with dementia. Alzheimer Disease &
Psychiatry, 12: 240–249. Associated Disorders, 22 (2): 131–136.
Schulz, R., O’Brien, A.T., Bookwala, J. and Fleissner, K. te Boekhorst, S., Pot, A.M., Depla, M. et al. (2008). Group
(1995). Psychiatric and physical morbidity effects of living homes for older people with dementia: The
dementia caregiving: Prevalence, correlates, and effects on psychological distress of informal caregiv-
causes. The Gerontologist, 35 (6): 771–791. ers. Aging & Mental Health, 12 (6): 761–768.
Schulz, R. and Williamson, G.M. (1997). The measurement Teri, L., Logsdon, R.G., Peskind, E. et al. (2000). Treatment
of caregiver outcomes in Alzheimer disease research. of agitation in Alzheimer’s disease: A randomized
Alzheimer Disease and Associated Disorders, 11: placebo controlled clinical trial. Neurology, 55:
117–124. 1271–1278.
160 Dementia

Teri, L., Logsdon, R.G., Uomoto, J. and McCurry, S. van Vliet, D., de Vugt, M.E., Bakker, C. et al. (2010). Impact
(1997). Behavioral treatment of depression in demen- of early onset dementia on caregivers: A review.
tia patients: A controlled clinical trial. Journal of International Journal of Geriatric Psychiatry, 25 (11):
Gerontology: Psychological Sciences, 52B: P159–P166. 1091–1100.
Teri, L., McCurry, S.M., Logsdon, R. and Gibbons, L.E. Vernooij-Dassen, M., Draskovic, I., McCleery, J. and
(2005). Training community consultants to help family Downs, M. (2011). Cognitive reframing for carers of
members improve dementia care: A randomized con- people with dementia. The Cochrane Database of
trolled trial. The Gerontologist, 45 (6): 802–811. Systematic Reviews, 11: CD005318.
Thompson, C.A., Spilsbury, K., Hall, J. et al. (2007). Vitaliano, P.P., Zhang, J. and Scanlan, J.M. (2003). Is care-
Systematic review of information and support inter- giving hazardous to one’s physical health? A meta-
ventions for caregivers of people with dementia. BMC analysis. Psychological Bulletin, 129: 946–972.
Geriatrics. 7 (1): 18. Wagner, D.L. (1997). Comparative Analysis of Caregiver
Tornatore, J. and Grant, L. (2002). Burden among family Data for Caregivers to the Elderly 1987 and 1997.
caregivers of persons with Alzheimer’s disease in nurs- Bethesda, MD: National Alliance for Caregiving.
ing homes. The Gerontologist, 42: 497–506. Wimo, A., Winblad, B. and Jönsson, L. (2007). An estimate
Torti, F.M.J., Gwyther, L.P., Reed, S.D. et al. (2004). A of the total worldwide societal costs of dementia in
multinational review of recent trends and reports in 2005. Alzheimer’s & Dementia, 3 (2): 81–91.
dementia caregiver burden. Alzheimer Disease and Withall, A. (2013). The challenges of service provision in
Associated Disorders, 18 (2): 99–109. younger-onset dementia. Journal of the American
Tun, S., Murman, D., Long, H. et al. (2007). Predictive Medical Directors Association, 14 (4): 230–232.
validity of neuropsychiatric subgroups on nursing Withall, A., Draper, B., Seeher, K. and Brodaty, H.
home placement and survival in Alzheimer’s disease (2014). The prevalence and causes of younger onset
patients. American Journal of Geriatric Psychiatry, 15: dementia in Eastern Sydney, Australia. International
314–327. Psychogeriatrics, 26 (12): 1955–1965.
Van’t Leven, N., Prick, A.E.J.C., Groenewoud, J.G. et al. World Health Organization and Alzheimer’s Disease
(2013). Dyadic interventions for community-dwelling International (2012). Dementia – A Public Health
people with dementia and their family caregivers: A Priority. United Kindgom: World Health Organization.
systematic review. International Psychogeriatrics, 25 Zarit, S. and Femia, E. (2008). Behavioral and psychosocial
(10): 1581–1603. interventions for family caregivers. American Journal
van der Lee, J., Bakker, T.J.E.M., Duivenvoorden, H.J. and of Nursing, 108 (9 Suppl.): 47–53; quiz 53.
Dröes, R.-M. (2014). Multivariate models of subjective Zarit, S.H., Reever, K.E. and Bach, S. (1980). Relatives of
caregiver burden in dementia: A systematic review. the impaired elderly: Correlates of feeling of burden.
Ageing Research Reviews, 15 (0): 76–93. The Gerontologist, 20: 649–655.
15
Dementia care in the community: Challenges
for primary health and social care

STEVE ILIFFE, JILL MANTHORPE AND VARI DRENNAN

The same phenomenon of un-integrated services is found


15.1 INTRODUCTION throughout Europe (Waldemar et al., 2007), the United
States (Dementia Initiative, 2013) and Australia (Grealish
Dementia is one of the main causes of disability in later et al., 2013).
life. Although the incidence of dementia in industrial- Services for people with dementia must cross boundaries
ized countries may have reached its peak (Matthews, 2013; between health and social care agencies, between second-
Alzheimer’s Disease International [ADI], 2014), its impact ary and primary care sectors and between the disciplines
remains profound. In terms of the global burden of disease, of medicine, nursing, allied professions and social care. The
it contributes 11.2% of all years lived with disability; higher purpose of this chapter is to identify effective ways to cre-
than stroke (9.5%), musculoskeletal disorders (8.9%), heart ate such cross-boundary working in the sector where most
disease (5%) and cancer (2.4%) (World Health Organization, people with dementia will receive most services and sup-
2003). Nevertheless, in comparison with other areas of long- port: in primary care. Different healthcare systems and
term disease management, the care of people with dementia funding streams are unhelpful regarding a prescriptive
currently constitutes only a small proportion of primary approach to the disease, nevertheless, we believe there are
care family doctors’ workload with only three to four new key ideas, approaches and techniques that can cross cul-
cases a year handled by a doctor in a demographically aver- tures and systems. To help this process of learning from
age population. While family doctors may have few cases, others’ experiences, we have adopted a polyglot terminol-
their engagement with those patients and their families is ogy. Following this terminology, caregiver is used to mean
likely to be over a long period, as the median length of time family members, neighbours or friends who provide practi-
from symptom onset to death is 10 years for those under 65 cal and emotional support to people with dementia and who
years old at diagnosis and 4 years for those over 80 years are called carers or informal carers in the United Kingdom.
at diagnosis (Xie et al., 2008). Even allowing for delays in Family doctors are primary care physicians (PCPs) in some
diagnosis, median survival from diagnosis is 3.5 years (Rait parts of the world and general practitioners (GPs) in other
et al., 2010). As populations rapidly age, this situation will parts. Primary care nurses include those nurses who work
change. Healthcare systems in many countries are antici- alongside family doctors as well as specialist nurses work-
pating the need to develop new services and reconfigure ing in the community (like the United Kingdom’s District
existing ones to respond to the increasing prevalence of Nurses or Community Nurses), particularly those working
dementia. with older people. Social workers do different things in dif-
In England, both the National Institute for Clinical ferent countries, but they all seem able to recognize each
Excellence and Social Care Institute for Excellence (NICE/ other and have a common practice base (Manthorpe and
SCIE) dementia guidelines (NICE, 2006) and the National Moriarty, 2007). Social care and support, on the other hand,
Dementia Strategy (Department of Health, 2009) proposed may be provided by commercial, local government or com-
a systematic approach to the continuing care of people with munity, voluntary and religious organizations, be part of
dementia in the community, to correct the evident deficits healthcare systems or separately funded.
in care identified over a period of many years, by a range Underpinning our argument is the knowledge that pro-
of bodies such as, the Alzheimer’s Disease Society (1995). posals for skill acquisition or service development rest on a

161
162 Dementia

weak evidence base, particularly for psychosocial interven- older person living on their own (Manthorpe and Iliffe,
tions. The systematic reviews that we cite make it clear that 2007) and suggest a consultation. However, the stigma of
the paucity of evidence arises from the quality of studies such a diagnosis, a general lack of awareness of the condi-
rather than the number. Only with the cholinesterase inhib- tion and the impression that little can be done for some-
itor drugs can we be confident that the evidence of effective- one suffering from an incurable disease may often block
ness is grounded in large trials with realistic time-frames. the diagnostic pathway preventing the patient or others
Dementia syndromes are progressive neurodegenerative from seeking medical help, because of embarrassment,
diseases which follow a variable but inexorable trajectory. shame, fear or uncertainty (Bradford et al., 2009). This
We have organized this chapter to follow that trajectory, delay occurs despite evidence that early cognitive impair-
starting with early symptoms, moving to diagnosis and ment is associated with increased frequency of consulta-
early support, then mid-stage dementia and behavioural tions in primary care (Ramakers et al., 2007).
and psychological disorders (BSPD). The chapter ends with The presenting symptoms of dementia may vary and do
a discussion of better care at home and in long-term care not always include memory loss. For example, in one study,
facilities such as care homes and nursing homes, including memory loss was the symptom that prompted help seek-
end-of-life care. In each section we attempt to answer four ing in 85% of new diagnoses of dementia, but 63% also had
questions: (1) what is known about this subject? (2) what do other behavioural and psychological symptoms of dementia
primary care practitioners need to learn? (3) how will they (BPSD) (Shigematsu, 2011). Symptoms can also fluctuate,
know they are effective? and (4) what service developments complicating decision-making, especially in mild cognitive
are needed? impairment, when between 40%–70% of patients notice an
improvement in their symptoms or are found not to have
dementia when reviewed (Le Couteur et al., 2013, Mitchell
and Shiri-Feshki, 2009).
15.2 EARLY RECOGNITION OF Some may make an initial, potentially inaccurate diag-
DEMENTIA IN PRIMARY CARE nosis of depression, although depression may coexist with
dementia.
15.2.1 WHAT IS KNOWN ABOUT THIS Early recognition of dementia syndrome is an area of
SUBJECT? practice where there is considerable potential for uncer-
tainty and confusion, including the difficulty in distin-
Although this is a surprisingly understudied subject (Le guishing between the (adverse) consequences of receiving
Couteur et al., 2013), the perceived benefits of reaching a the diagnosis report of dementia and the problems that
diagnosis include ending uncertainty about the cause of occur while living with dementia. Studies of older people’s
symptoms and behaviour change and promoting greater experiences of the assessment ‘journey’ have found that
understanding among family members with respect to many report feelings of confusion, uncertainty and anxiety
the problems experienced by the person suffering from over seemingly inexplicable time of waiting required in the
dementia. The aim is to increase access to helpful support diagnosis (Samsi et al., 2014). Some may be highly critical of
resources, to promote positive coping strategies and to facil- the systemic process of assessment and diagnosis disclosure
itate planning and fulfilment of short-term goals (Bamford although, generally positive of the practice of individual
et al., 2004). Using cholinesterase inhibitor medication to professionals (Samsi et al., 2014).
modify symptoms in a worthwhile way for a minority of
people with dementia who have Alzheimer’s disease is a 15.2.2 WHAT DO PRACTITIONERS NEED
further advantage (NICE, 2006). TO LEARN?
Dementia is probably under-diagnosed and under-
treated. Wide variation in family doctors’ abilities and Although lack of insight may be an early feature of the
confidence in diagnosing and managing dementia has dementia syndrome, people with dementia may be more
been consistently reported across different countries aware of their diagnosis than is suspected, because older
(Pucci et al., 2004). There is also evidence that diagnosis people and the general public are now much informed about
is sometimes delayed, with misattributed early symptoms dementia and its symptoms. Many primary care nurses and
as part of the process of normal ageing by both people social care staff are often asked if they think an older per-
with dementia and their families as well as professionals. son is showing symptoms of dementia. Some primary care
Carers and family members often prompt the seeking of nurses might lack confidence in responding to these ques-
a diagnosis as they may recognize symptoms before the tions, so they need to be knowledgeable about early symp-
person is affected, especially after a trigger event in the toms of dementia and also familiar with the locally agreed
patient’s life or health such as bereavement. Changes in first points of contact for people with such anxieties if they
personality and/or mood may also lead family members to are to answer such questions with confidence.
ask family doctors to identify what is wrong. Community Professional development among family doctors should
nurses and social care practitioners may be some of the aim to enrich the illness scripts that practitioners use to
first professionals to notice the changes that affect an recognize dementia, emphasizing the functional symptoms
Dementia care in the community: Challenges for primary health and social care 163

that can occur early in the disease process and reducing the at community level could postpone the onset of dementia
reliance on subjective memory loss as a cardinal symptom. syndromes and there is already some evidence of this hap-
Concerns about causing harm by over-zealous pursuit of pening in the United States (Langa et al., 2008) and other
early diagnosis can be offset by reframing the task as one industrialized countries (ADI, 2014). Public health initia-
of making timely diagnoses, in which changes in cognition tives may be situated in health services but they are as likely
or behaviour that worry the individual or those around to take place in sport, culture, environmental and transport
them are taken seriously, investigated and clarified. This is services, where opportunities for exercise and social partici-
essentially a clinical judgement rather than one that can be pation can be tuned to localities and populations.
made (in primary care) by use of cognitive function tests,
although such tests are useful adjunct to clinical reasoning.
15.3 DIAGNOSIS AND EARLY SUPPORT
15.2.3 HOW WILL THEY KNOW THEY ARE
EFFECTIVE?
15.3.1 WHAT IS KNOWN ABOUT THIS
Enhanced skills in earlier recognition of dementia will SUBJECT?
help to minimize misattribution and reduce the length of
time from symptom onset to diagnostic assessment, so that The problem of under-diagnosis of dementia is probably not
specialist services will see a change in their case mix, with due to lack of diagnostic skills, but rather due to the inter-
fewer late presentations during crises and more early ones. action of case-complexity, pressure on time and the nega-
Clinical records will contain more entries about risks being tive effects of reimbursement systems (Stoppe et al., 2007;
discussed and diagnostic hazards managed. These needs to Hinton et al., 2007). PCPs appear to adopt a watchful wait-
be shared with primary care nurses and social care prac- ing stance when it comes to people with potential demen-
titioners to ensure that mixed messages are not given to tia symptoms (Bamford et al., 2007). This may be due to a
the carers. Individuals will report processes that are clear, number of factors, including the tendency to assume that
person-centred and accommodate individuals’ preferences. such cognitive changes are merely due to ‘old age’ (Vernooij-
Professionals will provide accurate information, outline the Dassen, 2005), limited access to specialist mental health ser-
options and explain their relevance, including potential to vices and restrictions on the prescribing of cholinesterase
appoint proxy decision makers, draw up advance decisions inhibitors in the early stages of the illness in some countries.
and communicate wishes and preferences. Their patient In dementia, diagnosis is not a single act but a stepwise
record systems will contain such information and share process. De Lepeliere et al. (2008) explored the variety of
it with the consent of their patients or when in their best techniques used for detection of dementia throughout
interests. Europe and concluded that a systematic, stepwise strategy
could be employed to improve timely detection of demen-
15.2.4 WHAT SERVICE DEVELOPMENTS tia; that referral pathways could be more effective than
ARE NEEDED? specific guidelines; and that a strong, multidisciplinary
service infrastructure enables the diagnostic process. In
Professional development across community-based dis- other words, diagnosis may depend on the availability of
ciplines is the first priority in our view, although in some ‘­treatment’ – in the case of dementia this includes psycho-
countries, such as England, the development of memory social support for patients and their social networks as well
clinics has overtaken this in terms of investment. In addi- as medication.
tion to this, mechanisms are required at local levels for Telling people their diagnosis does seem to be match-
developing and disseminating agreed service access routes, ing with people suffering from dementia (Jha et al., 2001)
or care pathways for people with symptoms or concerns and younger professionals that their service provided is
across agencies. Without such ‘maps’, the potential for con- valuable (Sullivan and O’Connor, 2001) is essential, but
flict, omissions and delay increases (Waldorff et al., 2001), this is knowledge gleaned from those who seek help, not
but such maps need to be accurate and updated (Samsi and those who avoid it. The fear of triggering individual dis-
Manthorpe, 2014) and are no replacement for advice and tress, denial and withdrawal from contact with services
consideration of personal circumstances. is one factor that inhibits practitioners from discussing
There is a growing consensus that prevention of demen- dementia as a diagnosis (Iliffe and Wilcock, 2005) and
tia is practical and feasible. Service commissioners and pro- there appears to be a lack of clinical skill in managing this
viders should strengthen public health initiatives that have diagnostic transition (Samsi et al., 2014). Disclosure of the
the potential to reduce dementia prevalence by modifying suspected or a certain diagnosis is rated by PCPs as one
vascular risk in particular. Cognitive function changes are of the most difficult areas in dementia management and
measurable in people with cardiovascular risk factors in they are more likely than psychiatrists to use euphemisms
their middle age, making targeted primary prevention of (Bamford et al., 2004). The use of such euphemisms com-
both heart and brain disease imperatives. There is the real plicates relationships with other primary care practitio-
possibility that vigorous control of cardiovascular risks ners who may be uncertain about what the patient has
164 Dementia

been told and confused about whether to suggest the of shared understandings of professional practice and pres-
patient to be in touch with a support group such as the sures undergone by all (Iliffe et al., 2009). Learning in this
Alzheimer’s Society or otherwise and further exasperated way can usefully include people who have experienced ser-
as to what to record in their own notes. vices earlier, but it still may not be enough to change clini-
Counselling and support may preserve caregivers’ self- cal practice among family doctors in the absence of extra
rated health if they are already engaged in emotional or resources (Wilcock et al., 2013).
practical care work (Mittelman et al., 2007). Psychosocial
interventions that include group activity can reduce care- 15.3.3 HOW WILL THEY KNOW THEY ARE
giver’s feelings of ‘burden’ and increase their satisfac- EFFECTIVE?
tion (Andren and Elmståhl, 2008; Bunn et al., 2012). The
characteristics of effective psychosocial interventions are: People with dementia and their carers often report that
psycho-education (which addresses barriers to change); the breaking of the ‘bad news’ was handled as well as it
a family system’s perspective; being multifaceted in solving could be by others and that they did not feel abandoned.
problems and being able to apply them in a flexible manner Practitioners will know that the diagnostic process is
to meet different needs in different families. Specialist nurs- customized according to the people that they are seeing
ing services that have an explicit focus on supporting care- if there is an evident reduction in depressive responses,
givers, such as Admiral Nurses in the United Kingdom, are catastrophizing and if caregivers report less sense of aban-
highly valued, although there is limited evidence of effect donment. Reports from voluntary or third sector groups
on psychosocial impacts (Bunn et al., 2015). are useful as feedback experiences and as a link in the qual-
ity assurance feedback chain. Feedback from other practi-
15.3.2 WHAT DO PRACTITIONERS NEED tioners would be that the process or transition has been
TO LEARN? handled as well as possible and that, rather than avoiding
‘putting people through’ such a process they would have
A person-centred approach may help alleviate four key confidence that it will be supportive. Care home manag-
problems: fears associated with other people ‘finding out’ ers, for example, could be asked about the experiences of
the diagnosis, rapid deterioration in abilities, socially residents who may be showing signs suggestive of demen-
embarrassing behaviour and a loss of involvement in care tia. Would such a manager see it as being in the resident’s
planning (Edwards et al., 2014). The techniques avail- best interests to suggest mentioning this to the family doc-
able to all practitioners in primary care include combi- tor? What is the experience of staff who may accompany
nations of reality orientation, memory strategies and a resident to a memory assessment service or what is the
reframing. Reality orientation, for example, focusses on outcome of a visit from a memory assessment practitioner
the likely slow progression in early dementia and offsets to a resident?
catastrophic fears that may be triggered by mild memory
lapses. Memory enhancement strategies include short- 15.3.4 WHAT SERVICE DEVELOPMENTS
term goal setting and maintaining social and family roles ARE NEEDEED?
that reinforce memory. Reframing dementia as a disabil-
ity that can be accommodated shifts the emphasis from De Lepeliere et al. (2008) emphasizes the need for primary
preoccupations with the diagnosis to not being perceived and secondary collaboration in the diagnostic process
as a fool and fosters understanding of the anger or frus- and there is a case for creating integrated dementia care
trations associated with inabilities to perform daily tasks services (Bullock et al., 2007). Support groups for people
(Robinson et al., 2010). with newly diagnosed dementia seem to promote well-
Reluctance to make use of services or support is not being. These may be experiential group therapy allowing
uncommon, and practitioners need to remember that opt- people to explore the experience of dementia in a safe, sup-
ing for services is a result of a complex decision-making portive and secure setting (Bunn et al., 2012) or have less
process often unrelated to objective circumstances. Conflict ambitious aims of peer support. Memory cafes are ways
theory provides useful insights into such decision-making, in which social settings can be adapted or tailor-made for
particularly service avoidance (Markle-Reid and Browne, people with dementia, although, they are mostly found
2001), but we must remember that a number of services may in urban areas. Other groups for older people may focus
be inappropriate for people with early symptoms of demen- on couples or particular groups, such as men in particu-
tia and neither they nor their families may see them as good lar (Manthorpe and Moniz-Cook, 2008). Such efforts to
quality services (Bunn et al., 2013). widen the menu of support rests on the evidence that many
Dementia is one domain where training is not often caregivers of people with dementia make infrequent use of
needed for it may be confidence that is lacking rather than services (Brodaty et al., 2005) because they do not think
knowledge, or the opportunities to learn new skills and to they need those services. People with dementia may also
then put them into practise. There is an advantage in using be reluctant to use services. The accessibility and quality of
adult learning methods across professions and for profes- many services have been low compounding the stigma and
sionals to learn together. This can produce the added value negativism of dementia itself.
Dementia care in the community: Challenges for primary health and social care 165

view of incontinence in older people. Urinary inconti-


15.4 MID-STAGE DEMENTIA AND BPSD nence is more common in people with dementia than their
peers without dementia and those with both dementia and
15.4.1 WHAT IS KNOWN ABOUT THIS urinary incontinence are more likely to be catheterized
SUBJECT? (Grant et al., 2013).
There is some evidence to suggest that in the mid stage
Non-cognitive symptoms, from agitation and pacing around only a very small number of people with dementia have
to wandering and getting lost, are particularly distressing incontinence problems that cannot be treated or ame-
for families and social care staff. These BPSD are ‘behaviour liorated (Harari and Igbedioh, 2009), but it is an under-
that challenges’ and are common. Up to 20%–90% of people researched area (Drennan et al., 2012). A recent European
with dementia will experience BPSD at some time, particu- survey of 1181 family carers of people with dementia
larly, in the middle and later stages of the illness (Robinson reported that incontinence symptoms were more prob-
et al., 2010). BPSD precipitate crises and hospital admissions lematic rather than behavioural symptoms (Georges et al.,
and are associated with abuse and neglect. Although, the 2008). In part this may reflect the interface between loss
risks from BPSD are often not as high as many fear, BPSD of independence and behavioural problems. Toileting dif-
can lead to high levels of caregiver stress and curtailment ficulties described by carers include use of an inappropri-
of the activities of the person with dementia and can be ate receptacle or place, passive urinating or defecating
the crucial factor that leads to a move to a long-term care without active attempts to reach a toilet, hiding soiled or
facility (Balestreri et al., 2000). wet clothing, inability to clean after defaecation and clean-
Individual behaviour management therapy can reduce ing faeces off their hands inappropriately (Drennan et al.,
caregiver perceptions of problems, with a lasting effect 2011). Contributory factors for some of these behaviours
(Selwood et al., 2007). A recent systematic review (Livingston may include difficult access to the toilet, neuropsycho-
et al., 2014) of non-pharmacological interventions for agita- logical dysfunction, depression, fear and embarrassment
tion in dementia found that person centred care, communi- or medication side effects (Stokes, 2000). Caregivers have
cation skills training and adapted dementia care mapping reported primary care professionals’ responses as less
(observation of staff and resident interactions to improve than helpful in these situations and in some instances
care responsiveness) decreased severe agitation in people found solutions that have the potential to distress or harm
with dementia living in care homes immediately and this the person with dementia, such as restricting fluid intake
was sustained for up to 6 months afterwards. Activities and (Drennan et al., 2012). An absence of explicit discussion
music therapy by protocol decreased overall agitation while has been noted regarding the needs of people with demen-
sensory interventions decreased clinically significant agita- tia and their caregivers in best practiced incontinence and
tion immediately. service guidelines (Drennan et al., 2013). It has been sug-
There is widespread concern over the hazards of using gested that well contained incontinence should be consid-
antipsychotic drugs in dementia (Schneider et al., 2006) and ered as an objective in care planning and in supporting
these should be used sparingly for short periods and with caregivers (DuBeau et al., 2010). However, it is clear that
specialist advice. Recent positive evidence about the clinical the multifactorial nature of the problems require a broad
and cost-effectiveness of a manual based coping strategy has repertoire of types of solutions and responses from profes-
been provided by Livingston et al. (2014) where adherence sionals and services.
to the START (STrAtegies for RelaTives) manual reduced The broad objectives of primary care and social care
affective symptoms and case level depression among care- professionals at this point are: to maintain cognition and
givers. Caregivers’ quality of life also improved with this daily living skills, to reduce excess disability and distress,
intervention. to maintain the quality of life of patient and carers and to
Literature on the role of GPs in dementia has focussed sustain living in the community. Assessment of care quality
mainly on early diagnosis, access, attitudes, education and for people at this stage is complex and context-dependent
training, burden of workload and responsibility, but the (Zermansky et al., 2007), so a holistic approach is needed
quality of medical care they deliver to people with dementia (McMurdo and Witham 2007). Judgements passed about
in mid stage has been neglected. Although multiple comor- the quality of care are necessarily subjective and any qual-
bidities are equally common in older people with or without ity improvement should take into account not only the per-
dementia, people with dementia are less likely to report to spectives of the care providers but also of individuals and
be diagnosed or to receive optimal care for other comorbidi- their supporters (Orrell et al., 2008; Robinson et al., 2007 a).
ties (Connolly et al., 2013). Case management of people with dementia (includ-
The responses to incontinence symptoms are one exam- ing systematic follow up, provision of brief psychological
ple of this. Embarrassment may prevent the individual therapy and medication management) has the potential to
from seeking help, supported by carers who are trying provide a holistic approach and overcome service fragmen-
to protect the individual’s integrity in the outside world tation; it has been tested in the United States (Vickrey et al.,
(Drennan et al., 2011), the patient gains confidence. There 2006; Callahan et al., 2006) and Europe (Eloniemi-Sulkava
is also evidence that health professionals have a nihilistic et al., 2009).
166 Dementia

Case management for people with dementia can reduce admissions and limited or short-term psychotropic drug
hospital and emergency admissions (Vickrey et al., 2006), mit- use will be signs that dementia risks are being addressed
igate embarrassment, isolation and relationship strain (Clark effectively. Nevertheless, prevention may be a better solu-
et al., 2004) and caregiver stress (Callahan et al., 2006) as well tion. This may be assisted by tailor-made support plans,
as delay relocation of the person with dementia to a care home the use of skilled and experienced staff who can under-
(Pinquart and Sorensen, 2006). However, few studies have take diversionary and pleasurable activities and spot signs
recorded large effects (Koch et al., 2012). The heterogeneity of of distress, access physical activities and stimulate sympa-
patients in dementia case management studies and the lack thetic environments as well as understand people’s wishes,
of subgroup analyses make it difficult to identify the disease habits and patterns of activity and behaviour. Good care or
stage at which patients and their carers most benefit. A recent support planning will contain information that is transfer-
systematic review of randomized controlled trials (RCTs) of able but communication patterns need continual rehears-
case management for people with dementia and their carers ing. Audit and review can be the hallmark of support that is
concluded that evidence for the efficacy of case management seeking to be effective.
remains equivocal (Pimouguet et al., 2010).
A modelling and feasibility study in four general prac- 15.4.4 WHAT SERVICE DEVELOPMENTS
tices in the United Kingdom (Iliffe et al., 2014) suggests that ARE NEEDED?
case management of people with dementia, whether prac-
ticed by experienced nurses in dedicated sessional time, Investment in counselling, support planning and review
or by a seconded social worker devoted exclusively to case for people with dementia and caregivers and skill develop-
management, does not fit easily into practice routines. ment for community nurses, social workers and care home
staff may be more useful than investment in ‘memory clin-
15.4.2 WHAT DO PRACTITIONERS NEED ics’, unless they are able to undertake these support tasks
TO LEARN? in a consistent way. Commissioners or funders of care
and support may be able to use budgets in a flexible man-
The strategies people with dementia and their immediate ner to bolster or reward good preventive strategies rather
caregivers develop to protect their external persona and dig- than directing funds towards crises. Training and support
nity may mean that the extent of problems and their con- of front line workers may take much more of professionals’
sequences are not disclosed to professionals without direct time than individual case work, raising new challenges for
appropriately worded enquiry. Training in simple methods professionals to be in a position to demonstrate that their
for recognizing, understanding and responding to BPSD, input in supervision, training and prevention is effective
stress and continence problems should be part of profes- and efficient. Further, modelling studies are needed to clar-
sional development across disciplines. Social care providers ify the purpose of case management, to revisit the skills and
need help to develop and sustain basic levels of understand- attributes required for case management, to embed deliv-
ing and support from primary care teams. Opportunities ery of case management in primary care and to establish
for them to develop skills can be delivered by co-working whether the illness trajectory of dementia case management
rather than taking them away from care settings. Care has maximum cost-effectiveness.
staff can benefit from opportunities for reflective practice
in supervision. Mentoring through opportunities to learn
from senior care staff is possible if such care practitioners
15.5 HIGHER INTENSITY AND END
are not required to move away from care delivery. Night
staff and temporary or part-time workers need to be given
OF LIFE CARE (AT HOME AND IN
opportunities to develop their skills, particularly, given the CARE HOMES)
stresses of lone working or managing when staff is less in
number. Family doctors working at night or at weekends 15.5.1 WHAT IS KNOWN ABOUT THIS
need to be able to assist care staff or caregivers in difficulty, SUBJECT?
if necessary and risky hospital admission is to be avoided.
Given the hazards of using antipsychotic medication, fam- Specialist home care teams have (in some cases) demon-
ily doctors need to understand the limits of medication as strated greater flexibility and responsiveness to caregivers
a solution to BPSD and should be able to obtain specialist than task-oriented care services and they may also reduce
advice quickly and easily. caregiver stress and prevent crises. These outcomes depend
on the configuration of the service, including multidisci-
15.4.3 HOW WILL THEY KNOW THEY ARE plinary health and social care services’ input, care worker
EFFECTIVE? autonomy, continuous reassessment of client’s circum-
stances and capacity to develop long-term relationships
Effective management of BSPD will be shown in preven- through care-worker continuity (Rothera et al., 2008). Poor
tion and in symptom management if necessary. Fewer cri- quality services, of course, often lead to poor quality out-
ses, fewer out-of-hours (OOH) contacts, reduced hospital comes and it is rare for specialist services to be funded or
Dementia care in the community: Challenges for primary health and social care 167

employed in the same way as standard services, with their 15.5.2 WHAT DO PRACTITIONERS NEED
frequent problems of high staff turnover, short interven- TO LEARN?
tions (e.g. 15 minutes to undertake personal care and meal
preparation) and inflexibilities. There are promising results Understanding the caregiver’s strengths and their rela-
from ‘cash for care’ schemes in many parts of the developed tionship with the person suffering from dementia may be
world where individual or personal budgets can potentially an important guide to how support is offered and given.
provide much better support by responding to people’s Offering support early should become part of routine prac-
needs at the time necessary and supporting rather than tice, but only if the professional knows where to signpost the
undermining family and other assistance (Moran et al., patient or caregiver. Local service maps are important so
2014). However, the risk that these places additionally bur- that patients and carers do not face the frustration of being
den caregivers with, needs to be recognized. In some coun- passed from pillar to post. Professionals need to know the
tries, the effectiveness of community care polices enabling best sources of information about their locality and how to
people to stay at home means that people who do eventually report unmet needs to those responsible for locality plan-
enter care homes are often very ill or frail. High intensity ning and service development. Sometimes, these may be
care may be akin to palliative care. consolidated as part of dementia care pathways (Samsi and
The move to a care home may be determined by the Manthorpe, 2014), but like most information these need to
severity of cognitive impairment and functional loss, the be updated from time to time to be accurate.
presence of depression in the person with dementia and (in Compared with those with other conditions, people with
some but not all studies) incontinence (Thomas et al., 2004). dementia are more likely to experience symptoms, includ-
In the United Kingdom, the majority of people with ing persistent pain and are more likely to be untreated in
dementia live and die at home or in a care home rather than the last 6 months of life (McCarthy et al., 1997). The focus
in hospital (dying with dementia is rare in a hospice). There of attention may be on other problems, like nutrition and
are three ways in which people with dementia can die (Cox hydration, with pain being relatively neglected (Goodman
and Cook, 2002). First, there are people who have been diag- et al., 2010). This is a complex dilemma because of the
nosed of dementia, but their death is caused by another med- communication difficulties that are a feature of advanced
ical condition (e.g. cancer or heart disease). Second, people dementia and should, therefore, be the focus of professional
may die with interplay of another illness and dementia, development for all practitioners who contribute to end-of-
where the dementia has not impacted greatly on their func- life care. Early attention to such matters among those who
tioning. Third, there are people who are described as hav- may wish to make plans for their future care and treat-
ing end-stage dementia, where the associated consequences ment may be part of primary care consultations. In coun-
of the dementia impact upon all domains of their life and tries, such as England, there is evidence that more people
they ultimately die of the complications of this condition. with dementia are moving into care facilities with proxy
Each of these different ways will directly influence the place decision-makers appointed and statements of wishes in
­
and experience of death for an individual and their fam- place (Manthorpe and Samsi, 2014), meaning, that, primary
ily members and primary care involvement. A recent study care and care providers will need to share such information
talking to people with dementia living in care homes about and discuss its implications.
approaching end-of-life suggested that there was value in
creating opportunities to talk about these issues (Goodman 15.5.3 HOW WILL THEY KNOW THEY ARE
et al., 2013). There is, therefore, not a single experience and EFFECTIVE?
some patients and carers live with dementia for years often
with different trajectories of functional decline and needs Good care homes and good home care services can reduce
(Mularski et al., 2007; Barclay et al., 2014). fewer inappropriate hospital admissions and transfers as
Professional and policy guidance on care for people with well as out of hours contact. Good care homes will meet reg-
dementia nearing the end of life emphasizes the benefits ulatory demands but there will also be confidence among
of advance care planning, coordinated working between local communities and perhaps a sense of pride that a local
health and social care and the adaptation and use of pal- care home is part of the community. Care homes that work
liative care frameworks and clinical tools for people with with volunteers and are supportive of residents’ families
long-term conditions (Alzheimer Europe, 2008). However, will become well known. Local professionals often know
policy guidance and end-of-life initiatives, though laud- which homes they would want to enter or where they might
able, have outstripped the available evidence on end-of-life wish a relative to live if the need arose. We will know care
care for people with dementia and their caregivers that live homes are effective if they have limited staff turnover and
at home or in care homes (Goodman et al., 2010). There is if there are (short) waiting lists for their services. The same
some evidence that processes and emphasis on commu- applies to home care services. Staff may be able to support
nication and collaborative problem solving between fam- each other if this culture is fostered. They will then be more
ily doctors, care home staff and visiting specialist services able to offer support for families and other residents who
can improve the quality of end-of-life care and reduce costs are bereaved. Fewer admissions to hospital and greater con-
(Amador et al., 2014). fidence that people’s statements of wishes and any advance
168 Dementia

decisions will be respected can be indicators that end-of-life Andren, S. and Elmståhl, S. (2008). Psychosocial interven-
care is being well managed. Audits of records and face to tions for family caregivers of people with dementia
face quality assurance data collection will prove that peo- reduces caregiver’s burden: Development and effect
ple’s ability to make decisions and choices has been assessed after 6 and 12 months. Scandinavian Journal of Caring
and respected. Sciences, 22: 98–109.
Balestreri, L., Grossberg, A. and Grossberg, G.T. (2000).
15.5.4 WHAT SERVICE DEVELOPMENTS Behavioural and psychological symptoms of demen-
tia as a risk factor for nursing home placement.
ARE NEEDED?
International Psychogeriatrics, 12: 59–62.
The salience of flexibility, autonomy and responsiveness in Bamford, C., Lamont, S., Eccles, M. et al. (2004).
home care teams is an important lesson for other service Disclosing a diagnosis of dementia: A systematic
providers which might help others to learn lessons from review. International Journal of Geriatric Psychiatry, 19:
home care management’s style of functioning. End-of-life 151–169.
guidelines should be reviewed and monitored to make sure Bamford, C., Eccles, M., Steen, N. and Robinson, L. (2007).
that they are realistic and do not add to a ‘tick box culture’. Can primary care record review facilitate earlier diag-
Staff may need time to develop skills in this area and to pass nosis of dementia? Family Practice, 24: 108–116.
on these skills to new staff and to family members. The sup- Barclay, S., Froggatt, K., Crang, C. et al. (2014). Living in
port services for staff in large organizations, such as, coun- uncertain times: Trajectories to death in residential
selling or access to in-house employment support services care homes. British Journal of General Practice, 64
should be thought about in small or medium care enter- (626): e576–e583.
prises. Time for supervision and reflection should be built Bradford, A., Kunik, M.E., Schulz, P. et al. (2009). Missed
into contracts for care services. and delayed diagnosis of dementia in primary care:
Prevalence and contributing factors. Alzheimer
Disease and Associated Disorders, 23: 306–314.
Brodaty, H., Thomson, C., Thompson, C. and
15.6 CONCLUSION Fine, M. (2005). Why caregivers of people with demen-
tia and memory loss don’t use services. International
Kodner’s (2006) analysis of a successful model of inte- Journal of Geriatric Psychiatry, 20: 537–546.
grated care for frail older adults argues that provider net- Bullock, R., Passmore, P. and Iliffe, S. (2007). Can we
works which join together through standardized referral afford not to have integrated dementia services?
procedures, service agreements, joint training, shared Age & Ageing, 36: 357–358.
information systems and even common ownership of Bunn, F., Goodman, C. and Drennan, V.M. (2015).
resources enhance access to services, provide seamless Specialist nursing and community support for the
care and maintain quality. In this chapter we have drawn carers of people with dementia living at home:
on this and other evidence to suggest ways to create such An evidence synthesis. Health & Social Care in the
cross-boundary work in the sector where most people with Community, 24 (1): 48–67.
dementia will receive most services and support/primary Bunn, F., Goodman, C., Sworn, K. et al. (2012). Psychosocial
care. factors that shape patient and carer experiences of
dementia diagnosis and treatment: A systematic review
of qualitative studies. PLoS Med, 9 (10): e1001331.
REFERENCES Bunn, F., Sworn, K., Brayne, C. et al. (2013). Contextualizing
the findings of a systematic review on patient and
ADI (2014). Dementia and Risk Reduction: An Analysis carer experiences of dementia diagnosis and treat-
of Protective and Modifiable Factors. London: ment: A qualitative study. Health Expectations, 18 (5):
Alzheimer’s Disease International. 740–753.
Alzheimer’s Disease Society (1995). Dementia in the Callahan, C., Boustani, M., Unverzagt, F. et al. (2006).
Community: Management Strategies for General Effectiveness of collaborative care for older adults
Practice. London: Alzheimer’s Disease Society. with Alzheimer’s disease in primary care: A random-
Alzheimer Europe (2008). Position and ized controlled trial. JAMA, 295: 18.
Recommendations on End-of-Life Care. Luxembourg: Clark, P.A., Bass, D.M., Looman, W.J. et al. (2004).
Alzheimer Europe. Outcomes for patients with dementia from the
Amador, S., Goodman, C., King, D. et al. (2014). Exploring Cleveland Alzheimer’s managed are demonstration.
resource use and associated costs in end-of-life care Aging & Mental Health, 8: 40–51.
for older people with dementia in residential care Connolly, A., Campbell, S., Gaehl, E. et al. (2013). Under-
homes. International Journal of Geriatric Psychiatry, 29: provision of medical care for people with dementia in
758–766. doi:10.1002/gps.4061 primary care. BJGP, 63: 75–76.
Dementia care in the community: Challenges for primary health and social care 169

Cox, S. and Cook, A. (2002). Caring for people with Grant, R.L., Drennan, V.M., Rait, G. et al. (2013). First
dementia at the end of life. In J. Hockley and D. Clark. diagnosis and management of incontinence in older
(eds.), Palliative Care for Older People in Care Homes. people with and without dementia in primary care:
Buckingham: Open University Press, pp. 86–103. A cohort study using the health improvement network
De Lepeliere, J., Wind, A., Iliffe, S. et al. and the Interdem primary care database. PLoS Med, 10 (8): e1001505.
Group. (2008). The primary care diagnosis of dementia Grealish, L., Jamieson, M., Brown, J. et al. (2013). The
in Europe: An analysis using multi-disciplinary, multi- Interaction between Hospital and Community-based
national expert groups, Aging and Mental Health, 12: Services for People with Dementia and their Carers.
568–576. Sydney: Alzheimer’s Australia NSW.
Dementia Initiative (2013). Dementia care: The quality Harari, D. and Igbedioh, C. (2009). Restoring continence
chasm. Available at http://www.ccal.org/wp-content/ in frail older people living in the community: What
uploads/DementiaCareTheQualityChasm_020413. factors influence successful treatment outcomes?
pdf Department of Health (2009). Living Well with Age Ageing, 38: 228–233.
Dementia: The National Dementia Strategy. London: Hinton, L., Franz, C., Reddy, G. et al. (2007). Practice
Department of Health. constraints, behavioural problems and dementia
Drennan, V.M., Cole, L. and Iliffe, S. (2011). A taboo within care: Primary care physicians perspectives. Journal of
a stigma? A qualitative study of managing inconti- General Internal Medicine, 22 (11): 1625–1627.
nence with people with dementia living at home. BMC Iliffe, S., Jain, P., Wong, G. et al. (2009). Dementia diag-
Geriatrics, 14 (11): 75. nosis in primary care: Looking outside the educational
Drennan, V.M., Greenwood, N., Cole, L. et al. (2012). box. Aging Health, 5: 51–59.
Conservative interventions for incontinence in people Iliffe, S., Robinson, L., Bamford, C. et al. CAREDEM
with dementia or cognitive impairment, living at home: research team. (2014). Introducing case management
A systematic review. BMC Geriatrics, 12 (1): 1. for people with dementia in primary care: A mixed-
Drennan, V.M., Norrie, C., Cole, L. and Donovan, S. (2013). methods study. The British Journal of General Practice,
Addressing incontinence for people with dementia 64 (628): e735–e741.
living at home: A documentary analysis of local English Iliffe, S. and Wilcock, J. (2005). The identification of bar-
community nursing service continence policies and riers to the recognition of and response to dementia
clinical guidance. Journal of Clinical Nursing. 22: in primary care using a modified focus group method.
339–346. Dementia, 4: 12–23.
DuBeau, C.E., Kuchel, G.A., Johnson, T. et al. (2010). Jha, A., Tabet, N. and Orrell, M. (2001). To tell or not to
Fourth International Consultation on Incontinence: tell – Comparison of older patients’ reaction to their
Incontinence in the frail elderly: Report from the diagnosis of dementia and depression. International
4th International Consultation on Incontinence. Journal of Geriatric Psychiatry, 16: 879–885.
Neurourolgy Urodynamics, 29 (1): 165–178. Koch, T., Iliffe, S., Manthorpe, J. et al. (2012). The poten-
Edwards, R., Voss, S., Iliffe, S. (2014). Education about tial of case management for people with dementia:
dementia in primary care: Is person-centredness the A commentary. International Journal of Geriatric
key? Dementia, 13(1): 111-119 Psychiatry, 27: 1305–1314.
Eloniemi-Sulkava, U., Saarenheimo, M., Laakkonen, M.L. Kodner, D. (2006). Whole-system approaches to health
et al. (2009). Family care as collaboration: and social care partnerships for the frail elderly: An
Effectiveness of a multi-component support pro- exploration of North American models and lessons.
gram for elderly couples with dementia. Randomized Health and Social Care in the Community, 14: 384–390.
controlled intervention study. Journal of the American Langa, K.M., Larson, E.B., Karlawish, J.H. et al. (2008).
Geriatrics Society, 57: 2200–2208. Trends in the prevalence and mortality of cognitive
Georges, J., Jansen, S., Jackson, J. (2008). Alzheimer’s impairment in the United States: Is there evidence of
disease in real life – The dementia carer’s survey. the compression of cognitive morbidity? Alzheimer’s
International Journal of Geriatric Psychiatry, 23: Disease & Dementia, 4: 134–144.
546–551. Le Couteur, D., Doust, J., Creasey, H. and Brayne,
Goodman, C., Amador, S., Elmore, N. et al. (2013). C. (2013). Political drive to screen for pre-dementia:
Preferences and priorities for ongoing and end-of-life Not evidence based and ignores the harms of
care: A qualitative study of older people with demen- ­diagnosis. BMJ, 347: f5125.
tia resident in care homes. International Journal of Livingston, G., Barber, J., Rapaport, P. et al. (2013). Clinical
Nursing Studies, 50 (12): 1639–1647. effectiveness of a manual based coping strategy pro-
Goodman, C., Evans, C., Wilcock, J. et al. (2010). End of gramme (START, STrAtegies for RelaTives) in promot-
life care for community dwelling older people with ing the mental health of carers of family members with
dementia: An integrated review. International Journal dementia: Pragmatic randomised controlled trial. BMJ,
of Geriatric Psychiatry, 25 (4): 329–337. 347: f6276. doi:10.1136/bmj.f6276
170 Dementia

Livingston, G., Kelly, L., Lewis-Holmes, E. et al. (2014). Pinquart, M. and Sorensen, S. (2006). Helping caregiv-
Non-pharmacological interventions for agitation in ers of persons with dementia: Which interventions
dementia: Systematic review of randomised controlled work and how large are their effects? International
trials. The British Journal of Psychiatry, 205: 436–442. Psychogeriatrics, 18 (4): 577–595.
Manthorpe, J. and Moriarty, J. (2007). Models from other Pucci, E., Angelerie, F., Borsetti, G. et al. (2004). General
countries: Social work with people with dementia and practitioners facing dementia: Are they fully prepared?
their caregivers. In C. Cox (ed.), Dementia and Social Neurological Sciences, 24: 384–389.
Work Practice. New York: Springer. Rait, G., Walters, K., Bottomley, C. et al. (2010). Survival of
Manthorpe, J. and Moniz Cook, E. (eds.) (2008). Early people with a clinical diagnosis of dementia in primary
Psychosocial Interventions in Dementia. London: care. BMJ, 341: c3584.
Jessica Kingsley. Ramakers, I., Visser, P., Aalten, P. et al. (2007). Symptoms
Manthorpe, J. and Samsi, K. (2014). Care homes of preclinical dementia in general practice up to
and the Mental Capacity Act 2005: Changes in five years before dementia diagnosis. Dementia and
under-standing and practice over time. Dementia, Geriatric Cognitive Disorders, 24: 300–306.
pii: 1471301214542623 [Epub ahead of print]. Robinson, L., Hutchings, D., Corner, L. et al. (2007a).
Markle-Reid, M. and Browne, G. (2001). Explaining the use Balancing rights and risks – Conflicting perspectives
and non-use of community-based long-term care ser- in the management of wandering in dementia. Health,
vices by caregivers of persons with dementia. Journal Risk and Society, 9: 389–486.
of Evaluation in Clinical Practice, 7: 271–287. Robinson, L. Hutchings, D., Dickinson, H.O. et al. (2007b).
Matthews, F.E., Arthur, A., Barnes, L.E. et al. Medical Effectiveness and acceptability of non-pharmacolog-
Research Council Cognitive Function and Ageing ical interventions to reduce wandering in dementia:
Collaboration. (2013). A two-decade comparison of A systematic review. International Journal of Geriatric
prevalence of dementia in individuals aged 65 years Psychiatry, 22: 9–22.
and older from three geographical areas of England: Robinson, L., Iliffe, S., Brayne, C. et al. (2010). Primary care
Results of the cognitive function and ageing study I and dementia: 2 Long-term care at home: Psychosocial
and II. Lancet, 382 (9902):1405–1412. interventions, information provision, carer support and
McCarthy, M., Addington-Hall, J. and Altmann, D. (1997). case management. International Journal of Geriatric
The experience of dying with dementia: A retrospec- Psychiatry, 25 (7): 657–664.
tive study. International Journal of Geriatric Psychiatry, Rothera, I., Jones, R., Harwood, R. et al. (2008). An evalua-
12: 404–409. tion of a specialist multiagency home support service for
McMurdo, M. and Witham, M.D. (2007). Health and wel- older people with dementia using qualitative methods.
fare of older people in care homes. BMJ, 334 (7600): International Journal of Geriatric Psychiatry, 23: 65-72.
913–914. Samsi, K. and Manthorpe, J. (2014). Care pathways for
Mitchell, A.J. and Shiri-Feshki, M. (2009). Rate of progres- dementia: Current perspectives. Clinical Interventions
sion of mild cognitive impairment to dementia – Meta- in Aging, 9 (1–9): 2055–2063.
analysis of 41 robust inception cohort studies. Acta Samsi, K., Abley, C., Campbell, S. et al. (2014). Negotiating
Psychiatrica Scandinavica, 119: 252–256. a labyrinth: Experiences of assessment and diagnos-
Mittelman, M.S., Roth, D.L., Clay, O.J. and Haley, W.E. tic journey in cognitive impairment and dementia.
(2007). Preserving health of Alzheimer’s caregiv- International Journal of Geriatric Psychiatry, 29: 58–67.
ers: Impact of a spouse caregiver intervention. The Selwood, A., Johnston, K., Katona, C. et al. (2007).
American Journal of Geriatric Psychiatry, 15: 780–789. Systematic review of the effect of psychological inter-
Mularski, R., Dy, S., Shugarman, L. et al. (2007). A ­systematic ventions on family caregivers of people with dementia.
review of measures of end-of-life care and its outcomes. Journal of Affective Disorders, 101: 75–89.
Health Services Research, 42 (5): 1848–1870. Schneider, L.S., Dagerman, K. and Insel, P.S. (2006).
NICE/SCIE (2006). Dementia: Supporting People with Efficacy and adverse effects of atypical antipsychot-
Dementia and their Carers in Health and Social Care. ics for dementia: Meta-analysis of random placebo-
London: National Institute for Clinical Excellence and controlled trials. The American Journal of Geriatric
Social Care Institute for Excellence (NICE/SCIE). Psychiatry, 14: 191–210.
Orrell, M., Hancock, G.A., Liyanage, K.C.G. et al. (2008). Shigematsu, K. (2011). Motivation of dementia patients
The needs of people with dementia in care homes: in seeking initial medical consultation observed by
The perspectives of users, staff and family caregivers. outpatient clinic (initial consultation tends to be late).
International Psychogeriatrics, 20: 941–951. Alzheimer’s & Dementia, 7 (4): S438.
Pimouguet, C., Lavaud, T., Dartigues, J.F. and Stoppe, G., Knoblauch, A., Haak, S. and Maeck, L. (2007).
Helmer, C. (2010). Dementia case management effec- Physicians competence regarding the early diagnosis
tiveness on health care costs and resource utilization: of dementia: Differences between family physicians
A systematic review of randomized controlled trials. The and primary care neuropsychiatrists in Germany.
Journal of Nutrition, Health & Aging, 14 (8): 669–676. Psychiatrische Praxis, 34: 134–138.
Dementia care in the community: Challenges for primary health and social care 171

Stokes, G. (2000). Challenging Behaviour in Dementia: Waldorff, F.B., Bülow, L.B., Malterud, K. and Waldemar,
A Person Centred Approach. Bicester: Winslow Press. G. (2001). Management of dementia in primary health
Sullivan, K. and O’Connor, F. (2001). Should a diagnosis care: The experiences of collaboration between the
of dementia be disclosed? Aging & Mental Health, 5: GP and the district nurse. Family Practice, 18 (5):
340–348. 549–552.
Thomas, P., Ingrand, P., Lalloue, F. et al. (2004). Reasons Wilcock, J., Iliffe, S., Griffin, M. et al. (2013). Tailored
of informal caregivers for institutionalizing dementia educational intervention for primary care to improve
patients previously living at home: The Pixel study. the management of dementia: The EVIDEM-ED cluster
International Journal of Geriatric Psychiatry, 19 (2): randomised controlled trial. Trials, 14: 397.
127–135. World Health Organization (2003). Global burden of
Vernooij-Dassen, M.J., Moniz-Cook, E.D., Woods, R.T. dementia in the year 2000. Available at www.who.int/
et al. (2005). Factors affecting timely recognition and entity/healthinfo/statistics/bod_dementia.pdf.
diagnosis of dementia across Europe: From awareness Xie, J., Brayne, C., Matthews, F.E., Medical Research
to stigma. International Journal of Geriatric Psychiatry, Council Cognitive Function and Ageing Study
20 (4): 377–386. collaborators (2008). Survival times in people with
Vickrey, B.G., Mittman, B.S. and Connor, K.I. (2006). The dementia: Analysis from population based cohort
effect of a disease management intervention on quality study with 14 year follow-up. British Medical Journal,
and outcomes of dementia care – A randomized, con- 336: 258-262.
trolled trial. Annals of Internal Medicine, 145 (10): 713–726. Zermansky, A., Alldred, D.P., Petty, D.R. and Raynor,
Waldemar, G., Phung, K., Burns, A. et al. (2007). Access D.K. (2007). Striving to recruit: The difficulties of
to diagnostic evaluation and medical treatment for conducting clinical research on elderly care home
dementia in Europe. International Journal of Geriatric residents. Journal of the Royal Society of Medicine,
Psychiatry, 22 (1): 47–54. 100: 258–261.
16
Managing people with dementia in the
general hospital

ANDREW TEODORCZUK, ROWAN HARWOOD AND ELIZABETH SAMPSON

reasons, such as sensory impairment, learning disability, or


16.1 INTRODUCTION approaching the end of life (Inouye et al., 1999).
In this chapter, we explore the challenges of caring for
In the United Kingdom, older people with dementia occupy someone with dementia in the general hospital. Initially, the
a quarter of the general hospital beds (Alzheimer’s Society, epidemiology is detailed and issues of screening discussed.
2009). One out of three emergency medical admissions is The diverse clinical presentations and problems of physi-
of an older person with cognitive impairment; someone cal health, complexities and uncertainties are outlined.
with dementia is up to four times more likely to be admit- The chapter then considers specific areas of concern such
ted as an emergency case than someone without (Goldberg as pain suffered by the patient with dementia that may be
et al., 2012) and severity of dementia independently predicts overlooked. Key transition points, such as admission and
hospitalization (Albert et al., 1999). Arguably, dementia discharge of patients are further described and the chapter
now represents ‘core business’ for general hospital clinical ends with potential solutions to the difficulties that patients
practices. with dementia face in hospital, together with evidence-
The largest hospital medical speciality is geriatric medi- based educational messages for hospital staff.
cine, which espouse ‘comprehensive geriatric assessment’,
taking into account function, mental health, social and
environmental aspects alongside diagnosis and medical
management. Yet, current systems of care often presume
16.2 EPIDEMIOLOGY OF PEOPLE IN
non-confused patients with a single acute illness, some- HOSPITAL WITH DEMENTIA
times called the ‘acute medical model’ (Tullo et al., 2015).
This mismatch between hospital practices and the current It has been estimated that 25% of hospital inpatients in the
hospital demographic presents challenges to patients, fam- United Kingdom have dementia, but identifying those most
ily or other carers and staff alike. Typically, staff have not at risk can be challenging. Under-diagnosis of dementia in
been trained to work with patients suffering from dementia the community, particularly in primary care, is common
and find it difficult to implement good dementia care within and therefore, diagnosis often occurs at a time of crisis, for
outdated work structures and expectations (Marshall, 1999, example, after a fall, when the person comes into contact
2001; Clissett et al., 2014). with healthcare or social care services, and requires an
Patients with dementia are especially vulnerable in emergency hospital admission (Bourne, 2007).
acute hospital settings. Admission to hospital represents a However, even after general hospital admission, detec-
time of crisis usually caused by acute ill health or injury. In tion rates remain as low as between 37% and 46% (Harwood
addition to this, the setting forms a disruption to familiar- et al., 1997; Joray et al., 2004; Sampson et al., 2009). Diagnosis
ity and routine, which a person with dementia relies on to of dementia in the general hospital is made more complex by
enable them to cope successfully with daily activities. The concurrent delirium (see Chapter 71), the fact that patients
challenge, however, of improving dementia care also repre- may be sleep deprived and unwell in a strange and disori-
sents an opportunity. The elements of good dementia care entating environment and may have suffered an acute func-
enable good care for those who are vulnerable for other tional decline. Assessing cognition in these circumstances

172
Managing people with dementia in the general hospital 173

can be impossible. Delirium may occur in the absence of prior such as the Mini-mental State Examination (MMSE) (Folstein
dementia, but up to two-thirds of people with dementia in et al., 1975) and its use already is embedded in many hospitals.
hospital have superimposed delirium, recovery from which For both clinical and research purposes, a two-step approach is
may be slow or incomplete (Cole et al., 2009; Mukadam and essential with initial screening and then more detailed cogni-
Sampson, 2011; Whittamore et al., 2014). tive, functional and behavioural assessment is necessary. This
Screening for dementia in the community is controversial. should include discussion with a carer or relative to acquire
It has been argued that primary care doctors may not have the collateral history and then applying diagnostic criteria, such
specialist skills to make an accurate diagnosis and that offering as Diagnostic and Statistical Manual of Mental Disorders,
financial incentives to do this is unethical (Campbell-Taylor, Fourth Edition (DSM-IV) or International Classification of
2014). However, routine assessment of cognition is necessary Diseases, 10th Revision (ICD-10), or clinician’s judgement.
for inpatients in general hospital: delirium (and underlying The frailty of patients and age of people admitted to gen-
medical causes) must be diagnosed and those with previously eral hospitals reflects and then amplifies the prevalence of
known and unknown dementia identified in order to adjust to dementia found in the community. Prevalence estimates
meet their special needs (Goldberg et al., 2012). This includes for severe dementia in medical inpatients, over the age of
adapting communication and care approaches to enhance 65 years range from 9% to 50% in general hospital wards
understanding and minimize distress, communication and (Feldman et al., 1987; Hickey et al., 1997) and from 63% to
engagement with family carers and other stakeholders, collec- 80% in specialized geriatric medical wards (Adamis et al.,
tion of medical and personal background information from 2006; Torian et al., 1992). This illustrates how the preva-
informants to enable decision making for patients lacking lence of dementia is highest in older women and unlike
mental capacity and to plan discharge and future care. Many other criteria such as, ICD-10, the DSM-IV criteria does not
screening tools are available, but only the simple 10-point require 6 months duration of symptoms for diagnosis. Half
Abbreviated Mental Test Score (AMTS) has been validated in of the patients with hip fractures have cognitive impair-
more than one study in general hospitals (Jackson et al., 2013). ment (Holmes and House, 2000). For a summary of the
This tool is shorter than alternative cognitive screening tests prevalence of dementia in general hospitals, see Table 16.1.

Table 16.1 Prevalence of dementia in older general hospital patients – clinical studies 2000–2014
Age (mean, Female Sample Assessment tools and Prevalence
Paper Setting SD) % size diagnostic criteria %

Mixed wards
Uwakwe (2000) Medical, surgical, >60 (69.9, 38.7 109 24 item Self Reporting 2.8
Nigeria gynaecological 8.4) Questionnaire (SRQ-24),
wards Geriatric Mental State
(GMS), MMSE, ICD-10
Wancata et al. (2003) Medical wards, >60 (75.9, 72.3 372 Clinical Interview Schedule 27.4
Austria general 8.4) (CIS), DSM-IIIR and at
hospital least 2/5 on global
severity scale
Travers et al. (2013) Medical, surgical, >70 (80.4, 58.4 493 MMSE 20.7
Australia orthopaedic 6.5) Informant Questionnaire of
wards Cognitive Decline
(IQCODE)
DSM-IV

Geriatric wards/hospital
Laurila et al. (2004) Acute geriatric >70 (not 71.7 219 MMSE, Digit Span, parts of 40.2
Finland wards (medical given) WAIS, operationalized
and surgical), 59.4% DSM-IV criteria
geriatric over 85
hospitals years
Marengoni et al. Geriatric wards >65 (79.4, 54.1 1221 DSM-IV 9.6
(2011) n/a)
Italy
Zekry et al. (2008) Geriatric hospital >75 (85.2, 76.0 349 MMSE, Short Cognitive 43.3
Switzerland 6.7) Evaluation, DSM-IVTR
(Continued )
174 Dementia

Table 16.1 (Continued) Prevalence of dementia in older general hospital patients – clinical studies 2000–2014
Age (mean, Female Sample Assessment tools and Prevalence
Paper Setting SD) % size diagnostic criteria %
Acute medical wards
Margiotta et al. Acute medical >65 (79.8, 58 330 MMSE, DSM-IV 26.1
(2006) unit 8.0)
Italy
Sampson et al. (2009) Acute medical >70 (83.0, 59 617 MMSE, DSM-IV 42.0
United Kingdom wards 7.3)
Specific populations
Frohnhofen et al. Inpatients with Male 79, 8 63 1621 MMSE 52
(2011) COPD Female 81, 8 Global Deterioration
Germany scale (GDS)
Nightingale et al. Orthopaedic >65 n/a 731 GMS/AGECAT 40
(2001) Hip fracture (n/a) MMSE
United Kingdom
Gruber-Baldini et al. Orthopaedic >65 77.5 674 MMSE 28
(2003) Hip fracture (81, n/a) Chart review
United States
Abbreviations: SD, standard deviation; MMSE, Mini-mental State Examination; ICD-10, International Classification of Diseases, 10th
Revision; DSM-IV, Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition; WAIS, Wechsler Adult Intelligence Scale;
COPD, chronic obstructive pulmonary disease; n/a, not available.

Epidemiological studies vary in their findings, depending stroke and hip fracture all increase exponentially with age.
on the populations studied and the diagnostic tools used to Arthritis, heart, lung and bladder problems, Parkinson’s
study the population. disease, poor vision and deafness are also common at this
stage. Functional problems (disability) represent a final
common pathway for the effects of these multiple diagno-
16.3 CHARACTERISTICS OF PEOPLE sis and the impact of the environment, aids and available
WITH DEMENTIA IN HOSPITAL human help. People with dementia are prone to acute ill-
ness, injury, adverse drug effects and delirium (Box 16.1).
People with dementia in general hospitals are older by 5–9 The experience and well-being of a person with dementia
years (Erkinjuntti et al., 1986; Lyketsos et al., 2000), more is determined by symptoms more than their neurological
likely to be female, with lower education level (Sands et al., impairments. Personality, biography, other aspects of health
2003; Joray et al., 2004), more impaired in activities of daily (physical and mental) and social environment are impor-
living (Torian et al., 1992; Zekry et al., 2008) and more tant too (Kitwood, 1997). General physical health may be
likely to be admitted from nursing homes (Zekry et al., more amenable to intervention than other aspects, but may
2008; Sampson et al., 2009). Among acute medical inpa- be neglected if the dementia diagnosis is overemphasized
tients, dementia has a marked and independent effect on (so-called ‘diagnostic overshadowing’).
short- and longer-term mortality. Dementia significantly The term ‘frail’ means the propensity to deterioration in
increases the risk of death during the index admission even the face of a stressor, reflecting loss of physiological homeo-
after controlling severity of present illness considering the stasis, functional reserve and social resilience. Following
age and multimorbidities. This increased risk is found both this definition many people with dementia are ‘frail’ and
in medical inpatients and in patients who have suffered hip warrant proactive attempts to maintain health and pre-
fracture (Sampson et al., 2009; Marengoni et al., 2011). vent illness. Examples include vaccination, vascular risk
and bone protection, careful drug review and physical and
social activity promotion.
16.4 PHYSICAL HEALTH IN DEMENTIA Ultimately ill health will lead to death, more than half
of which occur in hospitals. This should be anticipated and
Multiple pathology is a defining feature of ill health in old planned for, in terms of preferred place of care and some indi-
age. People with dementia often have other things wrong as cation of preferences, beliefs and values with respect to health-
well and most of the patients are old. Moreover, dementia, care interventions (such as tube feeding and resuscitation). As
Managing people with dementia in the general hospital 175

breathlessness and incontinence. In dementia, the crisis


BOX 16.1: Older people differ medically may also be due to a sudden worsening of confusion state,
from the young or ‘behavioural’ problems, such as, wandering and getting
lost, aggression towards carers, or a grave misjudgement
Presentation is often non-specific or atypical. The of abilities. Among older patients admitted into hospitals,
classical non-specific presentations are confusion, delusions, hallucinations, agitation and aggression, each
immobility, falls and incontinence, or ‘inability to of these symptoms have 10%–20% prevalence. A third of
cope’ the patients are anxious or depressed, apathetic or awake
Diseases are multiple at night (Goldberg et al., 2012). Propensity to delirium is a
Lose abilities fast – so diseases can present with key issue; this must be recognized, diagnosed, distressing
their functional consequences features addressed and opportunity allowed for rehabilita-
More prone to complications of disease and their tion and recovery.
treatments Sometimes the presentation is due to the progression of
More dependent on the environment – someone dementia. This is especially the case with vascular dementia,
with an adverse environment will present where the steps of progression can look like delirium and be
symptoms sooner, someone with a supportive associated with problems, like, loss of balance or swallow-
environment often later and presentation of ing difficulties, or dementia with Lewy bodies where motor
symptoms may be due to a change in the features, fluctuation, psychosis and drug sensitivity may be
environment rather than the person concerned responsible.
Need for explicit rehabilitation

16.6 COMPREHENSIVE GERIATRIC
the disease progresses, it is appropriate to de-prioritize pre-
ventative drugs and to emphasize comfort and dignity.
ASSESSMENT

Health problems may present itself with typical symptoms,


such as, pain or breathlessness, but also in terms of their
16.5 EMERGENCY ADMISSION TO impact on everyday functions, such as, immobility, falls or
HOSPITAL incontinence. The ‘Comprehensive Geriatric Assessment’
(Box 16.2) tries to understand these problems, at the level
One of the most challenging times for people with demen- of body parts or organs (so-called ‘impairments’, such as
tia is dealing with acute illness, injury or other crises. Not weakness, dizziness, visual acuity or bladder function) to
all crises are medical and not all medical crises need hospi- identify underlying pathological diagnoses or syndromes.
tal admission. The exact causes of crises in old age can be From these specific treatments – pharmacological, surgi-
difficult to disentangle. Medical, mental and social factors cal, or rehabilitation – can be applied. Psychological, social
may all be operating and their relative contributions may be and environmental contextual factors are also taken into
unclear. For example, the cause of hallucinations or delu- account and may themselves be the target of therapeu-
sions may be delirium, physical illness, or the ability to cope tic, adaptive or supportive interventions, for example, by
with a chronic medical condition may depend on the physi- informing and educating family carers, or by resorting to
cal and social environment. environmental aids and adaptations.
People come to hospital because of a crisis that is, or may Medical care for people with dementia draws on a mul-
be, due to physical illness or injury, and where investigation, tiplicity of models (Box 16.3), often simultaneously, to
treatment or nursing care requirements cannot be met in address the complex mix of multiple acute, progressive and
another setting. In the face of such crises, general hospitals chronic problems, which impact function and well-being,
are called upon to diagnose or exclude physical disease, to
understand and take account of mental and social factors
and to provide refuge and relative safety while the crisis is BOX 16.2: Dimensions of Comprehensive
resolved. Hospital beds are an expensive and scarce resource, Geriatric Assessment
so must be used wisely; hospital stay should only continue if
necessary care cannot be delivered elsewhere. Hospitals must Diagnoses
integrate pathways of care with community or at-home reha- Function
bilitation provisions, primary care and community health Mental
teams (including mental health) and care homes. Social
Crises that cause admission in hospitals are frequently Environmental
functional, including falls, immobility, pain, dehydration,
176 Dementia

BOX 16.3: Models of health care for older people


Prevention Anticipatory intervention among high risk groups
Medical Diagnose, treatment
Rehabilitation Cycle of problem identification, analysis, multi-professional therapeutic intervention and
reassessment, directed at maximizing physical and social functioning
Palliative Meticulous management of symptoms, open communication, attention to psychological, social
and spiritual aspects
Person-centred care Value and respect for personhood, individualized care, empathic understanding,
communication and relationships to promote well-being and reduce distress
Recovery model Emphasize hope, set achievable goals, identify positive attributes and abilities, take risk, accept
failure
Social model Opportunities, environment and relationships adapted to personal abilities

in a varied and sometimes adverse social and physical envi- different ways, through behaviours. When in pain, a person
ronment (e.g. frail or stressed carers, residence in a care with dementia may respond with agitation, often shouting,
home, or an unadapted domestic environment). restiveness to care, depression or withdrawal (Ryden et al.,
1999; Tosato et al., 2012). This association is, however, com-
plex. Some observational pain scales are sensitive in detect-
ing pain in people with dementia but have a high rate of false
16.7 ELECTIVE ADMISSION positive alarm rate and may be detecting fear, frustration or
anger (Jordan et al., 2011). In the general hospitals, other
People with dementia may also be admitted to hospital factors such as delirium and the confusing ward environ-
for elective procedures. Special care is required to prevent ment may increase the risk of behavioural and psychologi-
delirium, detect and treat it if it occurs. Attention must be cal symptoms and these may also be communicating other
paid while dealing with the unavoidable disruption to rou- unmet needs such as fear and boredom (Ahn and Horgas,
tine, to ensure good communication with family or other 2013).
stakeholders, assessment of mental capacity and ability to The ‘gold standard’ for pain assessment is self-report.
consent provision of appropriately skilled rehabilitation. Some people with moderate to severe dementia can report
Problems may arise if previously undiagnosed dementia pain if supported by using visual pain scales (Zwakhalen
is present; assessment prior to admission in the hospital et al., 2006). One study in a geriatric hospital found out
should specifically assess cognition. that only 44% of patients with severe dementia were able to
report that they were in pain (Pautex et al., 2005). Clinicians
assessing behaviours that might indicate pain need to look
purposefully for potential sources of pain, in particular
16.8 PAIN joints, pressure areas, the mouth, abdomen and pelvis. It is
helpful to observe people with communication difficulties
Persistent pain is found in up to half of people with demen- over a period of time, in motion, including transfers from
tia in the community or in care homes, but is commonly bed to chair or repositioning and at rest, but people differ
under-detected and under-treated. It has been reported that greatly in how they express pain and hospital staff may not
some clinical staff believe that people with dementia actually be aware of an individual’s pain indicators. Pain manage-
experience less pain (Scherder et al., 2009). Under-treatment ment can be improved by contacting relatives or carers of
of pain causes distress and suffering for people with demen- the person with dementia who may be aware of individual
tia, their families included. In addition, pain increases the responses to pain (for example, social withdrawal or gri-
risk of delirium (Inouye, 1993), slows recovery, worsens macing). Few pain scales for people with dementia have
functional decline and is associated with poor sleep and been validated in the general hospital, despite the fact that
appetite (Scherder et al., 2009). Poorly managed pain can this is a frail older population likely to suffer from painful
slow down a person’s rehabilitation or even increase the risk multiple morbidities.
of delirium. These are major drivers of prolonged hospital In care homes, behavioural problems, particularly, verbal
stay and thus, raise the costs of care (Zwakhalen et al., 2006). aggression can be significantly reduced with systematic pain
Under-detection of pain may occur because people with management (Husebo et al., 2011). There have been no stud-
dementia may not remember they have been in pain before. ies conducted of such interventions in the general hospitals.
Some can tell you about pain if asked, but others lose the However, a number of barriers to improving pain management
words required to describe it. Pain can be communicated in in hospitals have been identified, including poor knowledge
Managing people with dementia in the general hospital 177

of how to respond when pain is detected, irregular adminis- Considerable uncertainty often surrounds the likely
tration of ‘as required’ medications and concerns about the success of a discharge. Attitude to risk vary greatly, among
side effects of some medications, particularly, opiates. Simple patients (when suffering from dementia or when they had
interventions with regular analgesia such as paracetamol, can mental capacity), families, care homes and hospital staff.
be extremely effective (Zwakhalen et al., 2006). Risk is often unavoidable and important functions are
to identify it, mitigate it where possible and then follow a
decision-making process to decide whether to take it up or
not. There is little evidence that discharge to a care home is
16.9 DISCHARGING PATIENTS WITH
‘safer’. A ‘trial of discharge’ to a person’s own home is often
DEMENTIA warranted, sometimes as part of a process of establish-
ing that institutional care is truly required (Stewart et al.,
Discharging a person with dementia from hospital requires 2005). The process is sometimes called ‘risk enablement’
planning and good communication. The person may be (Manthorpe and Moriaty, 2010).
able to cope well, or have well-established and satisfactory There is relatively little evidence on successful discharge
support arrangements, but equally well progression of dis- specific to patients with dementia. This reflects on the fact
ease; the consequences of acute illness, slow recovery, or that there are limited available reliable data, especially in
inadequate previous care support mean that explicit new the United Kingdom. However, the evidence suggests that
provision is required. The usual goal will be to return the the greatest variation in discharge processes and outcomes
person to the accommodation from which they were admit- is in older patients (Martin and Smith, 1996).
ted. About a quarter of people with dementia admitted in Studies have shown that patients with dementia have a
general hospitals are residents of care homes (Goldberg longer length of stay in hospitals, more readmissions and
et al., 2012). About a quarter will not return home, 10% die greater use of medical services compared to those without,
in hospital and a quarter will be newly placed in a care home even after controlling illness severity and functional impair-
within the next 6 months of their discharge. Readmissions ment (Holmes and House, 2000; Saravay et al., 2004; King
are common (18% within 30 days and nearly half within et al., 2006). Patients with dementia in the United Kingdom
6 months) (Bradshaw et al., 2013; Jencks et al., 2009). People typically stay for about 11 days in hospitals, 5–7 days longer
with dementia (and delirium) may be denied opportuni- than those without dementia (Alzheimer’s Society, 2009).
ties to recover and regain function in intermediate care and This conceals considerable variability: 25% are discharged
other rehabilitation centres. within 5 days, but 10% stay longer than 30 days (Goldberg
The goal of a safe discharge is to relocate a patient to et al., 2013).
accommodation in accordance with their preferences and Potential reasons for increased length of stay include
wishes that can meet their care needs and those of fam- greater functional impairment, slow or uncertain recov-
ily carers as well as other stakeholders. Efficient services ery, non-compliance with tests, treatment and therapy and
aim to minimize length of stay and readmissions in hos- carer strain. The need for explicit decision-making process
pital and use of expensive resources such as care homes, for those lacking mental capacity, concern about taking risk
while accepting that these are sometimes the right option on behalf of vulnerable adults, funding problems and the
for individuals. Studies seeking to understand what leads availability of suitably skilled community services and care
to a safe discharge suggest that it is dependent on patient homes can further delay discharge.
and hospital characteristics, social environment and Patients with dementia have a greater risk of readmis-
clinical practice (Westert et al., 1993; Martin and Smith, sion (Daiello et al., 2014). A United States retrospective
1996). Important patient characteristics include age, care- cohort study found that patients with dementia had greater
giver burden, comorbidity and polypharmacy. Social fac- odds ratio of readmission (1.8) compared to patients with-
tors that affect patients include availability of social care, out dementia. Higher readmission rates were further asso-
levels of poverty, provision of nursing homes and family ciated with antipsychotic use and discharge from nursing
support. homes. Potential reasons why patients with dementia may
Tailored discharge planning does reduce length of stay have greater readmission rates include factors such as they
and readmission rates (Shepperd et al., 2010), but the extent are less likely to benefit from discharge education, adhere
to which discharge planning impacts on other health out- to instructions and report symptoms (Becker et al., 2010).
comes including mortality is unclear. Important risks for Furthermore, patients with dementia are more likely to be
readmission include demographic factors such as, low admitted with ambulatory care conditions than those with-
socio-economic status, health literacy, reduced social net- out dementia (Phelan et al., 2012) and hence, readmission
work. Clinical factors that increased risk of readmission rates are greater.
include discharges against medical advice; polypharmacy However, it is not just patient factors that increase vul-
(over five medications) and having more than six chronic nerability to discharge and failed transitions of care. A
conditions. Specific conditions such as, chronic obstructive study into the practice gaps (difference between desired and
pulmonary disease, diabetes, heart failure, stroke, cancer actual practice) of hospital staff found that discharge was
and depression all increase risk of readmission. identified as a technical gap, however dependent on and
178 Dementia

preceded by attitudes of staff towards patients with demen- must be personal taking account of individual factors
tia and ownership of this complex group (Teodorczuk et al., including time of discharge and decision-making
2013). Hence, the socio-cultural ward environment and capacity. Educating patients and carers and sharing
degree to which staff are able to effectively own and manage information such as, a fact sheet on dementia, can help
patients with dementia within the ward environment are an further understanding the patient such as the Age UK
important determinant of discharge success (Teodorczuk Factsheet (Age UK, 2013).
et al., 2015). Personalizing a discharge plan involves ‘joining up’
However, a recent well conducted trial of routine geri- knowledge about the patient to promote and harness
atric care versus joint mental health medical care failed to collaborative practice. Key professionals involved in
show an improvement on the length of stay or readmission, the planning process include the pharmacist (concor-
though, those patients on the specialist units had greater dance and dispensing in the community), occupa-
satisfaction (Goldberg et al., 2013). Various reasons might tional therapy (to assess safety of home environment
support this finding, including the fact that frailty among risks and functionality), medical and nursing staff (to
dementia patients might be the driving factor behind the coordinate, follow up and proceed with treatment),
length of stay and readmission rate. Arguably, there is physiotherapy (mobility needs and fall risks) and
a need to look beyond these ‘harder’ outcomes towards social workers (to address funding issues, instigate
more relevant outcomes of patient and carer experience. carer support and reduce risks where present [e.g. door
Alternatively, it could have been the socio-cultural environ- alarms if risk of wandering]). At all points carers must
ment in the standard care unit that may have been optimal be part of the process.
and therefore, less appropriate as a comparator. Either way, 3. Assessing capacity at the right time
what is clear is that, in terms of understanding discharge Identifying whether a patient has capacity to
factors for patients with dementia it is a multifactorial com- decide on residence destination has been shown to
plex construct and there is a need for further work studying be a critical event. This is especially pertinent for
socio-cultural environments and exploring factors other patients with cognitive impairment who may lack
than such traditional measures as length of stay, as markers capacity, though, this is not always clear-cut. In
of good quality care. particular, it is essential to ensure that it is under-
taken at the right time, by the right person assessing
the right decision. Furthermore, staff undertaking
capacity assessment should have the right skills to do
16.10 IMPLICATIONS FOR CLINICAL it. In an ethnographic study (Poole et al., 2014) out of
PRACTICE 13 people deemed to have capacity nine went home
whereas, out of 16 deemed not to have capacity, only
Four general principles can guide, though not guarantee a one went home.
successful discharge and help address key factors important It is also important to ensure that, where
to effectiveness of safe discharge processes. They are listed patients have borderline capacity the right deci-
as follows sion is made and the patient has the best chance of
demonstrating capacity, if transfer to a step down
1. Considering discharge as a process and not an end point unit may be necessary. In some settings, if there is
Planning for discharge should begin at the begin- provision, a cognitive rehabilitation unit staff can
ning of hospitalization. The process can be broken be helpful, both physical and mental health staff can
down into pre-discharge interventions (e.g. medica- be effective.
tion reviews and discharge planning reviews), bridging 4. Ensuring a realistic contingency plan
interventions and post-discharge interventions such Where the risk of a failed discharge is high, a clear
as, a post-discharge phone call. Successful approaches and realistic contingency plan that both family and
for patients with dementia such as those piloted staff are aware of is crucial. If a patient is discharged
through the Royal Free My Discharge project in the to the community this should include identification
United Kingdom include allocating a care profes- of risks common to patients with dementia such as
sional (e.g. an occupational therapist) with a specific wandering, driving, operating gas cookers and medi-
remit to facilitate discharge (Royal Free London NHS cation management included within the discharge
Foundation Trust, 2014). Furthermore, bridging plan. Conversely, if a patient is discharged to a care
partnerships with other organizations in the voluntary home, education of staff to understand behaviours
sector and with community services are essential to and enhance communication is essential. A formu-
help plan a supported discharge. lation including antecedents and consequences of
2. Personalize the discharge plan behaviour as well as understanding behaviours in the
Arguably, the focus of hospital processes can be context of a patient’s background and medical ill-
on treating diseases rather than the patient. However, ness may help facilitate this process and increase the
more so for patients with dementia, discharge plans probability of a safe discharge.
Managing people with dementia in the general hospital 179

understanding of how frightened the patient is in hospi-


16.11 CHALLENGES AND SOLUTIONS tal, (4) promoting carer partnerships, (5) fostering person-
centred care, (6) understanding how to communicate with
The acute hospital is challenging for a person with demen- patients suffering from dementia, (7) recognition of cogni-
tia and disruptive to familiarity and routine that people tive impairment and (8) specific clinical needs (e.g. capacity
with dementia rely upon. Hospitals are busy, noisy, active assessments and discharge planning).
places. This can be over-stimulating and overwhelming for As demonstrated in Figure 16.1 these practice gaps or
those who are disorientated or with impaired reasoning or learning needs are hierarchically organized at a theoreti-
processing ability. The assessment model, in all disciplines, cal level, addressing the core level learning needs (e.g. of
is based on repeated and intensive questioning, by many attitudes and ownership) is essential in order to succeed
different people. Locations change rapidly (emergency with the more technical learning needs such as, discharge
department, admissions unit, ward, X-ray department, of patients. Furthermore, it is possible that the core level
physiotherapy gym), staff are busy and often stressed and learning needs may be related by means of a vicious circle.
nights can be very disturbing for both staff and patients. Staff may fail to gain confidence of a patient on the ward
In the United Kingdom, the majority (but not all) of and therefore, develop negative attitudes that can lead to a
people with dementia in hospitals are looked after by spe- patient feeling frightened and behaving in a manner that
cialist geriatric medical services. Consultant geriatricians is ‘challenging’, thereby further reducing the ownership of
have a broad training in medical sub-specialties and should such a patient in the ward.
be expert in clinical therapeutics, rehabilitation and pal- Evidence suggests that, if educational programmes are
liative care. The central role of multidisciplinary working, designed specifically to target these learning needs, with a
family engagement and consideration of mental health and clear focus on the core level needs and breaking the vicious
social factors and the importance of links with commu- circle, practice can be influenced effectively (Teodorczuk
nity rehabilitation, care homes and old age psychiatry are et al., 2014). To train staff successfully an interprofessional
acknowledged. Physiotherapists, occupational, speech and approach focussed on both formal and informal learning is
language therapists and nurses specializing in older peo- advocated and involving patients and carers in the teaching
ple bring eclectic approaches to addressing functional and processes can address issues like negative attitudes. In addi-
practical problems. Outreach and liaison services may exist, tion to this, it is clear that organizational processes should
especially for orthogeriatrics and admission units. Recently, be adapted to help the systems learn about the patient and
in the United Kingdom, there has been a great expansion staff to implement good dementia care practices. Successful
in older people’s liaison mental health services. This has organizational approaches underpinned by contempo-
been driven by evaluation of ‘RAID’ (Rapid, Assessment, rary educational theory have been reported in a study by
Interface and Discharge) models of care, where properly Teodorczuk et al. (2015). Approaches typically focus on
commissioned and funded psychiatric teams work in gen- encouraging learning about the patient not only at an indi-
eral hospitals. These services decrease costs and length of vidual level but equally at team and system levels. These
stay in the hospital and improve patient satisfaction with
care (Tadros et al., 2013). These teams work alongside gen-
eral hospital staff to provide assessment, advice, discuss
with families, plan and follow up, also establishes links
Ide d sp
bli

between inpatient and community mental health services


nti ots
n

and staff coaching or training.


fie
d

Specific
clinical needs
Par

16.12 LEARNING NEEDS OF HOSPITAL


tne

STAFF IN RELATION TO
rsh

Common clinical needs


ip

DEMENTIA CARE
con

Recognition, communication
cep

and person-centred care


t

The importance of educating staff has been alluded to


throughout the chapter. It is clear that patients with demen-
Pat

tia present significant challenges to staff. To overcome these


ien

Core non-clinical needs


challenges, training is essential, though not sufficient as a
t’s
fea

Ownership and negative attitudes


stand alone. In terms of the content of the training process,
r

the important areas to cover have been identified in an in


depth grounded theory study of staff practice gaps in rela-
tion to managing the confused older patients (Teodorczuk Figure 16.1 Hierarchy of practice caps including hid-
et al., 2013). These include (1) ownership of the confused den element. (From Teodorczuk, A. et al., International
patient, (2) addressing negative attitude, (3) developing an Psychogeriatrics, 25, 645–655, 2013.)
180 Dementia

include relaxing carer visiting times, reducing ward moves Bourne, J. (2007). Improving Services and Support for
and targeting senior managers to learn about hospital expe- People with Dementia. London, United Kingdom:
riences of dementia and delirium. Ultimately, if systems are National Audit Office.
adapted around the new epidemiology and demography of Bradshaw, L.E., Goldberg, S.E., Lewis, S.A. et al. (2013).
hospital inpatients, then it becomes easier for staff to oper- Six-month outcomes following an emergency hospi-
ate, own and manage patients with dementia. tal admission for older adults with co-morbid mental
health problems indicate complexity of care needs.
Age and Ageing, 42 (5): 582–588.
Campbell-Taylor, I. (2014). Increased screening for dementia:
16.13 CONCLUSIONS A potentially dangerous proposal. BMJ, 349: g6869.
Clissett, P., Porock, D., Harwood, R.H. and Gladman, J.R.
(2014). The responses of healthcare professionals to
As the population ages a period of epidemiological tran-
the admission of people with cognitive impairment to
sition follows. Morbidity of old age, mental and physical
acute hospital settings: An observational and interview
comorbidity of chronic diseases are now a norm in the
study. Journal of Clinical Nursing, 23: 1820–1829.
general hospitals. The systems and structures are inevita-
Cole, M.G., Ciampi, A., Belzile, E. and Zhong, L. (2009).
bly challenged as a result, increasingly burdening hospi-
Persistent delirium in older hospital patients: A sys-
tal staff managing a different demography and ultimately
tematic review of frequency and prognosis. Age and
having a negative impact on patients and carers. In this
Ageing, 38(1): 19–26.
context, this chapter aims to upskill and draw aware-
Daiello, L.A., Gardner, R., Epstein-Lubow, G. et al. (2014).
ness towards these challenges in relation to the patient
Association of dementia with early rehospitaliza-
admitted with dementia. Important areas such as pain
tion among Medicare beneficiaries. Archives of
and physical illness in patients suffering from dementia
Gerontology and Geriatrics, 59: 162–168.
as well as key educational messages for staff, are out-
Erkinjuntti, T., Wikstrom, J., Palo, J. and Autio, L. (1986).
lined. However, teaching and inculcating staff knowl-
Dementia among medical inpatients. Evaluation of
edge alone will fail unless followed by practice behaviour
2000 consecutive admissions. Archives of Internal
training. Perhaps the challenge for professionals working
Medicine, 146: 1923–1926.
with dementia patients is to move beyond the traditional
Feldman, E., Mayou, R., Hawton, K. et al. (1987).
­physical-mental health divide and adopt a holistic collab-
Psychiatric disorder in medical in-patients. The
orative approach towards practice that is relevant to man-
Quarterly Journal of Medicine, 63: 405–412.
aging the increasing complexity of dementia patients in
Folstein, M.F., Folstein, S.E. and McHugh, P.R. (1975).
the general hospitals.
“Mini-mental state”. A practical method for grading
the cognitive state of patients for the clinician. Journal
REFERENCES of Psychiatric Research, 12 (3): 189–198.
Frohnhofen, H., Heuer, H.C., Willschrei, H.P. and
Adamis, D., Treloar, A., Martin, F.C. and Macdonald, A.J. Falkenhahn, C. (2011). Dementia among older
(2006). Recovery and outcome of delirium in elderly in-­hospital patients with obstructive airway dis-
medical inpatients. Archives of Gerontology and ease: Frequency and consequences. Zeitschrift für
Geriatrics, 43 (2): 289–298. Gerontologie und Geriatrie, 44: 240–244.
Age UK (2013).Hospital discharge arrangements. Available Goldberg, S.E., Bradshaw, L.E., Kearney, F.C. et al. (2013).
at http://www.ageuk.org.uk/Documents/EN-GB Care in specialist medical and mental health unit
/Factsheets/FS37_Hospital_discharge_arrangements compared with standard care for older people with
_fcs.pdf?dtrk=true. Accessed 12 June 2016. cognitive impairment admitted to general hospital:
Ahn, H. and Horgas, A. 2013. The relationship between Randomised controlled trial (NIHR TEAM trial). BMJ,
pain and disruptive behaviors in nursing home resident 347: f4132.
with dementia. BMC Geriatr. 13: 14. Goldberg, S.E., Whittamore, K.H., Harwood, R.H. et al.
Albert, S.M., Costa, R., Merchant, C. et al. (1999). (2012). The prevalence of mental health problems
Hospitalization and Alzheimer’s disease: Results from a among older adults admitted as an emergency to a
community-based study. The Journal of Gerontology. general hospital. Age and Ageing, 41 (1): 80–86.
Series A, Biological Science and Medical Sciences, 54: Gruber-Baldini, A.L., Zimmerman, S., Morrison, R.S. et al.
M267–M271. (2003). Cognitive impairment in hip fracture patients:
Alzheimer’s Society (2009) Counting the Cost. London, Timing of detection and longitudinal follow-up.
United Kingdom: Alzheimer’s Society. Journal of American Geriatrics Society, 51: 1227–1236.
Becker, M.A., Boaz, T.L., Andel, R. et al. (2010). Predictors Harwood, D.M., Hope, T. and Jacoby, R. (1997).
of preventable nursing home hospitalizations: The Cognitive impairment in medical inpatients: Do phy-
role of mental disorders and dementia. The Americal sicians miss cognitive impairment? Age and Ageing,
Journal of Geriatric Psychiatry, 18: 475–482. 26 (1): 37–39.
Managing people with dementia in the general hospital 181

Hickey, A., Clinch, D. and Groarke, E.P. (1997). Prevalence Margiotta, A., Bianchetti, A., Ranieri, P. and Trabucchi, M.
of cognitive impairment in the hospitalized elderly. (2006). Clinical characteristics and risk factors of delir-
International Journal of Geriatric Psychiatry, 12: 27–33. ium in demented and not demented elderly medical
Holmes, J.D. and House, A.O. (2000). Psychiatric illness in inpatients. The Journal of Nutrition Health and Aging,
hip fracture. Age and Ageing, 29 (6): 537–546. 10: 535–539.
Husebo, B.S., Ballard, C., Sandvik, R., Nilsen, O.B. and Marshall, M. (1999). “They should not really be here” –
Aarsland, D. 2011. Efficacy of treating pain to reduce People with dementia in the acute sector. Age and
behavioural disturbances in residents of nursing Ageing, 28 (Suppl. 2): 9–11.
homes with dementia: Cluster randomised clinical trial. Marshall, M. (2001). The challenge of looking after people
BMJ 343: d4065. with dementia. BMJ, 323: 410–411.
Inouye, S.K. 1993. Delirium in hospitalized elderly Martin, S. and Smith, P. (1996). Explaining variations in
patients: Recognition, evaluation, and management. length of stay in the National Health Service. Journal
Conn. Med. 57(5): 309–315. of Health Economics, 15 (3): 279–304.
Inouye, S.K., Schlesinger, M.J. and Lydon, T.J. (1999). Mukadam, N. and Sampson, E.L. (2011). A systematic
Delirium: A symptom of how hospital care is failing review of the prevalence, associations and outcomes
older persons and a window to improve quality of of dementia in older general hospital inpatients.
hospital care. The American Journal of Medicine, 106: International Psychogeriatrics, 23: 344–355.
565–573. Nightingale, S., Holmes, J., Mason, J. and House, A.
Jackson, T.A., Naqvi, S.H. and Sheehan, B. (2013). (2001). Psychiatric illness and mortality after hip frac-
Screening for dementia in general hospital inpatients: ture. Lancet, 357 (9264): 1264–1265.
A systematic review and meta-analysis of available Pautex, S., Herrmann, F., Le, L.P., Fabjan, M., Michel,
instruments. Age and Ageing, 42 (6): 689–695. J.P. and Gold, G. 2005. Feasibility and reliability of
Jencks, S.F., Williams, M.V. and Coleman, E.A. (2009). four pain self-assessment scales and correlation with
Rehospitalizations among patients in the Medicare an observational rating scale in hospitalized elderly
fee-for-service program. The New England Journal of demented patients. J. Gerontol. A Biol. Sci. Med. Sci.
Medicine, 360 (14): 1418–1428. 60 (4): 524–529.
Joray, S., Wietlisbach, V. and Bula, C.J. (2004). Cognitive Phelan, E.A., Borson, S., Grothaus, L. et al. (2012).
impairment in elderly medical inpatients: Detection Association of incident dementia with hospitalizations.
and associated six-month outcomes. The American The Journal of the American Medical Association, 307
Journal of Geriatric Psychiatry, 12 (6): 639–647. (2): 165–172.
Jordan, A., Hughes, J., Pakresi, M., Hepburn, S. and Poole, M., Bond, J., Emmett, C. et al. (2014). Going home?
O’Brien, J.T. 2011. The utility of PAINAD in assessing An ethnographic study of assessment of capacity and
pain in a UK population with severe dementia. Int. J best interests in people with dementia being dis-
Geriatr. Psychiatry 26(2): 118–126. charged from hospital. BMC Geriatrics, 14: 56.
King, B., Jones, C. and Brand, C. (2006). Relationship Royal Free London NHS Foundation Trust (2014). My
between dementia and length of stay of general medi- discharge a proactive case management for discharg-
cal patients admitted to acute care. Australian Journal ing patients with dementia. In Shine. United Kingdom:
on Ageing, 25: 20–23. Health Foundation.
Kitwood, T. (1997). Dementia Reconsidered: The Person Ryden, M.B., Feldt, K.S., Oh, H.L., Brand, K., Warne, M,
Comes First. Buckingham, United Kingdom: Open Weber, E., Nelson, J. and Gross, C. 1999. Relationships
University Press. between aggressive behavior in cognitively impaired
Laurila, J.V., Pitkala, K.H., Strandberg, T.E. and Tilvis, R.S. nursing home residents and use of restraints, psycho-
(2004). Detection and documentation of dementia active drugs, and secured units. Arch. Psychiatr. Nurs.
and delirium in acute geriatric wards. General Hospital 13(4): 170–178.
Psychiatry, 26: 31–35. Sampson, E.L., Blanchard, M.R., Jones, L. et al. (2009).
Lyketsos, C.G., Sheppard, J.M. and Rabins, P.V. (2000). Dementia in the acute hospital: Prospective cohort
Dementia in elderly persons in a general hospital. The study of prevalence and mortality. The British Journal
American Journal of Psychiatry, 157 (5): 704–707. of Psychiatry, 195: 61–66.
Manthorpe, J. and Moriaty, J. (2010). Nothing Ventured Sands, L.P., Yaffe, K., Covinsky, K. et al. (2003). Cognitive
Nothing Gained: Risk Guidance for People with screening predicts magnitude of functional recovery
Dementia. London, United Kingdom: Department of from admission to 3 months after discharge in hos-
Health. pitalized elders. Journal of Gerontology. Series A,
Marengoni, A., Corrao, S., Nobili, A. et al. (2011). Biological Sciences and Medical Sciences, 58: 37–45.
In-hospital death according to dementia diagno- Saravay, S.M., Kaplowitz, M., Kurek, J. et al. (2004). How
sis in acutely ill elderly patients: The REPOSI study. do delirium and dementia increase length of stay of
International Journal of Geriatric Psychiatry, 26: elderly general medical inpatients? Psychosomatics,
930–936. 45: 235–242.
182 Dementia

Scherder, E., Herr, K., Pickering, G., Gibson, S., Benedetti, Travers, C., Byrne, G., Pachana, N. et al. (2013).
F. and Lautenbacher, S. 2009. Pain in dementia. Pain Prospective observational study of dementia and delir-
145(3): 276–278. ium in the acute hospital setting. Internal Medicine
Shepperd, S., McClaran, J., Phillips, C.O. et al. (2010). Journal, 43 (3): 262–269.
Discharge planning from hospital to home. Cochrane Tullo, E., Khoo, T.K. and Teodorczuk, A. (2015). Preparing
Database of Systematic Reviews, 20 (1): CD000313. to meet the needs of an ageing population – A chal-
Stewart, R., Bartlett, P. and Harwood, R.H. (2005). Mental lenge to medical educators globally. Medical Teacher,
capacity assessments and discharge decisions. Age 37 (2): 105–107.
and Ageing, 34 (6): 549–550. Uwakwe, R. (2000). Psychiatric morbidity in elderly
Tadros, G., Salama, R., Kingston, P. et al. (2013). Impact of patients admitted to non-psychiatric wards
an integrated rapid response psychiatric liaison team on in a ­general/teaching hospital in Nigeria.
quality improvement and cost savings: The Birmingham International Journal of Geriatric Psychiatry,
RAID model. The Psychiatric Bulletin, 37: 4–10. 15: 346–354.
Teodorczuk, A., Corbett, S., Welfare, M. and Mukaetova- Wancata, J., Windhaber, J., Krautgartner, M. and
Ladinska, E. (2013). Reconceptualising models of Alexandrowicz, R. (2003). The consequences of non-
delirium education: Findings of a Grounded Theory cognitive symptoms of dementia in medical hospital
study. International Psychogeriatrics, 25: 645–655. departments. International Journal of Psychiatry
Teodorczuk, A., Mukaetova-Ladinska, E., Corbett, S. and Medicine, 33: 257–271.
Welfare, M. (2014). Learning about the patient: An inno- Westert, G.P., Nieboer, A.P. and Groenewegen, P.P. (1993).
vative interprofessional dementia and delirium educa- Variation in duration of hospital stay between hospitals
tion programme. The Clinical Teacher, 11 (7): 497–502. and between doctors within hospitals. Social Science
Teodorczuk, A., Mukaetova-Ladinska, E, Corbett, S. and and Medicine, 37 (6): 833–839.
Welfare, M. (2015). Deconstructing dementia and delir- Whittamore, K.H., Goldberg, S.E., Gladman, J.R. et al.
ium hospital practice: Using cultural historical activity (2014). The diagnosis, prevalence and outcome
theory to inform education approaches. Advances in of delirium in a cohort of older people with men-
Health Sciences Education: Theory and Practice, 20: tal health problems on general hospital wards.
745–764. International Journal of Geriatric Psychiatry, 29 (1):
Torian, L., Davidson, E., Fulop, G. et al. (1992). The effect 32–40.
of dementia on acute care in a geriatric medical unit. Zekry, D., Herrmann, F.R., Grandjean, R. et al. (2008).
International Psychogeriatrics, 4: 231–239. Demented versus non-demented very old inpatients:
Tosato, M., Lukas, A., van der Roest, H.G., Danese, P., The same comorbidities but poorer functional and
Antocicco, M., Finne-Soveri, H., Nikolaus, T., Landi, F., nutritional status. Age and Ageing, 37 (1): 83–89.
Bernabei, R. and Onder, G. 2012. Association of pain Zwakhalen, S.M., Hamers, J.P., Abu-Saad, H.H. and
with behavioral and psychiatric symptoms among nurs- Berger, M.P. (2006). Pain in elderly people with severe
ing home residents with cognitive impairment: Results dementia: A systematic review of behavioural pain
from the SHELTER study. Pain 153(2): 305–310. assessment tools. BMC Geriatrics, 6: 3.
17
The role of nursing in the management of
dementia

MAREE MASTWYK AND BEVERLEY WILLIAMS

Dementia care is not the care of the dying: it is about meaningful life for the living.

—Wylie (2003, p. 35)

17.1 INTRODUCTION BOX 17.1: The nurse needs

The nurse’s role in the management of dementia patients ●● Understanding, patience, resilience, sense of
encompasses many areas including, but not limited to, humour
assessment and education in memory clinics, provision of ●● Interest/passion in caring for the person with
support services in the home and direct care in hospitals dementia
and care facilities. This chapter will focus on the direct ●● Education, guidance and support in the role
care provided to the person with dementia (PWD) who is
cognitively impaired, is surrounded by strangers and may
be driven by emotions, particularly, fear. The fundamental
tenet of nursing is to provide a safe, secure environment, so
17.2 PATIENT AUTONOMY – ANY
we need to ameliorate that fear. STAGE OF THE ILLNESS IN
Dementia care is a nursing speciality. It is important ANY SETTING
that the nurse has an interest in or, better still, has a pas-
sion in this area and recognizes that the journey through The emphasis today is on person-centred care (Kitwood,
dementia is unique for each individual. In simple words, 1993) to maintain autonomy and dignity. This can only
it is of crucial importance to make the journey as comfort- be accomplished if the opinions of the PWD are sought
able as possible. A nursing assessment is key to achieving and respected (Pringle-Specht et al., 2009). The PWD
this and should include consultation with the PWD and needs a partner to compensate for cognitive losses and
their family; learning their history, background, abilities promote more efficient use of remaining functional abili-
and shortfalls, likes and dislikes. Keep in mind the basic ties (Pringle-Specht et al., 2009), not one who takes con-
human needs: nutrition, hydration, exercise, sleep, hygiene, trol. Nurses need to foster the PWD’s independence for
safety and comfort and be aware that if these needs are not as long as possible. Not only cognitive/functional deficits
being met, the PWD may exhibit ‘challenging’ behaviours need to be considered when drafting a care plan, but the
in order to be heard (Box 17.1). PWD’s abilities, interests and culture are important too.

183
184 Dementia

Self-esteem has an impact on the way we feel about our-


selves, interact with other people, handle problems and it BOX 17.2: Role of nursing
determines our level of relaxation and safety (ReachOut
Australia, 2009). It should be understood that the same ●● Promote independence
does for the PWD as well. ●● Provide a safe, secure environment for patients
PWD can share their knowledge, experience and needs and staff
and are now included in focus groups with other stake- ●● Advocate on the patient’s behalf, when needed
holders, e.g. driving cessation (Perkinson et al., 2005). ●● Educate patient, family, other nurses and staff with
Alzheimer’s Association fact sheets include suggestions regard to patient’s behaviour
from PWD (Alzheimer’s Australia, 2012b). Pringle-Specht ●● Promote health, prevent illness/injury
and Bossen consulted Taylor, a person with probable
Alzheimer’s disease (AD) for their paper Partnering for
Care: The Evidence and The Expert, in March 2009. PWD ●● INTRODUCE yourself, ask if the PWD remembers you
are able to guide us in designing strategies for care for them when you go into the room on later occasions and intro-
and others suffering like them. duce yourself again if necessary.
In later stages, the family can be a valuable resource of ●● EXPLAIN every procedure, even if it happens every
information about the PWD. However, until unable to com- day during the admission, and even if you are only
municate, he or she can be consulted directly about future in the room to replace a filled urine bottle. You may
plans, needs and wants. Alzheimer’s Australia (2012a) rec- need to remind the PWD frequently why they are in
ommends planning with the PWD soon after diagnosis to hospital. This may be frustrating for you but is more
enable them to express their desires, wants and needs for frustrating and confusing for the PWD if there is no
consideration at the time when they are no longer able to understanding.
express themselves, including medical, financial and legal ●● Focus the PWD’s attention. If two nurses are required
concerns. This planning should also include end-of-life for a procedure, only one should instruct the PWD if
decisions (see Chapters 33 through 35). their cooperation is required to complete a task.
Planning by the family on behalf of the PWD can be dis- ●● If you need a distraction, chat about the PWD’s past
tressing for the family. If important decisions are made in life – job, family, hobbies. Remember that these early
consultation with the PWD early, this can remove a poten- memories will be the last to fade away.
tial source of stress from the family as the PWD’s life nears ●● Ask permission before helping yourself to the locker or
its end. wardrobe. We see these cupboards as an extension of
Person-centred care requires person-centred staff and our working environment – the PWD may misinterpret
management to promote and provide a supportive envi- your movements as theft.
ronment. Residents in a nursing home will still have some ●● The PWD may not be able to report pain – look for non-
self-awareness and some ability to participate in decisions. verbal cues: grimacing, restlessness, agitation.
Allowing them to make small simple choices for themselves ●● Use caution when giving pro re nata (prn) medication
can help maintain their self-esteem (Box 17.2). for agitation – pain may be the cause.
●● Extend visiting hours for next of kin, other family
members and friends – hospitals are extremely noisy
17.3 HOSPITAL NURSING CARE environments and some noises can be frightening for
the patient. If a trusted visitor is present, they can help
explain what is going on and keep the PWD calm.
Caring for a PWD in a hospital setting requires patience ●● Place an orientation board in the line of sight of the
and a calm approach. Most PWDs will recall aspects of PWD with the day, date, location and why the PWD is
a hospital admission, despite their dementia. Even if the in the hospital.
patient has severe dementia, unpleasant feelings and emo- ●● Facilitate watching favourite programmes on television
tions from previous admissions may still be remembered, for the PWD.
so nurses need to approach the patient in a friendly, non- ●● Ask the family to bring familiar objects and photos into
threatening manner. Remember, the PWD is in a strange, the room and to play the PWD’s favourite music.
noisy environment and surrounded by strangers.

17.3.1 CARE OF A PWD IN A GENERAL 17.3.2 EMERGENCY DEPARTMENT (ED)


WARD
Finding a quiet corner for the PWD in an ED is going to be
●● Provide a single room if possible to minimize noise and difficult. It is important to have family close and attempt
confusion; preferably one with an en suite so that the to minimize the stimulus from hospital equipment/hustle
toilet can be found easily. and bustle as much as possible. Every time you approach
●● Greet the PWD by name. the PWD, introduce yourself. Increased stress and pain can
The role of nursing in the management of dementia 185

analgesic regimen and continue to observe, evaluate and


BOX 17.3: Key points to caring for people document patient behaviour.
with dementia Medications, when used well, can assist pain manage-
ment. However, the fact that older people have an increased
Key points when caring for a person with dementia in sensitivity to some drugs, especially opiates, must be con-
any environment are: sidered. If an increased sedative effect is present, this may
●● Remember: the patient is in a strange environ-
lead to falls and fractures, loss of muscle strength, immobil-
ment, surrounded by strangers
ity and decreased cognition (see Chapter 16).
●● INTRODUCE yourself and the task to be carried
Depending on the cause of pain, regular paracetamol is a
out to prevent fear in the patient
safer option than anti-inflammatories, which may have gas-
●● Allow autonomy, enhance self-esteem (allow
trointestinal or other side effects.
choices, maintain abilities)
An effective approach to pain relief is to combine
●● One stimulus/voice at a time to focus the patient’s
­medication with a non-drug approach: massage, heat, exer-
attention
cise – all to increase circulation – or distraction. As tired-
●● Treat patient with dignity
ness can increase pain, take into account the PWD’s sleep
●● Maintain routine
pattern too.
●● Do not ‘infantilize’ the patient
Regular medication reviews are required to avoid unnec-
●● The patient will remember EMOTION
essary polypharmacy. Medications prescribed earlier in
life may now be unnecessary or causing side-effects. Used
wisely, medications can help with symptoms of pain, agita-
cause changes in behaviour – ‘exiting behaviour’ in particu- tion and anxiety. Medication can improve sleep at night and
lar, trying to ‘walk away’ from the cause of pain or change increase daytime participation in activities.
in facial expressions (James and Hodnett, 2009) are com-
mon. Hospital volunteers can be of benefit to the PWD,
staff and carers by remaining with the PWD and explaining
procedures where appropriate (James and Hodnett, 2009). 17.5 RESIDENTIAL CARE
Providing training in dementia care for staff will improve
outcomes by reducing stress for both PWD and staff (James 17.5.1 ASSESSMENT
and Hodnett, 2009) (Box 17.3).
Dementia care today is based on person-centred care,
founded on a holistic model designed and developed by
the late Tom Kitwood and Kathleen Bredin during the late
17.4 PAIN AND MEDICATION 1980s (Kitwood, 1993). The basic tenets of this work are the
following:
Experiencing pain is different for everybody. People have
different pain thresholds which may further be lowered by ●● Uniqueness – Everyone is different.
fatigue and depression. For those who are cognitively intact, ●● Complexity – We all see things differently and are
the presence of pain is easily articulated. Thus preferred, influenced by the changes and events that occur during
effective medications and other methods of pain relief are our lifetime.
immediately provided.
Cognitive impairment is a substantial barrier to the Most facilities will have their own PWD assessment tool.
assessment and management of pain. It is common for Whichever tool is used, the assessment needs to be person-
older PWDs to experience pain associated with arthritis, centred and keep to the PWD’s at home routine as much
spinal canal stenosis, diabetic neuropathy, painful legs, etc. as possible. For example, if they showered in the evening
A PWD may be incapable of reporting pain verbally. The at home, can this be maintained? Information gathering
nurse needs to know the PWD’s past record and current should involve the PWD, include the family and close
medical history, both, to identify possible causes of pain and friends, list abilities, needs and details of the person’s life
to facilitate appropriate management. story so that new staff can quickly gain an insight into the
If the PWD is unable to communicate adequately, our person they may be caring for on a particular day. Of course,
observations and knowledge of the PWD need to be relied assessment of fall risks, compliance with special diets, etc.,
upon. Gait, facial expressions (frown, grimace), skin colour, also need to be included, but the emphasis needs to be on
behaviour, placing cold or hot things on a body part – can the PWD – their life, culture, things they love, things they
all be observable indicators of pain. Vital signs (heart rate, hate, usual routine. Moving into a care facility will cause con-
blood pressure, temperature) can provide important infor- fusion and the PWD will need support. Talking to the PWD
mation, especially in the assessment of non-verbal, elderly helps them feel welcome and use of knowledge obtained
dementia patients. Observe, gather information from all about PWD will assist in maintaining their self-esteem and
team members, consult with the doctor, commence an safety, thus reducing anxiety and contributing to a settled
186 Dementia

environment for all. A brief, dot-point biography of the ●● To avoid surfaces/paints that reflect light. Sun glare may
PWD in front of the notes documented would be helpful. give the appearance of spills on the floor.
Once the assessment and a comprehensive care plan are ●● Floor coverings that are easy to clean, not slippery
complete, it is important to establish a routine in the daily care and absorb noise. The floor surface should be level.
of the PWD. If they are capable of dressing with supervision Particular care needs to be taken if bedroom areas are
it would be an insult to dignity, autonomy and self-esteem to carpeted and day rooms are surfaced with linoleum.
dress them yourself for expediency. It is important to work at Surface changes may contribute to falls.
the pace of the PWD. Information gathered during the assess- ●● Hand rails on all walls.
ment can also be used to distract the PWD at times of potential ●● Adequate lighting. No dark corners or shadows which
distress/anxiety during the provision of care and can also be may distort shapes to produce something frightening.
used in reminiscence and validation therapy (see Chapter 23). ●● Appropriate signage – at eye level, perhaps use the
The care plan needs to be reviewed and amended regularly as accepted universal symbols for the toilet as well as the
needs of PWD change over time. Two PWDs with the same word toilet; pictorial dinner plate with knife and fork on
diagnosis may be very different in their daily functioning. either side for the dining room, etc.
●● An outside area to walk in, sit for awhile or, perhaps,
17.5.2 A NOTE ON CULTURE participate in gardening activities.
●● Background music – PWD choice, or relaxing,
Some residential facilities are culture specific and have car- ­non-stimulating music.
ers from the same cultural background. The PWD’s culture ●● To provide a PWD’s individual bedroom area –
needs to be considered if this is not the case. Despite cog- ­personalized with familiar furniture and recognizable
nitive deficits, participating in religious practices can still belongings, family photograph on the door, pictures/
bring comfort to the patient. Family or friends can help photographs of relatives on the walls. Asking the family
provide this. PWDs of non-English speaking backgrounds to help with this arrangement will help them feel they
may have spoken the local language fluently prior to diag- are still contributing towards caring for the PWD.
nosis, but with the progression of dementia, may revert back
to their native tongue. PWDs approached with dignity and
respect are more likely to respond positively. Non-verbal
communication – facial expression, tone of voice, gentle 17.7 ENVIRONMENT – SOCIAL
directing – can be powerful.
Everyone requires daily mental stimulation. A PWD who
cannot walk, talk or participate actively in group activities
can still be a passive observer. Previous leisure activities can
17.6 ENVIRONMENT – PHYSICAL still be enjoyed with a little creative modification. Most peo-
ple enjoy dancing. PWDs who cannot dance will still benefit
PWDs with dementia may have perceptual difficulties. Even from being present in the room while others dance – they
in the absence of cognitive difficulties, an older person may can enjoy the movement, music and rhythm. Sports fans
have impaired sight, hearing or a gait disturbance. Older can still enjoy watching sports on television. Appropriate
people need a simple, uncluttered, well lit environment diversion therapies can be provided by occupational or
that is safe to navigate. Family can assist by personalizing diversional therapists (see Chapter 19).
the PWD’s living space to make it identifiable to the PWD. Multi-sensory stimulation can be provided with a
Colour and material choices are also important. Even in an Snoezelen room, a Dutch initiative, which may be useful in
old building appropriate change can be made during build- calming agitated PWDs. As its objective is to stimulate the
ing maintenance. An ideal environment for the PWD needs: PWD’s senses – sight, sound, smell, touch and taste – the
room may require: comfortable seating, a solar projector,
●● To be calm and welcoming and not too busy. Advise all bubble column, a revolving mirror ball, soft gentle music
staff to avoid unnecessary noise (e.g. not to drop a large (i.e. sounds of nature, aromatherapy oils/candles and many
bundle of cutlery into a sink). other innovative ideas that assist to soothe the PWD). Such
●● Contrasting colours between walls, floors and doors. rooms promote relaxation through sensory stimulation and
Toilet doors particularly, need to be of a different colour can be calming for agitated PWDs, though monitoring is
to differentiate them from other doors, so that the PWD needed because everyone’s experience is different and some
can identify and find the toilet when needed. In bath- PWDs may become distressed or more agitated by these
rooms, if the walls, floor, tiles, toilet, basin are all white, a arrangements (Wylie, 2003).
PWD with visuospatial difficulties may be unable to find Any sensory stimulus can be therapeutic. This might
the toilet and may become agitated, incontinent or use include sitting in the fresh air, enjoying the warmth of the
the black-lined rubbish bin instead. Coloured toilet role sun, the sounds of leaves in the breeze and birds chirping,
holders, towel dispensers and taps make them easier to or smelling the aroma of cooking when one is unable to
locate. cook.
The role of nursing in the management of dementia 187

Family and friends can contribute to the This Is Your Life 17.8.2.1 Perceptual impairment
book (see Section 17.9.1.2) continually and can see who has
visited when and it can be used to provide conversation/ Due to confusion, the PWD could find themselves living in
reminiscence prompts. a state of fright perpetually. A black jumper on furniture
Music can be an entertainment, used as a therapy to may look like an animal; a PWD with Balint’s syndrome
promote memories, stimulate sensation and conversation (difficulties with visual perception) may not be able to see
(Baird and Samson, 2015), or to calm agitated PWDs (see what is directly in front of them causing feelings of insecu-
Chapter 29). The choice of music should be the PWDs, not rity, making them prone to falls and unable to see food on
what the staff or family chooses. It is the PWD’s home, the plate right in front of them. Dementia may affect visuo-
everybody else is a guest there. spatial awareness: glaring light on pale floors may look like
water and the PWD may try to step over it risking a fall.
The nurse needs to be alert to the PWD’s sensory impair-
ments in order to provide assistance when required. This may
17.8 TIME, PLACE, PERSON include helping with meals, moving clothes from one place to
another or directing the PWD away from potentially danger-
ous or anxiety provoking situations. This may be as simple
17.8.1 ORIENTATION as using coloured crockery for distinguishing food items
The PWD’s peace of mind is more important than their or standing in a patch of light on the floor to avoid visual
knowledge of time and place. If a 93-year-old woman thinks misperception.
that her aunt still lives down the certain street ‘near here’,
there is no point in causing distress by telling her that she is
93, the year is 2016 and that her aunt could not possibly still
be alive. It would be better to comment on the fond memo-
17.9 COMMUNICATION THERAPIES
ries she must have of her aunt and encourage her to talk
about those. Deficits in the ability to communicate are often an early
When orientation of the PWD is important, such as meal symptom of dementia: word finding difficulties, partial
or shower times, the PWD should be greeted appropriately comprehension of conversation, problems with reading and
in a non-threatening way, to alleviate any fear felt at being writing. Communication is made up of three parts: 55%
approached by a stranger and to gain cooperation. A little body language (facial expression, posture, gestures); 38%
thought is required to provide the information needed for tone and pitch of the voice; only 7% is speech (Alzheimer’s
this task without putting the PWD under pressure. For Australia, 2012b). It is dependent on the listener listening
example, ‘Hello Therese, I’m Ann, one of the nursing staff. and patiently giving the speaker time to speak. Give the
It’s 12 o’clock and lunch will be served very soon. Let me PWD the respect of waiting for their response rather than
walk with you to the dining room’. In this context, orienta- second guessing for them or speaking on their behalf.
tion to person, time of the day and purpose of the activity
has been explained and the PWD does not need to make a 17.9.1 THERAPIES FOR MODERATE TO
decision. SEVERE DEMENTIA
When PWDs do not recognize family members, it is
often distressing to the family but not necessarily to the
PWD. The PWD may still remember the emotion associated 17.9.1.1 Reminiscence therapy (RT)
with that family member and smile or say ‘hello’ when they RT involves discussion of past events and can be a posi-
enter the room. It is important to point out to the family tive activity even if the PWD cannot participate actively
that even though the PWD cannot recall their name, they (Alzheimer’s Australia, 2012c). Photographs, pictures, music,
are still happy to see them. TV shows, books, can be used to trigger memories. It is
important to know your PWD before involving them in RT,
as some memories may be distressing. RT does not always
17.8.2 PERCEPTION
need to be conducted as structured group therapy. It can also
Other factors to consider are disturbances of perception be used as a device for distraction at any time – in the ED or
and misinterpretation of stimuli. The PWD’s reality is dif- anywhere when the PWD is becoming agitated.
ferent from ours. They may tell their family that they want
to go home. If the family takes the PWD home for a visit, 17.9.1.2 Validation therapy (VT)
the PWD may say that he or she still wants to go home.
Perhaps ‘home’ means a calm and safe place where they The PWD’s reality is different from ours. By entering their
feel comfortable and at peace. Whilst in a state of anxiety, a reality, we can establish feelings of trust and security in
PWD may become agitated and become a challenge for staff the PWD and alleviate distress and anxiety (Alzheimer’s
attempting to provide care. Australia, 2012b). VT consists of exploring an issue raised
188 Dementia

by the PWD, even if it obviously may have no bearing on All activities need to be tailored according to the needs
the present. It is useful when the PWD’s short-term memory and capabilities of the individual (Alzheimer’s Australia,
has deteriorated to the point that understanding of the pres- 2012d).
ent is cognitively beyond them and they have reverted to
memories from their past to survive (Alzheimer’s Australia,
2012c). The person attending the PWD can promote discus-
sion about their memories and the emotions they felt in the 17.11 THE NEED FOR SLEEP
past. It does not matter if the details do not correlate with
reality. The PWD’s peace of mind is the goal to be achieved. Sleep hygiene must be considered. Lack of sleep has a
VT can be used to engage the PWD in conversation and negative impact on the PWD’s behaviour and ability to
promote mental stimulation or to distract or redirect them participate/function in activities in a meaningful way. An
from doing something inappropriate without causing them appropriate sleeping environment must be provided and
distress. other basic needs met: absence of pain, hunger, empty
A This Is Your Life book compiled by a family member or bowel/bladder. Staff should be mindful of daytime rest
friend can be an excellent communication tool. Such a book periods – catnaps are acceptable, a lengthy period of sleep
can include photographs, letters, personal details, such as: is not. PWDs need to be involved in daytime activities/exer-
immediate family tree, details of birth, anecdotes from fam- cise both indoors and outdoors in order to feel tired enough
ily folklore, qualification certificates. Keep one idea to each to sleep at night. The sleep/wake cycle can be reversed in
page in the book to avoid overwhelming the PWD and place dementia and daytime activities can assist normal diurnal
labels on photos rather than testing the PWD’s memory awareness. Night sedation should be the last resort after all
(Alzheimer’s Australia, 2012b). other strategies have been attempted and have failed.

17.10 EXERCISE/ACTIVITIES 17.12 NUTRITION

‘Enjoyment doesn’t require memory’ (Alzheimer’s Nutrition for PWD can be assisted by the environment in
Australia, 2012d). Exercise has a beneficial effect on both which food is served. A noisy environment can be distracting
physical and mental health. In a literature review, Leone and frightening. A lack of sensory cues (smell, sight, touch,
et al. (2008) found that exercise, particularly, can have a taste and sound) will not encourage a person with dementia
positive effect on depressive symptoms, chronic diseases to eat (Marshall, 2003). PWDs who have come from a small
and on the behavioural and psychological symptoms of home unit into a multi-bed facility may find meal times
dementia (BPSD). Exercise can stimulate cognition and confusing and overwhelming. A calm environment, back-
the performance of activities of daily living in PWD. The ground music and the opportunity to help with cooking and
risk of falls can be reduced through balance and strength serving may stimulate appetite (Marshall, 2003).
training. Food should be presented attractively. Smaller, more fre-
Benefits of exercise include: quent meals or nutritious finger foods may benefit small eat-
ers. Fluids should be offered frequently and in small quantities
●● Improved general well-being to be taken easily. Some PWDs may require assistance to eat
●● Improved blood circulation because of a perceptual difficulty or severe dementia. Some
●● Strengthened immune and hormone systems may need a trigger (coloured crockery or cutlery handles) or
●● Increased lean muscle mass cue, such as a guiding hand over theirs, to remind them of the
●● Increased metabolism action of eating. A staff member eating with the PWDs may
●● Improved balance and coordination benefit. Adequate time to eat must be provided and plates not
●● Positive effect on BPSD removed hastily because kitchen staff needs to go for lunch.
●● Enjoyment Care is provided to benefit the PWD and not the staff.

Types of exercises that are beneficial:


17.13 BEHAVIOURAL AND PSYCHIATRIC
●● Aerobic (walking, dancing, any exercise which requires
movement)
SYMPTOMS OF DEMENTIA (BPSD)
●● Progressive strength training (resistance and weight
bearing) Not all PWDs exhibit BPSD. Nursing education requires
●● Flexibility and balance training (tai chi, bending/ knowledge of the different types of dementia, associated
stretching) symptoms and expected progression of each disease. Basic
The role of nursing in the management of dementia 189

understanding of the functional parts of the brain must


underpin any understanding of the behaviours exhibited. BOX 17.4: Behavioural and psychological
Two people with the same diagnosis and same degree of symptoms of dementia
atrophy may not necessarily display the same problem in
their behaviours. Symptoms and responses to treatment Depression, lowered mood, apathy, aggression,
change during the course of the illness. agitation, resistance to personal care, wandering,
Challenging behaviours are not deliberate and staff intrusiveness, pacing, sleep disturbance, verbal
should not take such behaviours personally. It is a waste of outbursts or obscene abusive language, stealing,
time arguing with a PWD who does not have the capacity hiding, h
­ oarding, inappropriate toileting, inappropri-
to reason. A PWD may not be able to process your words, ate sexual behaviour, repetitive acts, suspicion and
however, if delivered with a smile, warmth, a soft tone and accusations, anxiety, delusions, hallucinations
gentle touch, the person may respond in a more appropri-
ate manner. Communication is both verbal and non-verbal.
The message conveyed in a non-verbal manner may be more 17.13.1 DISTRACTIONS – ‘QUICK-FIX’
powerful than spoken words (Box 17.4). SOLUTIONS
When are these behaviours most likely to occur?
Another technique to deal with BPSDs is to recognize
●● Attending personal care/bathing the precipitant and redirect or distract the PWD to try
●● Dressing to prevent such behaviour. The fundamental require-
●● Meal times ment for a quick fix solution is to know your PWD. If the
●● Late afternoon (sundowner syndrome occurs due to PWD is becoming anxious, agitated or frustrated, greet-
fading light) ing them warmly, addressing by name with and an open,
●● Night-time outstretched hand may be all that is needed to calm them
●● Anytime down. Engage the PWD in a meaningful conversation – life
pre-retirement, their pastimes – anything to distract them
Those behaviours that are disruptive to others need to and focus their attention on something else.
be addressed. However, some behavioural patterns, such There are many reasons why a PWD may exhibit problem
as pacing and repetitive acts, need not necessarily disrupt behaviour, most probably precipitated by an unmet need.
anyone. PWDs can rummage through a box or drawer pro- The solution does not always need to be pharmacological.
vided a purpose, or wander around if it is safe in the ward In managing a challenging behaviour, a holistic problem-
area. Sundowner syndrome may be alleviated by turning on solving approach can be adopted to formulate strategies to
lights, closing curtains and not allowing shadows to be cast modify/eliminate/better manage their behaviour.
in the room. Any behaviour only requires intervention if it A simple problem solving model consists of:
is distressing to the PWD or others. The nursing aim is to
minimize the occurrence of any negative behaviour. Gathering information: What is the behaviour of concern?
Bathing often triggers BPSD. This is perhaps due to fear, When does it happen? What are the triggers? How
embarrassment and/or physical discomfort. Previously, long does it persist? Who/what is it directed towards?
this PWD has bathed in private. An alternative to routine Is it due to boredom? Is it stimulated by a past mem-
showering or bathing, such as a traditional bed/sponge bath ory? Can the family shed any light into the matter? Is
using warm cloth steeped in non-rinse cleanser may help there an organic basis: urinary tract infection, pain,
(O’Connor et al., 2009). sensory loss, constipation?
Multi-sensory activities and exercise can help minimize Assessment: By team – pooling of information from all
BPSD. Mental stimulation and exercise are basic human sources.
needs and unmet needs can create problems. Planning: All members of the team contribute to develop-
Other stimulating strategies to moderate or prevent ing a strategy to change the behaviour of the con-
BPSD are addressed in Chapters 23 and 24. Most require cerned PWD. A review date is set.
human contact, so the personal interaction between the Implementation: ALL members of the team follow the
PWD and person providing care may be the key to prevent plan on a daily basis; no saboteurs.
BPSD (O’Connor et al., 2009). Evaluation: By team members regarding success or failure
Sudden behavioural changes may be due to delirium of the strategy. Accurate documentation and commu-
caused by an infection (commonly urinary tract infection nication between team members is imperative.
[UTI]), dehydration, electrolyte imbalance, constipation Reiteration: Modification of management strategies based
or nutritional deficiency. These are all treatable, reversible on the known successes/failures of those strategies
conditions. already tried.
190 Dementia

Training needs to be relevant and meaningful. A recent


17.14 CARE OF THE FAMILY Australian paper found out that staff were aware of the need
for education and training but ‘were critical of the content
…loved ones are not lost to us because they relevancy to direct care practices’ (Jones et al., 2013, p. 52).
have dementia. Staff turnover in nursing homes varies greatly. Recent
research indicates that effective leadership, education and
—Young-Mason (2009, p. 43) support contribute to staff retention in nursing homes
(Chenoweth et al., 2010). The provision of education
Family members grieve and fear for the PWD from the and clinical supervision alleviates stress and positively
moment of diagnosis. At different times they will feel lost, affects job satisfaction (Brodaty et al., 2003). It needs to be
angry and frustrated. There are carer support s­ervices acknowledged that the work can be unpleasant and seem-
­available to which the nurse can refer carers while provid- ingly unrewarding but education and support can mitigate
ing care at home and during the ­transition from home care the negativity.
to a residential facility. Alzheimer’s Associations conduct The people providing the care need to have a voice.
sessions for carers to learn ­effective ways of ­dealing with Supportive facility managers acknowledge the need for reg-
problem behaviours (see Chapters 15 and 36). ular staff forums and provide a budget for them. Strategies
The issues faced by carers after institutionalization of to provide a forum for nurses to receive education and sup-
the PWD are under-researched. Some carers visit rarely port in the workplace do not need to be expensive. They
whilst others stay on for most days. Some carers feel could include:
redundant, guilty or isolated. All carers will require sup-
port from the nurse as well, especially when the PWD 1. Regular support sessions
is newly admitted. It helps the family to involve them- 2. Regular education sessions
selves with the PWD’s care – with the assessment, deco- 3. Debriefing sessions or extraordinary meetings to
rating the PWD’s room, providing stimulation through discuss specific issues that occur between regular
reminiscence, taking the PWD for walks, etc. They will meetings, such as the death of a long-term resident,
require explanations if the PWD’s behaviour is problem- a challenging PWD behaviour or a difficult family
atic or if health is compromised. A family member with member
enduring power of attorney may need guidance regarding 4. Remunerate night staff for attendance and keep a
choice of appropriate treatment at times, if an advanced register to ensure that every staff member has the
care plan is not in place. This issue should be referred opportunity to attend regularly
appropriately.
Perhaps, by providing a structured, safe environment for
the PWDs and a supportive environment for the nursing
staff, an effective milieu for both can be created.
17.15 CARE OF THE CARE PROVIDERS

Qualified nurses working exclusively with PWDs will


largely be working in administration, management or in 17.16 CONCLUSIONS
the community. Most of the care in dementia specific nurs-
ing homes is provided by personal care attendants (PCAs), This chapter is written from a practical perspective. When
supervised by a registered nurse. It is incumbent on the all else fails, it is important to remember that common
nurse manager to educate, support, mentor and lead by sense is a great tool when dealing with the complexity and
example in the workplace. diversity we face with people. PWDs, in some cases, amplify
the requirement that enables us to think clearly about and
17.15.1 STAFF EDUCATION make well-judged and informed decisions. The victims of
dementia are not limited to PWD alone. This chapter aims
In any medical facility there are a host of different workers to provide ideas to stimulate discussion that result in better
who come in contact with PWDs – from medical staff to support for the care of the unfortunate people who endure
cleaners. All staff in a facility that regularly caters to PWDs these degenerative and devastating illnesses.
requires some degree of training. In a facility in Suffolk,
England, nonclinical staff doubted the necessity of demen- People are whole people until the moment of
tia training given to them, but post-training reviews dem- their death.
onstrated that training was valuable to all departments,
including the laundry staff (Moden, 2009). —Pringel-Specht et al. (2009, p.16)
The role of nursing in the management of dementia 191

REFERENCES Kitwood, T. (1993). The person and process in demen-


tia. International Journal of Geriatric Psychiatry,
Alzheimer’s Australia (2012a). Helpsheet – About 8: 541–545.
Dementia 07 ‘Early Planning.’ Available at https: Leone, E., Deudon, A. and Robert, P. (2008) Physical
//f­ightdementia.org.au. Accessed 16 October 2014. activity, dementia and BPSD. Journal of Nutrition and
Alzheimer’s Australia (2012b). Helpsheet – Caring for Aging, 12: 457–360.
Someone with Dementia 01 ‘Communication.’ Marshall, M. (2003). Nutrition. In R. Hudson (ed.),
Available at https://fightdementia.org.au. Dementia Nursing, A Guide to Practice. Melbourne,
Accessed 16 October 2014. Australia: Ausmed Publications Pty Ltd.
Alzheimer’s Australia (2012c). Helpsheet – Caring Moden, L. (2009). Passionate about dementia care train-
for Someone with Dementia 02 ‘Therapies and ing. Nursing Older People, 21 (1): 15.
Communication Approaches.’ Available at https: O’Connor, D.W., Ames, D., Gardner, B. and King,
//fightdementia.org.au. Accessed 16 October 2014. M. (2009). Psychosocial treatments of behaviour
Alzheimer’s Australia (2012d). Helpsheet, Caring for symptoms in dementia: A systematic review of
Someone with Dementia 05 ‘Activities.’ Available at reports meeting quality standards. International
https://fightdementia.org.au. Accessed 16 October Psychogeriatrics, 21 (2): 225–240.
2014. Perkinson, M.A., Berg-Weber, M.L., Carr, D., et al. (2005).
Baird, A. and Samson, S. (2015). Music and dementia. Driving and dementia of the Alzheimer type: Beliefs
Progress in Brain Research, 217: 207–235. and cessation strategies among stakeholders. The
Brodaty, H., Draper, B. and Low, L.-F. (2003). Nursing Gerontologist, 45 (5): 676–685.
home staff attitudes towards residents with dementia: Pringle-Specht, J.K., Taylor, R. and Bossen, A.L. (2009).
Strain and satisfaction with work. Journal of Advanced Partnering for care: The evidence and the expert.
Nursing, 44: 583–590. Journal of Gerontological Nursing, 35: 16–22.
Chenoweth, L., Jeon, Y.-H., Merlyn, T. and Brodaty, ReachOut Australia. (2009). Fact sheet ‘Feeling OK
H. (2010). A systematic review of what factors attract about who you are.’ Available at www.reachout.com.
and retain nurses in aged and dementia care. Journal Accessed 19 August 2009.
of Clinical Nursing, 19 (1–2): 156–167. Wylie, K. (2003). Enriching the environment.
James, J. and Hodnett, C. (2009). Taking the anxiety out In R. Hudson (ed.), Dementia Nursing, A Guide to
of dementia. Emergency Nurse, 16: 10–13. Practice. Melbourne, Australia: Ausmed Publications
Jones, C., Moyle, W. and Stockwell-Smith, G. (2013). Pty Ltd.
Caring for older people with dementia: An exploratory Young-Mason, J. (2009). Family and friends create hope
study of staff knowledge and perception of training in and understanding for those with dementia. Nursing
three Australian dementia care facilities. Australasian and the Arts, 23 (1): 43–44.
Journal on Ageing, 32: 52–55.
18
Social work and care/case management
in dementia

DAVID CHALLIS, JANE HUGHES AND CAROLINE SUTCLIFFE

Moreover, there is evidence of its effectiveness within this


18.1 INTRODUCTION group provided that it is appropriately targeted. In many
developed countries, care/case management services have,
Social work, like any professional or occupational group, is it has long been argued, been intended for those people
inseparable from the organizational and legislative context at risk of admission to long-term care reflecting a policy
in which it takes place since this defines the parameters of of ‘downward substitution’ by the provision of more cost-
practice. Prior to the introduction of care/case management, effective community-based alternatives (Challis, 2003).
descriptions of social work in services for adults, including Two examples demonstrate the international applicability
older people, tended to focus on profession specific activi- of care/case management as a means to provide effective
ties, to the neglect of outputs such as the function of link- community-based care for older people with dementia.
ing the individual to networks of care. Moreover, social Early research in the United States revealed that this target
work with older people, including those with dementia, was group experienced a greater reduction in their use of health
typically a short term intervention (Hunter et al., 1990). care and had lower costs within an approach where care/
Assessments were often undertaken in a relatively narrow case managers had small caseloads; undertook frequent
and service-oriented fashion, followed by an allocation of home visits; with appropriate use and knowledge of local
service prior to closure (Challis, 2003). Continuing manage- resources (Eggert et al., 1990). In England, an exemplar
ment of long-term problems for people living in the com- care/case management site also demonstrated its capacity
munity was neglected. The advent of care/case management to support those with dementia in their own homes. Key
required a useful redefinition of social work in relation to characteristics of the service were the following: trained
long-term care of older people. It can be most simply defined and experienced staff; small caseloads targeted on the
as a strategy for organizing and coordinating care services most vulnerable older people; long-term support; finan-
at the level of the individual. Care/case management, there- cial management arrangements in which care costs were
fore, involves mobilizing and influencing various agencies explicit, care packages costed and, within limits, bud-
and services to achieve clearly formulated goals, rather than gets devolved to front-line staff; and systematic records
each provider pursuing separate and perhaps diverse goals for assessment and monitoring (Challis et al., 2002a). A
(Challis, 1993; Schultz and McDonald, 2014). It is helpfully recent Cochrane review of case management and dementia
understood in terms of six criteria: the performance of core indicated reduced admissions to nursing and residential
tasks, effective coordination, explicit goals, a specific target homes and lower healthcare costs from case management.
population, a long-term care focus and an impact on service However this was dependent upon key factors: targeting
development as well as individual cases. This is summarized the intervention on the correct population; a clear and
in Box 18.1. Various terms are used to describe this process explicit specification of the form and content of the case
including case management, care management and care management intervention and related to this, model fidel-
coordination (Challis and Hughes, 2012). ity (Reilly et al., 2015). Other work indicates the impor-
Specialist care/case management for older people with tance of the degree of influence over resources which case
dementia is an area of practice in which there is much managers possess, where more control is associated with
scope for innovation and development (Challis et al., 2009). better outcomes (Challis, 2003).

192
Social work and care/case management in dementia 193

services (Welsh Assembly Government, 2011). In Australia,


BOX 18.1: Key characteristics of care the strategic vision for the development of services suggests
management a not dissimilar role for social workers, that of helping peo-
ple with dementia and their families ‘navigate’ the commu-
●● Core tasks: case finding and screening; assess- nity care system with provision located within mainstream
ment; care planning; monitoring and review services (AHMC, 2006; Brodaty and Cumming, 2010). By
●● Functions: coordination and linkage of care contrast, the Norwegian strategy for people with dementia
services emphasizes care planning, the implementation of home care
●● Goals: providing continuity and integrated care; services and day care programmes and the development of
increased opportunity for home-based care; make a competent workforce (Norwegian Ministry of Health and
better use of resources; promote well-being of Care Services, 2007; Engedal, 2010).
older person The core tasks of long-term care embodied in care/case
●● Target population: long-term care needs; multiple management, detailed in Figure 18.1, offer an approach suit-
service requirements; risk of institutional placement able for the community-based care of vulnerable people with
●● Differentiating features of long-term care: inten- long-term conditions. Since older people with dementia typi-
sity of involvement; breadth of services spanned; cally have complex health and social care needs, it is impor-
lengthy duration of involvement with older person tant that agencies have in place procedures and protocols
●● Multi-level response: linking practice-level activities within care/case management arrangements which facilitate
with broader resource and agency level activities an appropriate level of response, a feature more likely to be
associated with a differentiated approach to care/case man-
agement. This requires a distinction to be made between
older people with complex needs requiring a multiservice
18.2 PATTERNS OF PRACTICE
response and those with less complex needs which are met
by a single service response provided by one agency (Hughes
Increasingly countries are developing strategies for the care et al., 2005); however, it is not always available. In England,
of people with dementia which include enhanced care at for example, little progress was made in the development of
home, thereby demonstrating the important contribution a differentiated approach to care/case management follow-
of social work and care/case management to such service ing the community care reforms of the 1990s. Intensive care/
development. This is detailed in international strategies and case management, an important component of a differenti-
plans in, for example, France, Holland, Ireland and Japan for ated approach, was not widespread within older people’s ser-
people with dementia, with care/case management identified vices (Sutcliffe et al., 2008; Chester et al., 2015). The purpose
as the means to coordinate the necessarily multiple service of intensive care/case management is to permit more flexible
inputs as an alternative to care home admission (Ministry responses through improved coordination and appropriate-
of Health Welfare and Sports, 2009; Arai et al., 2010; Cahill, ness of care by use of a designated care manager, and is a
2010; Guisset-Martinez, 2012). The four countries within the prerequisite for the provision of complex packages of care to
United Kingdom have their own dementia strategies which enable older people with dementia to continue living in their
have either adopted a care management/care coordination own homes (Applebaum and Austin, 1990; Challis, 2003;
approach (Department of Health, 2009, 2013; Department of Department of Health, 2009; NICE, 2013).
Health, Social Services and Public Safety, 2011; The Scottish A review of case management operation and process has
Government, 2013), or have proposed better integration of indicated that clarity of definition about the form, content

Re-referral

Assessment Brief
Referral Closure
intervention

(i) Short-term interventions

Case finding Care Monitoring Closure


Assessment planning and review
and screening

(ii) Long-term care

Figure 18.1 A model of care.


194 Dementia

and quantity was essential (Reilly et al., 2010). A model of Table 18.1 Lewisham Case Management Scheme: setting
care/case management that can successfully support older and role
people with dementia at home has been proposed using
Shorter-term care
eight standards of good practice that include intensive
care/case management. Four of these were related to the Community mental health team for older people
framework within which care/case management is under- Doctors, nurses, psychologist
taken: provision by specialist multidisciplinary teams; Occupational therapists, social workers
integrated arrangements for commissioning domiciliary
and respite services; joint financial arrangements facilitat- Key workers
ing health and social care provision; and organizational Longer-term care
arrangements promoting a differentiated approach to
ensure an appropriate level of response. The remaining Case managers
four were associated with the process of care/case manage- Social workers with budget
ment: a continuing targeting process to ensure a level of Service to patients with dementia
response suitable to a person’s needs; a multidisciplinary
Care management
assessment process appropriate in terms of content and
Source: Challis, D. et al., Supporting People with
timing; care planning, which supports and enhances qual-
Dementia at Home Challenges and Opportunities
ity of life; and sufficient monitoring and review to ensure
for the 21st Century, Ashgate, UK: Aldershot,
adjustments to care plans occur when required (Hughes
2009. With permission.
et al., 2005). These features remain important (Abell et al.,
2010; Challis et al., 2011, 2014a; Clarkson et al., 2011a,
2011b, 2012; Sutcliffe et al., 2010).
BOX 18.2: Practice interventions and
strategies
18.3 INTENSIVE CARE/CASE Case finding and screening
MANAGEMENT
●● Targeting of appropriate cases

The Lewisham Case Management Scheme offered a practi- Assessment


cal demonstration of care management providing commu- ●● Structured approach
nity support to older people with dementia living at home ●● Severity of need assessed
(Challis et al., 2009) and an illustration of the role of social
work within this. It was cited in the government’s National Implementing the care plan
Dementia Strategy as an examplar of intensive care/case ●● Goal defined
management preventing inappropriate care home admis- ●● Strategies of intervention and resources
sions and was one of a family of similar studies (Challis identified
and Davies, 1986; Challis et al., 1995, 2002a; Department of
Health, 2009). Specifically, it was established within a mul- Monitoring, review and case closure
tidisciplinary setting for a target population of individuals ●● Achievement of goals reassessed
with a diagnosis of dementia, identified as having unmet ●● Problems reviewed
needs and at the risk of entry to institutional care, despite ●● Outcome by domain of assessed need
input from statutory services. Its aim was to provide effective evaluated
integrated community-based long-term care spanning the
health and social service interface. The service was provided
by social services care/case managers based in a commu- and organizational. The most frequently reported were
nity mental health team for older people. Most importantly, supportive (68%), therapeutic (66%) and practical (56%).
care/case managers were able to purchase care in response Supportive goals were almost exclusively intended for the
to identified need within defined parameters. This enabled benefit of informal carers, the most frequently reported cat-
the development of a specialist paid helper service which was egories were to relieve carer burden, provide respite and assist
available to those receiving care/case management input. carers. Therapeutic interventions were most often directed
The service setting and roles are summarized in Table 18.1. at devising strategies to reduce problem behaviours associ-
Box 18.2 details the process of assessment and care/case ated with the individual’s deteriorating state. Practical goals
management within the scheme. With regard to implement- were most frequently focused on assistance with personal
ing the care plan, the goals of intervention in care/case man- care, healthcare and domestic care of the older person. These
agement were identified by retrospective analysis of 80 care demonstrated that the prime objectives of the scheme were to
plans. These were analyzed under seven categories: support- support, sustain and enhance the quality of life of the older
ive, therapeutic, practical, preventive, social, destinational person in his/her own home, and thereby to assist the carers.
Social work and care/case management in dementia 195

In the second year of the Lewisham Case Management secondary health care with the decisions made in the con-
Scheme, its effects began to show in a lower rate of admission text of care/case management (Brocklehurst et al., 1978;
to care homes for older people with dementia receiving care/ Peet et al., 1994; Sharma et al., 1994; Challis et al., 2004).
case management than for those receiving standard care. At 6 Research conducted within a specialist mental health team
months, older people receiving case management were more for older people has found that the process of assessment can
satisfied with their lifestyle at home, and there was evidence be undertaken by any of the members within it (Lindesay
of a reduction in their needs associated with activities of daily et al., 1996), and more generally, growing role substitution is
living. At 12 months, there was a significant reduction in the emerging in community mental health teams for older peo-
amount of care input and distress experienced by carers of ple (Challis et al., 2014a). More recent work has suggested
the older people in receipt of case management. other gains arising from closer integration between social
Social care provider costs were higher for those receiving care and old age mental health services (Sutcliffe et al., 2008;
care/case management, since care/case manager visits and the Clarkson et al., 2011a, 2011b; Challis and Hughes, 2012).
paid helper service were not available to those receiving stan- The experience of Australia, which has the community care
dard services. On the other hand, there was a positive gain for reforms most analogous to those in England, gives credence
the carers of those in receipt of care/case management, since to this type of arrangement (Challis et al., 1995; Howe and
financial and other costs were lower for this group compared Kung, 2003; Government of South Australia, 2009).
to the carers of those receiving standard services. Moreover, Other work in relation to social work and social care
when all costs to society were accounted for, including infor- integrated with health provision would seem to concur
mal care, the weekly cost of supporting older people with with this. Multidisciplinary assessments in the commu-
dementia and their carers was not significantly higher for nity, involving social workers and secondary healthcare
those receiving intensive care/case management, although professionals, have increased over time and are associ-
that of the main component, statutory services, was greater. ated with better care outcomes for people with cognitive
As demonstrated above, this specialist scheme, located in impairment (Clarkson et al., 2011a, 2011b; Sutcliffe et al.,
a multidisciplinary team which integrated health and social 2010). Furthermore, the use of more standardized assess-
care services, was successful in providing care to older ment processes in social work and social care improves
people with dementia to enable them to remain at home identification of people with dementia in the commu-
rather than enter long-term care and provided effective sup- nity, and potentially earlier diagnosis and access to ser-
port to their carers. This approach is consistent with other vices (Clarkson et al., 2012). It has also been suggested
UK and international evidence relating to the provision that delayed discharges of older people with cognitive
of specialist care for this group of older people (Chu et al., impairment may be reduced by the adoption of a case
2000; Eloniemi-Sulkava et al., 2001; Minkman et al., 2009). management approach (Challis et al., 2014b). However,
Moriarty and Webb (2000) noted factors that could opti- a cautionary note remains in that the case management
mize care for people with dementia and their carers. These alone is not a panacea. Effective case management cannot
included responsive care management systems, effective be delivered in isolation but depends upon access to and
monitoring systems, and home care and day care services influence over the allocation of services (Challis, 2003;
sensitive to changes in an individual’s care needs and which Challis et al., 2011; Reilly et al., 2010). It is anticipated that
provide practical support to carers. The latter is particularly the effective care of people with dementia will benefit from
important since the centrality of support for carers remains these new service configurations and arrangements which
a constant theme (Moriarty, 1999; Brodaty et al., 2005; pay less attention to traditional boundaries whether pro-
Gaugler et al., 2005; Sutcliffe et al., 2013). National and inter- fessional or organizational than hitherto.
national policy has endorsed this approach (AHMC, 2006;
Department of Health, 2009; Republique Francaise, 2011a,
b) although a recent report has suggested that care/case
management could substantially reduce health and social
18.5 CONCLUSIONS
care costs but increase the costs of unpaid care (Knapp et al.,
2014). The emergence of dementia specific services may lead to
further interprofessional role blurring which, if managed
correctly, could be the basis of improved care for people
with dementia and their carers. Within this, social work
18.4 SOCIAL WORK IN INTEGRATED and care/case management have a distinct role in rela-
CARE SETTINGS tion to the provision of long-term community-based care.
Developments in England over the last 25 years provide evi-
Within multidiscplinary teams, arrangements need to be in dence of a change in the role, focus and setting for social
place to facilitate the multidisciplinary assessment of people workers and other professions working with people with
with dementia and particularly to ensure that specialist cli- dementia with an increasing emphasis on care/case man-
nicians contribute to this process. A number of UK s­ tudies agement as a means of coordinating complex packages of
have suggested potential gains from greater integration of care as an alternative to care home placement (SSI/SWSG,
196 Dementia

1991; Department of Health, 2001; 2009). Care coordination Available at http://www.dcnet.gr.jp/retrieve/kaigai/


features strongly in official guidance with service commis- pdf/au09_02NationalFramework.pdf. Accessed 7 June
sioners advised to work with local dementia partnerships to 2016.
agree and implement a robust service model for care coor- Brodaty, H., Thomson, C., Thompson, C. and Fine, M.
dination (NICE, 2013). (2005). Why caregivers of people with dementia and
While a changed role for social workers has emerged with memory loss don’t use services. International Journal
the introduction of personal budgets for social care services of Geriatric Psychiatry, 20: 537–546.
(cash-for-care), permitting older people and their carers to Brodaty, H. and Cumming, A. (2010). Dementia services in
organize and manage their own care (Department of Health, Australia. International Journal of Geriatric Psychiatry,
2008; Department of Health (Australia), 2012), social work- 25: 887–895.
ers will still be required to undertake care coordination for Brocklehurst, J., Carty, M., Leeming, J. and Robinson,
the most frail elders. Indeed, there is evidence that greater J. (1978). Care of the elderly: Medical screening of
flexibilities and personalized care can be provided by care/ old people accepted for residential care. Lancet, 2:
case managers with budgets (Challis and Davies, 1986; 141–142.
Challis et al., 1995, 2002a; Sutcliffe et al., 2012). Concurrently Cahill, S. (2010). Developing a national dementia strategy
there has been an increase in both community-based multi- for Ireland. International Journal of Geriatric Psychiatry,
disciplinary teams for older people with mental health prob- 25: 912–916.
lems and those with social workers as core members (Challis Challis, D. (1993). Alternatives to institutional care. In
et al., 2002b, 2014a; Tucker et al., 2009). R. Levy, R. Howard and A. Burns (eds.), Treatment
For social work, there are two challenges consequent on and Care in Old Age Psychiatry. Petersfield, England:
these changes which have an international resonance. First, Wrightson.
the profession-specific contribution to the care of older peo- Challis, D. (2003). Achieving co-ordinated and integrated
ple with dementia and their carers has changed primarily to care among long term care services: The role of care
that of responsibility for the care for people living at home management. In J. Brodsky, J. Habib and M. Hirschfeld
and, by virtue of changes to long-term care funding, gate- (eds.), Key Policy Issues in Long Term Care. Geneva,
keepers for admission to residential or nursing home care. Switzerland: World Health Organization.
Second, and in antithesis to this clarity, the social work role Challis, D. and Davies, B. (1986). Case Management in
in respect of the care of people with dementia has become Community Care. Aldershot, United Kingdom: Gower.
less profession specific. Within multidisciplinary teams, Challis, D. and Hughes, J. (2012). Systems of care for older
there are more opportunities for the sharing of tasks offer- people – Case management. In M. Gosney, A. Harper
ing informal small-scale opportunities for service substi- and S. Conroy (eds.), Oxford Desk Reference: Geriatric
tution. There is thus a tendency for less specificity of role Medicine. Oxford, United Kingdom: Oxford University
for staff as services become more community based. This Press, pp. 394–395.
leads unsurprisingly to a degree of insecurity among pro- Challis, D., Darton, R., Johnson, L. et al. (1995). Care
fessional groups, because each derives their unique qual- Management and Health Care of Older People: The
ity from a training and occupational function. Roles come Darlington Community Care Project. Aldershot, United
to be negotiated rather than simply professionally defined Kingdom: Arena.
within groups to a greater extent in community-based ser- Challis, D., Chesterman, J., Luckett, R. et al. (2002a).
vices and therefore may be seen as less clearly delineated by Care Management in Social and Primary Health Care:
professional origin. This process may well occur to a greater The Gateshead Community Care Scheme. Aldershot,
extent as the process of closer integration between health United Kingdom: Ashgate.
and social care develops. Challis,D., Reilly, S., Hughes, J. et al. (2002b). Policy,
organisation and practice of specialist old age psy-
REFERENCES chiatry in England. International Journal of Geriatric
Psychiatry, 17: 315–325.
Abell, J., Hughes, J., Reilly, S. et al. (2010). Case manage- Challis, D., Clarkson, P., Williamson, J. et al. (2004). The value
ment for long-term conditions: Developing targeting of specialist clinical assessment of older people prior to
processes. Care Management Journals, 11: 11–18. entry to care homes. Age and Ageing, 33: 25–34.
Applebaum, R. and Austin, C. (1990). Long Term Care Challis, D., Sutcliffe, C., Hughes, J. et al. (2009).
Case Management, Design and Evaluation. New York, Supporting People with Dementia at Home Challenges
NY: Springer. and Opportunities for the 21st Century. Aldershot,
Arai, Y., Arai, A. and Mizuno, Y. (2010). The national United Kingdom: Ashgate.
dementia strategy in Japan. International Journal of Challis, D., Hughes, J., Berzins, K. et al. (2011).
Geriatric Psychiatry, 25: 896–899. Implementation of case management in long-term
Australian Health Ministers Conference (AHMC) (2006). conditions in England: Survey and case studies.
National Framework for Action on Dementia: Journal of Health Services Research and Policy, 18
2006–2010. Sydney, Australia: Department of Health. (Suppl. 1): S8–S13.
Social work and care/case management in dementia 197

Challis, D., Tucker, S., Wilberforce, M. et al. (2014a). Eggert, G.M., Friedman, B. and Zimmer, J.G. (1990).
National trends and local delivery in old age mental Models of intensive case management. In L. Reif and
health services: Towards an evidence base. A mixed- B. Trager (eds.), Health Care of the Aged: Needs,
methodology study of the balance of care approach, Policies and Services. New York: The Haworth Press.
community mental health teams and specialist Eloniemi-Sulkava, U., Notkola, I.L., Hentinen, M. et al. (2001).
mental health outreach to care homes. Programme Effects of supporting community-living demented
Grants for Applied Research, 2 (4): 1–480. patients and their caregivers: A randomized trial. Journal
Challis, D., Hughes, J., Xie, C. and Jolley, D. (2014b). An of the American Geriatrics Society, 49: 1282–1287.
examination of factors influencing delayed discharge Engedal, K. (2010). The Norwegian dementia plan 2015 –
of older people from hospital. International Journal of ‘Making most out of the good days’. International
Geriatric Psychiatry, 29: 160–168. Journal of Geriatric Psychiatry, 25: 928–930.
Chester, H., Hughes, J., Sutcliffe, C. et al. (2015). Exploring Gaugler, J.E., Kane, R.L., Kane, R.A. and Newcomer, R.
patterns of care coordination within services for older (2005). Early community-based service utilization and
people. International Journal of Care Coordination, 18: its effects on institutionalization in dementia caregiv-
5–17. ing. The Gerontologist, 45: 177–185.
Chu, P., Edwards, J., Levin, R. and Thomson, J. (2000). Government of South Australia (2009). Health Service
The use of clinical case management for early stage Framework for Older People 2009–2016: Improving
Alzheimer’ patients and their families. American Health and Wellbeing Together. Adelaide, South
Journal of Alzheimer’s Disease and Other Dementias, Australia: Department of Health, South Australia.
15: 284–290. Guisset-Martinez, M. (2012). Three Alzheimer plans
Clarkson, P., Brand, C., Hughes, J. and Challis, D. (2011a). in France (2008–2012). In A. Innes, F. Kelly and L.
Integrating assessments of older people: Examining McCabe (eds.), Key Issues in Evolving Dementia
evidence and impact from a randomised controlled Care: International Theory-based Policy and Practice.
trial. Age and Ageing, 40: 388–391. London: Jessica Kingsley, pp. 122–136.
Clarkson, P., Hughes, J., Abendstern, M. et al. (2011b). Howe, A. and Kung, F. (2003). Does assessment make a
Involving specialist clinicians in policies for integrated difference for people with dementia? The effective-
care. Journal of Integrated Care, 19: 14–22. ness of the aged care assessment teams. International
Clarkson, P., Abendstern, M., Sutcliffe, C. et al. (2012). The Journal of Geriatric Psychiatry, 18: 205–210.
identification of dementia and its correlates in a social Hughes, J., Sutcliffe, C. and Challis, D. (2005). Social work.
services setting: Impact of a national study in England. In A. Burns (ed.), Standards in Dementia Care. London:
Dementia, 11: 617–632. Taylor and Francis.
Department of Health (2001). The National Service Hunter, S., Brace, S. and Buckley, G. (1990). The interdisci-
Framework for Older People. London: Department of plinary assessment of older people at entry into long-
Health. term institutional care: Lessons for the new community
Department of Health (2008). Transforming Adult Social care arrangements. Research Policy and Planning, 11:
Care, LAC (DH) (2008)1. London: Department of 2–9.
Health. Knapp, M., Comas-Herrera, A., Wittenberg, R. et al.
Department of Health (2009). Living Well with Dementia: (2014). Scenarios of Dementia Care: What Are the
A National Dementia Strategy. London: Department of Impacts on Cost and Quality of Life?. London: Personal
Health. Social Services Research Unit, London School of
Department of Health (2013). The Prime Minister’s Economics and Political Science.
Challenge on Dementia. Delivering Major Lindesay, J., Herzberg, J., Collighan, G. et al. (1996).
Improvements in Dementia Care and Research by Treatment decisions following assessment by multidis-
2015: Annual Report of Progress. London: Department ciplinary psychogeriatric teams. Psychiatric Bulletin, 20:
of Health. 78–81.
Department of Health (Australia) (2012). Living Longer Minkman, M., Ligthart, S. and Huijsman, R. (2009).
Living Better. Canberra, Australia: Department of Integrated dementia care in the Netherlands: A mul-
Health, Government of Australia. tiple case study of case management programmes.
Department of Health, Social Services and Public Safety Health and Social Care in the Community, 17: 485–494.
(2011). Improving Dementia Services in Northern Ministry of Health Welfare and Sports (2009). Guideline
Ireland: A Regional Strategy. Available at http://www. for Integrated Dementia Care [excerpt] An Aid for
dhsspsni.gov.uk/improving-dementia-services-in- the Development of Integrated Dementia Care. The
northern-ireland-a-regional-strategy-november-2011. Hague, Netherlands: Ministry of Health Welfare and
pdf. Accessed 15 June 2015. Sports.
198 Dementia

Moriarty, J.M. (1999). Use of community and long-term Schultz, E.M. and McDonald, K.M. (2014). What is
care by people with dementia in the UK: A review of care coordination? International Journal of Care
some issues in service provision and carer and user Coordination, 17: 5–24.
preferences. Aging and Mental Health, 3: 311–319. Sharma, S., Aldous, J. and Robinson, M. (1994). Assessing
Moriarty, J. and Webb, S. (2000). Part of Their Lives: applicants for part 3 accommodation: Is a formal
Community Care for Older People with Dementia. clinical assessment worthwhile? Public Health, 108 (2):
Bristol, England: The Policy Press. 91–97.
National Institute for Health and Care Excellence (NICE) Social Services Inspectorate/Social Work Services Group
(2013). Support for Commissioning Dementia Care, (SSI/SWSG) (1991). Care Management and Assessment:
NICE commissioning guides CMG48. London: Managers Guide. London: HMSO.
National Institute for Health and Care Excellence. Sutcliffe, C., Hughes, J., Abendstern, M. et al. (2008).
Available at www.nice.org.uk/guidance/cmg48. Developing multidisciplinary assessment – Exploring
Accessed 15 June 2015. the evidence from a social care perspective.
Norwegian Ministry of Health and Care Services (2007). International Journal of Geriatric Psychiatry, 23:
Dementia Plan 2015, Making the Most of Good Days, 1297–1305.
Subplan of Care Plan 2015. Oslo, Norway: Norwegian Sutcliffe, C., Hughes, J., Chester, H. et al. (2010).
Government Administration Services Distribution Changing patterns of care coordination within old-age
Services. services in England. Care Management Journals, 11 (3):
Peet, S., Castleden, C., Potter, J. and Jagger, C. (1994). 157–169.
The outcome of a medical examination for applicants Sutcliffe, C., Hughes, J., Xie, C. et al. (2012). Social care
to Leicestershire homes for older people. Age and in older people’s services – Facilitating the flexible
Ageing, 23: 65–68. use of resources. Care Management Journals, 13:
Reilly, S., Hughes, J. and Challis, D. (2010). Case manage- 100–107.
ment for long-term conditions: Implementation and Sutcliffe, C.L., Roe, B., Jasper, R. et al. (2013). People
processes. Ageing and Society, 30: 125–155. with dementia and carers’ experiences of dementia
Reilly, S., Miranda-Castillo, C., Malouf, R. et al. (2015). care and services: Outcomes of a focus group study.
Case Management Approaches to Home Support for Dementia, 14 (6): 769–787.
People with Dementia (Review). Cochrane Database of The Scottish Government (2013). Scotland’s Dementia
Systematic Reviews. Chichester, United Kingdom: John Strategy 2013–2016. Available at http://www.gov.
Wiley & Sons. scot/Resource/0042/00423472.pdf. Accessed 15 June
Republique Francaise (2011a). National French plan 2015.
for “Alzheimer and related diseases” 2008–2012. Tucker, S., Baldwin, R., Hughes, J. et al. (2009). Integrating
Available at http://www.plan-alzheimer.gouv.fr/IMG/ mental health services for older people in England –
pdf/. Accessed 15 June 2015. From rhetoric to reality. Journal of Interprofessional
Republique Francaise (2011b). National French plan for Care, 23: 341–354.
“Alzheimer and related diseases” 2008–2012. Available Welsh Assembly Government. (2011). National Dementia
at http://www.plan-alzheimer.gouv.fr/IMG/pdf/A_ Vision for Wales. Available at http://gov.wales/docs/
short_summary_of_Plan_Alzheimer_2008-2012_as_ dhss/publications/110302dementiaen.pdf. Accessed
of_30_june_2011.pdf. Accessed 15 June 2015. 15 June 2015.
19
Occupational therapy in dementia care

ALISSA WESTPHAL

19.1 INTRODUCTION 19.2 OCCUPATIONAL THERAPY ROLE

Occupational therapy (OT) is concerned with maximiz- The role of OT in caring for people with dementia has
ing a person’s engagement in meaningful and required received growing research interest over the past decade as
occupations and roles. The profession is built on the phil- part of the shift to provide evidence based practice. The
osophical foundation that engaging in meaningful and OT’s role varies according to the level of experience and
purposeful occupations and roles is essential for main- training of the OT (McGrath and O’Callaghan, 2014), the
taining health and well-being (American Occupational specific care setting, presenting issues, severity of the per-
Therapy Association [AOTA], 2008). The OT clinical son’s dementia and the cultural context (Letts et al., 2011).
model of practice recognizes occupational function- Roles of OT can include
ing as arising from a systemic and dynamic relationship
between the person, the physical, social and cultural ●● Contributing to diagnostic formulation, including early
environment and the occupation (Law et al., 1996). This identification of cognitive impairment and disease
relationship between the person, environment and occu- progression.
pation is often disrupted by physical and psychiatric con- ●● Facilitating recovery and/or maintenance of skills,
ditions. Occupational therapists (OTs) use rehabilitative e.g. physical functioning, developing compensatory
and adaptive approaches to address these occupational memory strategies.
performance issues and maximize functioning (Fisher, ●● Maximizing the person’s independence in everyday
2006a; Hopkins and Smith, 1993). activities through modifications of the activity, environ-
Dementia causes skill loss, resulting in deteriorations ment, communication and approach and use of adaptive
in occupational functioning. The disease trajectory, whilst equipment where required.
progressive, differs for each person and is frequently ●● Maintaining the person’s sense of self-worth and support-
accompanied by additional functional difficulties caused ing their identity through facilitating their engagement in
by age-related physical conditions and behavioural and meaningful, necessary and interesting activities and roles.
psychological symptoms of dementia (BPSD) (see Chapters ●● Recommending supports which allow the person to
8, 9 and 16). Those with dementia are commonly supported remain in their preferred care environment or alternate
by caregivers who themselves experience difficulty dur- accommodation, when required.
ing the progression of the disease. The OT clinical model ●● Minimizing risk and maximizing safety of the person’s
provides a biopsychosocial–occupational–­environmental occupational performance within their environment,
framework, offering OTs a significant and unique role in e.g. driving capacity.
holistic response to the complex and progressive occu- ●● Addressing BPSD using non-pharmacological approaches.
pational dysfunction, which is unique to the experience ●● Developing and reviewing activity programmes and
of each person with dementia and their caregivers (Letts environments which support the needs of people with
et al., 2003). dementia in a group or residential setting.

199
200 Dementia

●● Supporting and building the capacities of caregivers and medication management, home and money
services to effectively care for people with dementia. management, leisure, telephone use, driving and
●● Developing evidence to inform and advance clinical transport, work and technology use
practice. b. Other activities: Work, sleep and rest, social engage-
●● Contributing to the development of policy in areas of ment and leisure
health and ageing. c. Task issues: Demands, complexity, sequencing,
environmental context, cues and familiarity
The OT process involves assessment, treatment plan- d. Risk issues: Risks to the person with dementia,
ning, implementation and evaluation. Best practice advo- caregiver and others associated with occupational
cates the engagement of both the person with dementia and dysfunction
their caregiver(s) throughout this process (Strong, 2003). 4. The caregiver
a. Knowledge: Understanding of the person they are
caring for and dementia related changes
b. Caring role: Relationship, difficulties, burden,
19.3 ASSESSMENT changes, motivation, concerns and readiness to
engage and adopt interventions
19.3.1 PURPOSE AND PROCESS OF THE c. Communication and approach
OT ASSESSMENT 5. Behaviour and occupational performance
a. The person with dementia: Occupational engage-
The goal of OT assessment is to gain a comprehensive ment and BPSD (incidence, triggers and responses)
understanding of the abilities, strengths and deficits of the b. The caregiver: Role engagement
patient and their caregivers and the available resources to
do so (Letts et al., 2003). Completion of OT assessments can OT assessment facilitates include identification of the
provide specific information in the following areas: person and caregivers’ needs and intervention priorities.
Whilst, overlapping at times, the information gathered
1. The person complements that obtained by other professional disciplines
a. Cognitive function: Memory acquisition, storage providing a unique picture of the impact of dementia on the
and retrieval, learning, orientation, judgement, person’s activities of daily living (ADL) and occupational
insight, decision making, planning, concentration, performance (Fossey, 2001).
task shifting, reasoning, problem solving, mental The focus of assessment changes with disease progres-
flexibility, emotional and behaviour regulation sion, reflecting the changing needs of the person and care-
b. Perceptual and sensory function: Visual, auditory, giver (Zgola, 1999). In the early stages of dementia, the
proprioceptive sensation, stereognosis, praxis, OT’s role is focussed on maintenance of ADL performance
agnosia, illusions and misidentification within the home. With disease progression, the focus
c. Physical functioning: Movement, balance, mobil- typically shifts to maximizing and providing meaningful
ity and transfers, hand function, coordination and engagement and facilitating appropriate care and support.
impact of illness on the patient OT assessment information is collected through inter-
d. Communication skills: Comprehension, verbal and views, observation and standardized assessments. No sin-
non-verbal social skills, receptive and expressive gle assessment measures all areas of interest. Instead, the
dysphasia and adaptive communication OT chooses the most appropriate tool. Understanding the
e. Psychological state: Mood, anxiety, past trauma and present issues and the person’s background, including their
motivation level of education, premorbid functioning and roles and
f. Past and current occupational preferences: Interests, cultural history is necessary to facilitate informed selection
roles, habits, routines and meaningful activities and interpretation of assessment tools. Finally, developing a
2. The environment trusting and supportive relationship, adapting communica-
a. Physical, social, temporal, cultural and spiritual tion and approach, focussing on abilities and engaging the
characteristics: Contrasts, temperature, accessibil- caregiver, all assist with the assessment process and maxi-
ity, level of sensory stimulation, lighting, design, mize the accuracy of the results.
furnishings, cues, personalization, language use,
values, expectations, supports, knowledge, toler- 19.3.1.1 Interview
ance and flexibility
3. Occupational functioning Interviews provide a foundation for identification of
a. Personal, domestic and community activities of daily areas requiring assessment, allowing the OT to gain an
living (Personal Activities of Daily Living [PADLs], understanding of the life story of the person with demen-
Domestic Activities of Daily Living [DADLs] and tia. Interviews may be conducted with the person suffer-
Community Activities of Daily Living [CADLs]): ing from dementia, their caregivers, family members and
Eating, dressing, grooming, bathing, toileting, other relevant persons. Caregivers and family members are
Occupational therapy in dementia care 201

frequently interviewed in order to obtain a collateral history ●● ADL and leisure functioning: Assessment of motor and
and understand their views on the person’s functioning. process skills (Fisher, 2006b); perceive, recall, plan and
Building rapport, basic counselling skills, use of interpret- perform system of task analysis (Chapparo and Ranka,
ers, adapting communication and language use in response 2006); interview for deterioration in daily activities in
to the caregiver’s level of education and communication dementia (Teunisse and Derix, 1997); Barthel index
deficits are common approaches used during interviews to (Mahoney and Barthel, 1965); functional assessment
assist in developing therapeutic relationships and obtaining measure (Hall, 1997); functional independence mea-
information. Interviews may be used instead of observation sure (Guide from the Uniform Data Set for Medical
or standardized assessments when time or funding prohibit Rehabilitation, 1993); structured assessment of indepen-
more comprehensive assessment. dent living skills (Mahurin et al., 1991); activities of daily
living questionnaire (Johnson et al., 2004); daily activities
19.3.1.2 Observations questionnaire (Oakley et al., 1991); kitchen task assess-
ment (Baum and Edwards, 1993); Lawton instrumental
The functioning of a person suffering from dementia may activities of daily living scale (Lawton and Brody, 1969);
be observed when participating in work, daily living and Bristol activities of daily living scale (Bucks et al., 1996);
leisure activities that are relevant to them. Observational functional activities questionnaire (Pfeffer et al., 1992);
assessments provide a flexible approach to identifying Canadian occupational performance measure (Law et al.,
functional performance issues (Alexander, 1994) and may 1991); Cleveland scale of ADLs (Patterson et al., 1992);
be completed at the pace and manner in which the person pool activity level instrument (Pool, 2008) and assess-
is capable or accustomed. Where possible, observations of ment of living skills and resources (Williams et al., 1991).
functional performance are completed within the environ- ●● Environmental and quality of care assessments: Safety
mental contexts familiar to the person, often during home assessment scale (de Poulin et al., 2006); dementia care
visits. This aims to minimize the patient’s anxiety, their mapping (Bradford Dementia Group, 1997); home envi-
need to learn new skills and maximize their orientation. ronment protocol assessment (Gitlin et al., 2002); safety
assessment of function and environment for rehabili-
19.3.1.3 Standardized tests tation (Oliver et al., 1993) and in-home occupational
performance evaluation (Stark et al., 2010).
Assessment tools are often used by occupational therapists ●● Engagement: Expanded group activity form
in addition to the aforementioned methods to (Thorgrimsen et al., 2002: expanded from Bender et al.,
1987); pool activity level instrument (Pool, 2008); activ-
●● Gain an objective and measurable picture of a person’s ity in time and context (Wood et al., 2005) and positive
performance in particular areas, including BPSD. response schedule for severe dementia (Perrin, 1997).
●● Assess the severity of cognitive or occupational dys- ●● Carer assessments: Zarit carer burden interview (Zarit
function compared to a normative group. et al., 1980); perceived change index (Gitlin et al., 2006);
●● Establish a baseline measure of functioning against task management strategy index (Gitlin et al., 2002);
which subsequent assessments may be compared, readiness to change (Gitlin and Corcoran, 2005) and
e.g. quantifying change or efficacy of interventions. caregiver assessment of management problems (Gitlin
and Corcoran, 2005).
A wide range of tools exists for use of practitioners for
people who have dementia. They can be divided into cogni-
tive, functional performance, environmental, risk, quality
of life, caregiver and behavioural assessments. A selection of 19.4 INTERVENTION
assessments commonly reported in the occupational ther-
apy literature includes the following: OT interventions focus on maximizing functioning, pri-
marily using adaptive or compensatory rather than rehabil-
●● Cognitive and perceptual skill assessments: Mini-mental itative approaches. Interventions are individually tailored,
state examination (Folstein et al., 1975); hierarchical reflecting the differences in the presenting issues and goals,
dementia scale (Cole and Dastoor, 1996); Lowenstein the stage of the person’s dementia, the need and abilities of
occupational therapy cognitive assessment for geri- the caregiver and the results of the completed assessment
atric population (Elazer et al., 1996); Allen cogni- (Graff et al., 2006). Ongoing review of OT interventions is
tive level screen (Allen et al., 2007); Allen diagnostic required to ensure they best address the issues being expe-
module placemat (Earhart, 2006); revised memory and rienced by patients and needs of the person with dementia
behaviour problems checklist (Teri et al., 1992); execu- and their caregivers. Evidence supporting OT interven-
tive function performance test (Baum et al., 2008); tions with the person and their caregivers is growing, with
Middlesex elderly assessment of mental state (Golding many practices still guided by good clinical care principles
and Cognitie, 1989) and Addenbrooke’s cognitive (Jensen and Padilla, 2011; Letts et al., 2011; Padilla, 2011a,
examination – revised (Mioshi et al., 2006). 2011b; Thinnes and Padilla, 2011).
202 Dementia

19.4.1 THE USE OF ACTIVITIES ●● Experience and variety (Hellen, 1998; Truscott, 2004):
Activities provide opportunities for sharing experi-
Activity interventions aim to preserve the person’s dignity ences, reminiscing, building friendship, giving pleasure,
and self-esteem by successfully engaging them in meaning- allow emotional expression and creativity. The focus of
ful, required and purposeful activity that is congruent with activities shifts from the product or outcome to the pro-
their occupational history, interests, roles and motivations. cess of doing. Variety of activity opportunities remains
Knowledge about the person, including their retained abili- important.
ties, allows the OT to modify and provide activities that best ●● Physical activity (Thuné-Boyle et al., 2012): Access to
fit their skills, throughout the disease trajectory. General indoor and outdoor opportunities for physical activity
principles for activity interventions include the following: can assist in maintaining physical and mental function-
ing, along with managing BPSD.
●● Familiar and novel activities (Perrin et al., 2008): ●● Individualized (Padilla, 2011a; Robert Dooley and
Familiar activities provide continuity and familiarity Hinojosa, 2004): Caregivers can be engaged in providing
for the person with dementia, allowing them to partic- activities congruent with the person’s life story and tai-
ipate using well-learned skills. Activities can be used lored to their interests, needs and abilities. Individualizing
to provide role continuity and reinforce identity in the activities can be cost effective, pleasurable for the person
person, for example, a doll or child representational and assist in reducing BPSD and in addition, benefit
therapy, pet therapy, domestic chores and men’s sheds the caregiver by reducing the time providing care and
are helpful. Role-based activities become increasingly improving their sense of confidence in their role (Graff
important, as many roles are removed with the pro- et al., 2006; Gitlin et al., 2008, 2009, 2010).
gression of dementia in the patient. On the other hand,
novel activities have fewer preconceived expectations 19.4.2 ENVIRONMENT ADAPTATIONS
of performance potentially providing failure free
experiences. Robotic technologies (e.g. Paro) are an Environmental adaptations involve changes to the physi-
example of novel activities that can increase interac- cal, social and organizational/institutional environments
tion, interest, pleasure and quality of life (Roger et al., of the person with dementia and their carers. Modifying
2012; Klein et al., 2013; Moyle et al., 2013; Robinson environments can maximize the person’s occupational per-
et al., 2013). formance by providing accessible opportunities for engage-
●● Simplifying activity demands and structure (Hellen, ment, compensating for skill deficits, utilizing strengths of
1998; Padilla, 2011b): OT’s grade activities according the patient, minimizing risk and managing BPSD (Cooper
to the skills and abilities of the person. Reducing the and Day, 2003; Brawley, 2006; Padilla, 2011b).
number of steps in an activity, providing assistance only Greater evidence exists for environmental interventions
where needed and limiting choices to match the per- in long-term care settings (Fleming and Purandare, 2010).
son’s abilities, all reduce the task demand and support Despite this, many of the principles of environmental inter-
successful occupational performance. Simplification ventions are applicable across other care settings, including the
of tasks by caregivers has demonstrated improvements homes and throughout the disease trajectory. Environmental
in the person’s functioning and BPSD, decreasing the intervention principles include the following:
number of upset and burdened caregivers, as well as
improved effect on patients (Gitlin et al., 2003, 2005). ●● Promoting comfort (Calkins, 2005): Preferred tempera-
Where OT skill is unavailable, Montessori activities ture, familiar furnishings and objects, appropriate
which offer a graded activity approach may be used to pressure care, preferred size of objects and familiar
increase engagement and reduce some BPSD (Camp, caregivers provide comfort.
2006; Lin et al., 2009; van der Ploeg et al., 2013). Care ●● Optimal sensory stimulation (Calkins, 2004, 2005):
should be taken to ensure the activity remains mean- Sensory stimulation can be altered to meet the needs of
ingful for the person and not simply utilized to exercise the person by changing the location or level of sensory
cognitive, perceptual and motor skills and to occupy stimulation, e.g. reducing noise in the environment.
time. ●● Contrasts and lighting (Brawley, 2006): Maximizing
●● Temporal adjustments (Pool, 2008): Allow the person contrast between items can optimize orientation to
to set the pace and provide activities within a familiar and awareness of objects. Conversely, minimizing
routine. When this is not possible, due to time limita- contrasts can be used to camouflage or reduce cueing.
tions, priority should be given to those which are most Glare, shadows, poor lighting levels and sudden lighting
important to the person. changes should be addressed.
●● Engage multiple senses (Pool, 2008): Activities that ●● Cueing, shaping and compensating for occupational
engage the senses become increasingly important with performance issues (Cooper and Day, 2003; Brawley,
disease progression, affording opportunities for engage- 2006; Marcy-Edwards et al., 2005; Padilla, 2011b):
ment to those with severely impaired cognitive and OTs prescribe and recommend adaptive equipment
communication skills. to optimize the person’s functioning, address safety
Occupational therapy in dementia care 203

concerns and reduce caregiver burden (Gitlin and Chee, Thinnes and Padilla, 2011). This process minimizes excess
2006; Graff et al., 2006). These include shower stools, disability for the person by assisting caregivers often through
grab rails, non-slip mats, hand held showers, com- role play or modelling, to learn skills and generate strategies
mode chairs, contrasting toilet seats, modified clothing, effectively matching their communication, approach and
large calendars, talking clocks, modified telephones assistance to the person’s abilities. Finally, OTs assist in min-
(with large photo buttons), contrasting crockery and imizing disruption to the caregiver’s occupational identity
napery, modified cutlery, specialized seating, medica- by encouraging use of support services and engagement in
tion dispensing packs, personal alert alarms, isolation meaningful activities (Hasselkus and Murray, 2007).
switches for ovens and stoves, stove dial covers, smoke
and gas detectors, signs and labels. Structured routines, 19.4.4 MANAGING BPSD
development of activity specific areas, task-related items
placed in the task environment, furniture placement, The emergent paradigms for understanding and treating
placement of photos and objects personally familiar to BPSD view these symptoms as arising from unmet needs,
the person may be used to cue and support the person’s difficulties coping with stress, reinforced learning and
engagement. Objects may be removed or disguised to changes in pathology (O’Connor et al., 2009; see Chapters 8
be as unobtrusive as possible. This can reduce cueing or and 9). The OT model of practice provides scope for under-
allow use of objects where engagement is not desired or standing the multifactorial and changing aetiology of BPSD
is unsafe. Finally, environments should make sense, be and intervening in a patient’s care in a caregiver-centred
visually accessible, contain familiar aspects and provide manner. Through this model, OTs view BPSD as arising
clear cues for what is expected of the person (Fleming from poor fit or mismatch between the environment and/or
and Purandare, 2010). occupation and skills and attributes to the person, many of
●● Simplification (Calkins, 2004): Reducing clutter and dis- which are listed in Section 19.3.1. Short-term OT interven-
tractions can minimize the complexity of the environ- tions modifying the aspects of the activity and environment
ment, reducing the demands placed on the person. and building caregiver capacities have demonstrated ben-
●● Privacy (Brawley, 2006): Provision of privacy during efits for the person and caregivers (Gitlin et al., 2001, 2003,
PADLs and other activities remains crucial to caring for 2005, 2008, 2009, 2010; Graff et al., 2003, 2006, 2007). But,
the person with dementia. OTs require training in this area to maximize intervention
outcomes (Voigt-Radloff et al., 2009, 2011). OT interven-
tions focus on identifying triggers and optimizing fit and
19.4.3 WORKING WITH FAMILIES use many of the principles and approaches outlined earlier.
AND CARERS Additional principles include the following:

Caregivers have a crucial role in facilitating the person’s ●● Utilization of the BPSD: Some behavioural symptoms
occupational performance. This role changes with disease can be effectively utilized. Pacing may be provided with a
progression in the patient. Interventions with caregivers purpose through engaging the person in physical activi-
aim to improve their confidence and role performance and ties such as sweeping, going for a walk outside or cleaning
reduce burden by windows. An apron customized to include multi-sensory
occupational opportunities may be used to reduce cloth-
●● Building on the caregiver style of caring and increas- ing removal due to ‘fiddling’ with shirt buttons where
ing their willingness to utilize interventions (Corcoran, discomfort is not the triggering issue. Finally, where the
2011; Gitlin and Corcoran, 2005). BPSD can be understood in the context of the person’s
●● Building caregiver capacities to understand, care for previous occupational identity, jobs may be provided that
and support continued engagement of the person with addresses this need. For example, tactile floor tiles may
dementia in meaningful activities. be laid out for the person who previously was a tiler and
●● Supporting caregiver to attend to their occupational now crawls around feeling the floor surface.
roles and needs. ●● Refocussing attention: Providing a person with an
alternate purposeful activity that is not reliant on
Elements of the OT intervention process with caregivers cognitive processing, or placing them in an alternate
include flexibility, affording choice, making small changes environment, can assist in distracting and shifting their
gradually, providing education and information that is rel- behaviour from the BPSD.
evant and timely, encouraging caregiver breaks, drawing on ●● Environmental placement: Environments influence and
their experiences as an expert on the person with dementia, often engender particular behaviours (Law et al., 1996).
validating their experience, readjusting interventions based For example, libraries, churches and playschools are
on the caregiver’s feedback and both written and multi-­ areas associated with quieter activity. Environments can
sensory learning to communicate the recommended inter- be manipulated to promote or cue alternate behaviours.
ventions (Josephsson et al., 2000; Graff et al., 2003; Gitlin In some cases, positioning in the environment can cue
and Corcoran, 2005; Egan et al., 2006; Letts et al., 2011; BPSD. For example, for a person who has difficulty
204 Dementia

sitting long enough to consume their meal, distracting Bucks, R.S., Ashworth, D.L., Wilcock, G.K. and Siegfried,
them away from changes in visual stimuli (people walk- K. (1996). Assessment of activities of daily living in
ing by) can assist in reducing cues that encourage them dementia: Development of the Bristol activities of daily
to get up and walk away. living scale. Age and Ageing, 25: 113–120.
Calkins, M.P. (2004). Articulating environmental press in
Thorough assessment is always required when faced with environments for people with dementia. Alzheimer’s
BPSD. Reversible causes should always be treated and inter- Care Quarterly, 5: 165–172.
ventions should be individualized, risk addressed and the Calkins, M.P. (2005). Environments for late-stage demen-
caregiver supported in understanding, responding and cop- tia. Alzheimer’s Disease Quarterly, 6: 71–75.
ing with the BPSD. Finally, OTs should regularly review Camp, C.J. (2006). Montessori-Based Activities for Persons
interventions to ensure the current trends and address the with Dementia, Vol. 2. Beachwood, OH: Menorah Park
issues of concern. Center for Senior Living.
Chapparo, C. and Ranka, J. (2006). The PRPP System of
Task Analysis: Users Training Manual, Research Edition.
Sydney, Australia: OP Network.
19.5 CONCLUSION Cole, M.G. and Dastoor, D.P. (1996). The hierarchic
dementia scale: Conceptualisation. International
Whilst the specific role of the OT differs across settings Psychogeriatrics, 8: 205–212.
and is based on the severity of the person’s dementia and Corcoran, M.A. (2011). Caregiving styles: A cognitive
surrounding issues, occupational dysfunction remains a and behavioral typology associated with dementia
common theme. OTs are skilled in assessing and provid- family caregiving. The Gerontologist, 51: 463–472.
ing unique individualized and responsive interventions that doi:10.1093/geront/gnr002
address performance issues and BPSD experienced by the Earhart, C.A. (2006). Allen Diagnostic Modules: Manual,
person suffering from dementia. Addressing these issues Second Edition. Colchester, CT: S&S Worldwide.Egan,
assists in optimizing functioning and maximizing the qual- M., Hobson, S. and Fearing, V.G. (2006). Dementia and
ity of life of the patient and their caregiver. occupation: A review of the literature. The Canadian
Journal of Occupational Therapy, 73: 132–140.
REFERENCES Elazer, B., Itzkovitch, M. and Katz, N. (1996). LOTCA-G
Manual. Pequannock, NJ: Maddak.
Alexander, K. (1994). Occupational therapy. In E. Chiu and Fisher, A.G. (2006a). Assessment of Motor and Process
D. Ames (eds.), Functional Psychiatric Disorders of the Skills. Volume 1: Development, Standardisation and
Elderly. Cambridge, UK: Cambridge University Press, Administration Manual, Sixth Edition. Fort Collins, CO:
pp. 522–543. Three Star Press.
Allen, C.K., Austin, S.L., David, S.K. et al. (2007). Manual Fisher, A.G. (2006b). Assessment of Motor and Process
for the Allen Cognitive Level Screen-5 (ACLS-5) and Skills. Volume 2: User’s Manual, Sixth Edition. Fort
Large Allen Cognitive Level Screen-5 (LACLS-5). Collins, CO: Three Star Press.
Camarillo, CA: ACLS and LACLS Committee. Fleming, R. and Purandare, N. (2010). Long-term care
American Occupational Therapy Association (AOTA) for people with dementia: Environmental design
(2008). Occupational therapy practice framework: guidelines. International Psychogeriatrics, 22:
Domain and process, Second Edition. The American 1084–1096.
Journal of Occupational Therapy, 62: 625–683. Folstein, M.F., Folstein, S.E. and McHugh, P.R. (1975).
Baum, C. and Edwards, D.F. (1993). Cognitive perfor- Mini-mental State: A practical method for grading
mance in senile dementia of Alzheimer’s type: The cognitive status of patients for the clinician. Journal of
kitchen task assessment. The American Journal of Psychiatric Research, 12: 189–198.
Occupational Therapy, 47: 431–436. Fossey, E. (2001). Effective interdisciplinary teamwork:
Baum, C.M., Connor, L.T., Morrison, T. et al. (2008). An occupational therapy perspective. Australasian
Reliability, validity, and clinical utility of the executive Psychiatry, 9: 232–235.
function performance test: A measure of executive Gitlin, L.N. and Chee, Y.K. (2006). Use of adaptive equip-
function in a sample of people with stroke. American ment in caring for persons with dementia at home.
Journal of Occupational Therapy, 62: 446–455. Bender, Alzheimer’s Care Today, 7: 32–40.
M., Norris, A. and Bauckham, P. (1987). Group Work Gitlin, LN, Corcoran, M.A. (2005) Occupational Therapy
with the Elderly. Bichester, Oxon: Winslow. and Dementia Care: The Environmental Skill-building
Bradford Dementia Group (1997). Evaluating Dementia Program for Individuals and Families. Bethesda, MD:
Care: The DCM Method. Bradford, England: Author. American Occupational Therapy Association.
Brawley, E.C. (2006). Design Innovations for Ageing and Gitlin, L.N., Corcoran, M., Winter, L. et al. (2001). A
Alzheimer’s: Creating Caring Environments. Hoboken, randomized, controlled trial of a home environmental
NJ: John Wiley & Sons. intervention. The Gerontologist, 41: 4–14.
Occupational therapy in dementia care 205

Gitlin, L.N., Hauck, W.W., Dennis, M.P. and Winter, Guide from the Uniform Data Set for Medical
L. (2005). Maintenance of effects of the home environ- Rehabilitation (1993). Adult FIM, Version 4. Buffalo, NY:
mental skill-building program for family caregivers and State University of New York.
individuals with Alzheimer’s disease and related disor- Hall, K.M. (1997). The functional assessment measure (FAM).
ders. The Journals of Gerontology Series A: Biological Journal Rehabilitation Outcomes Measure, 1: 63–65.
Sciences and Medical Sciences, 60: 368–374. Hasselkus, B.R. and Murray, B.J. (2007). Everyday occupa-
Gitlin, L.N., Hodgson, N., Jutkowitz, E. and Pizzi, L. (2010). tion, well-being and identity: The experience of care-
The cost-effectiveness of a nonpharmacologic inter- givers in families with dementia. The American Journal
vention for individuals with dementia and family care- of Occupational Therapy, 61 (1): 9–20.
givers: The tailored activity program. The American Hellen, C.R. (1998). Alzheimer’s Disease: Activities-
Journal of Geriatric Psychiatry, 18: 510–519. focused Care, Second Edition. Woburn, MA:
Gitlin, L.N., Schinfeld, S., Winter, L. et al. (2002). Butterworth-Heinemann.
Evaluating home environments of people with demen- Hopkins, A. and Smith, H. (1993). Willard and Spackman’s
tia: Interrater reliability and validity of the Home Occupational Therapy, Eighth Edition. Philadelphia,
Environmental Assessment Protocol (HEAP). Disability PA: J.B. Lippincott Co.
and Rehabilitation, 24 (1–3): 59–71. Jensen, L.E. and Padilla, R. (2011). Effectiveness of inter-
Gitlin, L.N., Winter, L., Burke, J. et al. (2008). Tailored ventions to prevent falls in people with Alzheimer’s
activities to manage neuropsychiatric behaviors in disease and related dementias. American Journal of
persons with dementia and reduce caregiver burden: Occupational Therapy, 65: 532–540.
A randomized pilot study. The American Journal of Johnson, N., Barion, A., Rademaker, A. et al. (2004). The
Geriatric Psychiatry, 16: 229–239. activities of daily living questionnaire: A validation
Gitlin, L.N., Winter, L., Corcoran, M. et al. (2003). Effects study in patients with dementia. Alzheimer’s Disease
of the home environmental skill-building program on and Associated Disorders, 18: 223–230.
the caregiver–care recipient dyad: 6-Month out- Josephsson, S., Backman, L., Nygard, L. and Borell, L.
comes from the Philadelphia REACH initiative. The (2000). Non professional caregivers’ experience of
Gerontologist, 43 (4): 532–546. occupational performance on the part of relatives with
Gitlin, L.N., Winter, L., Dennis, M.P. and Hauck, W.W. dementia: Implications for caregiver program in occu-
(2006). Assessing perceived change in the well-being pational therapy. Scandinavian Journal of Occupational
of family caregivers: Psychometric properties of the Therapy, 7: 61–66.
Perceived Change Index and response patterns. Klein, B., Gaedt, L. and Cook, G. (2013). Emotional robots:
American Journal of Alzheimer’s Disease and Other Principles and experiences with Paro in Denmark,
Dementias, 21: 304–311. Germany, and the UK. GeroPsych: The Journal of
Gitlin, L.N., Winter, L., Earland, T.V. et al. (2009). The Gerontopsychology and Geriatric Psychiatry, 26 (2):
Tailored Activity Program to reduce behavioral symp- 89–99.
toms in individuals with dementia: Feasibility, accept- Law, M., Cooper, B., Strong, S. et al. (1996). The per-
ability, and replication potential. The Gerontologist, son-environment-occupation model: A transactive
49: 428–439. approach to occupational performance. Canadian
Golding, E. and Cognitie, O. (1989). Middlesex Elderly Journal of Occupational Therapy, 63: 9–23.
Assessment of Mental State (MEAMS). Bury St. Law, M., Polatajko, H., Pollok, N. et al. (1991). Canadian
Edmunds, England: Thames Valley Test Company. Occupational Performance Measure. Toronto, Canada:
Graff, M.J., Vernooij-Dassen, M.J., Thijssen, M. et al. Canadian Association of Occupational Therapists.
(2006). Community based occupational therapy Lawton, M.P. and Brody, E.M. (1969). Assessment of older
for patients with dementia and their care ­givers: people: Self-maintaining and instrumental activities of
Randomised controlled trial. BMJ, 333 (7580): daily living. The Gerontologist, 9: 179–186.
1196–1199. Letts, L., Baum, C. and Permutter, M. (2003). Person-
Graff, M.J., Vernooij-Dassen, M.J., Thijssen, M. et al. environment-occupation assessment with older adults.
(2007). Effects of community occupational therapy on Occupational Therapy Practice, 2: 27–33.
quality of life, mood, and health status in dementia Letts, L., Edwards, M., Berenyi, J. et al. (2011). Using
patients and their caregivers: A randomized controlled occupations to improve quality of life, health and
trial. The Journals of Gerontology Series A: Biological wellness, and client and caregiver satisfaction for
Sciences and Medical Sciences, 62: 1002–1009. people with Alzheimer’s disease and related demen-
Graff, M.J.L., Vernooij-Dassen, M.J.F.J., Hoefnagels, tias. American Journal of Occupational Therapy, 65:
W.H.L. et al. (2003). Occupational therapy at home 497–504.
for older individuals with mild to moderate cogni- Lin, L.C., Yang, M.H., Kao, C.C. et al. (2009). Using
tive impairments and their primary caregivers: A pilot acupressure and Montessori-based activities to
study. Occupational Therapy Journal of Research, 23: decrease agitation for residents in dementia: A cross-
155–164. over trial. Journal of the American Geriatric Society,
206 Dementia

57: 1022–1029.Mahoney, F. and Barthel, D. (1965). Perrin, T., May, H. and Anderson, E. (2008). Wellbeing
Functional evaluation: The Barthel Index. Maryland in Dementia Care: An Occupational Approach for
State Medical Journal, 2: 61–65. Therapists and Carers, Second Edition. Philadelphia,
Mahurin, R., DeBettignies, B. and Pirozzolo, F. (1991). PA: Churchill Livingstone, Elsevier.
Structured assessment of independent living skills: Pfeffer, R.I., Kurosaki, T.T., Harrah, C.H. et al. (1992).
Preliminary report of a performance measure of func- Measurement of functional activities in older adults in
tional abilities in dementia. Journal of Gerontology, the community. Journal of Gerontology, 37: 323–329.
46: 58–66. Pool, J. (2008). The Pool Activity Level Instrument for
Marcy-Edwards, D., Grant, N.K. and Slater, S. (2005). The Occupational Profiling: A Practical Resource for Carers
exploration of self-determined behaviour and activity of People with Cognitive Impairment, Third Edition.
vignettes: A descriptive exploratory study. Alzheimer’s London, UK: Jessica Kingsley Publishers.
Disease Quarterly, 6: 35–51. de Poulin, C.L., Genlinas, I., Gauthier, S. et al. (2006).
McGrath, M. and O’Callaghan, C. (2014). Occupational Reliability and validity of the safety assessment scale
therapy and dementia care: A survey of practice in the for people with dementia living at home. The Canadian
Republic of Ireland. Australian Occupational Therapy Journal of Occupational Therapy, 73: 67–75.
Journal, 61: 92–101. Robert Dooley, N. and Hinojosa, J. (2004). Improving
Mioshi, E., Dawson, K., Mitchell, J. et al. (2006). The quality of life for persons with Alzheimer’s disease
Addenbrooke’s Cognitive Examination Revised and their family caregivers: Brief occupational therapy
(ACE‐R): A brief cognitive test battery for dementia intervention. The American Journal of Occupational
screening. International Journal of Geriatric Psychiatry, Therapy, 58: 561–569.
21: 1078–1085. Robinson, H., MacDonald, B.A., Kerse, N. and Broadbent,
Moyle, W., Cooke, M., Beattie, E. et al. (2013). Exploring E. (2013). The psychosocial effects of a companion
the effect of companion robots on emotional expres- robot: A randomized controlled trial. Journal of the
sion in older adults with dementia: A pilot randomized American Medical Directors Association, 14: 661–667.
controlled trial. Journal of Gerontological Nursing, Roger, K., Guse, L., Mordoch, E. and Osterreicher,
39: 46–53. A. (2012). Social commitment robots and dementia.
Oakley, F., Sutherland, T., Hill, J.L. et al. (1991). The daily Canadian Journal of Aging, 31: 87–94.
activities questionnaire: A functional assessment Stark, S.L., Somerville, E.K. and Morris, J.C. (2010).
for people with Alzheimer’s disease. Physical and In-home occupational performance evaluation
Occupational Therapy in Geriatrics, 10: 67–81. (I–HOPE). American Journal of Occupational Therapy,
O’Connor, D.W., Ames, D., Gardner, B. and King, 64: 580–589.
M. (2009). Psychosocial treatments of behavioural Strong, J. (2003). Seeing beyond the clouds: Best practice
symptoms in dementia: A systematic review of and occupational therapy. The Canadian Journal of
reports meeting quality standards. International Occupational Therapy, 70: 197–199.
Psychogeriatrics, 21: 225–240. Teri, L., Traux, P., Logsdon, R. et al. (1992). Assessment
Oliver, R., Blathwayt, J., Brackley, C. and Tamaki, T. (1993). of behavioural problems in dementia: The revised
Development of the safety assessment of function memory and behaviour problems checklist (RMBPC).
and the environment for rehabilitation (SAFER) tool. Psychology and Ageing, 7: 622–631.
Canadian Journal of Occupational Therapy, 60: 78–82. Teunisse, S. and Derix, M.M.A. (1997). The interview for
Padilla, R. (2011a). Effectiveness of environment-based deterioration in daily living activities in dementia:
interventions for people with Alzheimer’s disease and Agreement between primary and secondary care-
related dementias. American Journal of Occupational givers. International Psychogeriatrics, 9 (Suppl. 1):
Therapy, 65: 514–522. 155–162.
Padilla, R. (2011b). Effectiveness of interventions designed Thinnes, A. and Padilla, R. (2011). Effect of educational
to modify the activity demands of the occupations and supportive strategies on the ability of caregivers
of self-care and leisure for people with Alzheimer’s of people with dementia to maintain participation in
disease and related dementias. American Journal of that role. American Journal of Occupational Therapy,
Occupational Therapy, 65: 523–531. 65: 541–549.
Patterson, M.B., Mack, J.L., Neundorfer, M.M. et al. Thorgrimsen, L., Kennedy, L., Douglas, C. et al. (2002).
(1992). Assessment of functional ability in Alzheimer The group activity form: Is it valid and reliable? The
disease: A review and a preliminary report on the British Journal of Occupational Therapy, 65: 283–287.
Cleveland Scale for Activities of Daily Living. Alzheimer Thuné-Boyle, I.C.V., Iliffe, S., Cerga-Pashoja, A. et al.
Disease and Associated Disorders, 6: 145–163. (2012). The effect of exercise on behavioral and psy-
Perrin, T. (1997). The positive response schedule for severe chological symptoms of dementia: Towards a research
dementia. Aging and Mental Health, 1: 184–191. agenda. International Psychogeriatrics, 24: 1046–1057.
Occupational therapy in dementia care 207

Truscott, M. (2004). Adapting leisure and creative activi- caregivers-the design of a pragmatic randomised con-
ties for people with early stage dementias. Alzheimer’s trolled trial evaluating a Dutch programme in seven
Care Quarterly, 5: 92–102. German centres. BMC Geriatrics, 9: 44.
van der Ploeg, E.S., Eppingstall, B., Camp, C.J. et al. Williams, J., Drinka, T., Greenberg, J. et al. (1991).
(2013). A randomized crossover trial to study the Development and testing of the assessment of living
effect of personalized, one-to-one interaction using skills and resources (ASLAR) in elderly community-
Montessori-based activities on agitation, affect, and dwelling veterans. Gerontologist, 31: 84–91.
engagement in nursing home residents with Dementia. Wood, W., Harris, S., Snider, M. and Patchel, S.A. (2005).
International Psychogeriatrics, 25 (4): 565–575.Voigt- Activity situations on an Alzheimer’s disease special
Radloff, S., Graff, M., Leonhart, R. et al. (2011). Why care unit and resident environmental interaction,
did an effective Dutch complex psycho-social interven- time use, and affect. American Journal of Alzheimer’s
tion for people with dementia not work in the German Disease and Other Dementias, 20: 105–118.
healthcare context? Lessons learnt from a process Zarit, S.H., Reever, K.E. and Bach-Peterson, J. (1980).
evaluation alongside a multicentre RCT. BMJ Open, Relatives of the impaired elderly: Correlates of feeling
1 (1): e000094. doi:10.1136/bmjopen-2011-000094 of burden. The Gerontologist, 20: 649–655.
Voigt-Radloff, S., Graff, M., Leonhart, R. et al. (2009). Zgola, J. (1999). Care That Works: A Relationship
WHEDA study: Effectiveness of occupational therapy Approach to Persons with Dementia. Baltimore,
at home for older people with dementia and their MD: Johns Hopkins University Press.
20
Speech and language therapy in dementia
assessment and management

BRONWYN MOORHOUSE AND CAROLINE A. FISHER

across many of the cortical dementia syndromes, and are


20.1 INTRODUCTION often diagnostically important for differentiating between
underlying dementia pathologies. The following sections
Communication is integral to successful living (Lubinski, outline these changes.
1991); however, in dementia its breakdown is well docu-
mented. People with dementia (PWD) live for up to 20 years 20.2.1 MILD COGNITIVE IMPAIRMENT
after diagnosis and require help with communication to
optimize quality of life (Bourgeois, 2002), circumvent com- Mild cognitive impairment (MCI) captures the juncture
munication related behaviour problems and reduce care- between normal ageing and dementia. MCI is diagnosed
giver burden (Savundranayagam et al., 2005). This chapter in individuals who experience subjective cognitive difficul-
outlines cognitive-linguistic deficits seen in dementia and ties, as well as mild cognitive reductions on neuropsycho-
the assessments used by Speech and Language Therapists logical assessment, but experience no significant difficulties
(SLTs) to examine them. It explores how SLTs can help PWD with activities of daily living (Gauthier et al., 2006). MCI
and their carers to optimize interactions, and discusses safe includes amnestic, non-amnestic and multi-domain sub-
eating and swallowing. This chapter highlights, and further types (Petersen and Negash, 2008) and having MCI places
advocates for, the ongoing need for SLT services in dementia individuals at up to 50% risk of developing dementia, within
diagnosis and care. 5 years (Gauthier et al., 2006).
Over the last decade, research into early language
changes in MCI has increased significantly. Semantic
20.2 COGNITIVE-LINGUISTIC AND category fluency performances often differentiate those
with MCI from normal controls (see Taler and Phillips,
OROMOTOR CHANGES IN
2008, for a review). However, deficits in letter fluency
DEMENTIA performances also occur frequently, with some studies
showing differences according to MCI subtypes (Brandt
Subtle changes in language functioning occur as part of and Manning, 2009; Carter et al., 2012; Weakley et al.,
normal ageing. Word-finding difficulties and reductions 2013). Deficits in naming in MCI often contribute to a diag-
in confrontation naming, verbal fluency and spelling nosis of non-amnestic and multi-domain MCIs. A number
have been observed in a number of studies from the sixth of studies have reported poorer naming in people with
decade of life onwards (Au et al., 1995; Stuart-Hamilton MCI than controls (Taler and Phillips, 2008; Carter et al.,
and Rabbitt, 1997; Machulda et al., 2013). Grammar sim- 2012). Naming deficits may predict higher likelihood of
plification and reductions in the understanding of complex future dementia, although outcomes differ depending on
(written) language begin from the middle of the eighth MCI subtype and dementia syndrome (Testa et al., 2004;
decade (Bayles and Tomoeda, 2007). Beyond normal age- Belleville et al., 2014). In conversation, reductions in the
ing, more pronounced language changes are observed use of thematic information (Harris et al., 2008) and the

208
Speech and language therapy in dementia assessment and management 209

quality of complex discourse production (Fleming, 2014) 20.2.3 HIPPOCAMPAL SCLEROSIS (HS) –


have also been observed. Further, subtle input processing AGEING SUBTYPE
deficits have been observed in auditory, reading and oral
spelling comprehension (Tsantali et al., 2013). In later old age, there is a second neurodegenerative disease
that is characterized by significant memory problems. The
20.2.2 ALZHEIMER’S DISEASE primary features of HS, ageing subtype, are hippocampal
atrophy and significant memory problems in the absence of
Alzheimer’s disease (AD) is the most common neurode- seizure activity (cf. HS due to temporal lobe epilepsy). HS
generative syndrome. There are significant primary deficits is difficult to distinguish from AD and increases in preva-
in memory functioning and secondary difficulties in other lence from the age of 85 years (Nelson et al., 2011). On cog-
cognitive domains, including language, which restrict daily nitive evaluation, HS bears a number of similarities to AD
functioning (Dubois et al., 2010). From a language perspec- with significant deficits in new learning and memory, and
tive, word-finding difficulties in early AD are common. reductions in language measures, including confrontation
Conversation is often vague and semantically empty (Bayles picture naming and verbal fluency (Corey-Bloom et al.,
and Tomoeda, 2007). Communication is usually successful 1997; Nelson et al., 2011). Two studies have indicated that
for brief periods, but breaks down during lengthy interac- verbal fluency performances may be less affected in HS than
tions (Haak, 2002). Basic language comprehension is often AD (Zabar et al., 1998; Nelson et al., 2011). However, this
intact (Bayles et al.,1992) but may show some decline for finding is not consistent across studies, with confrontation
longer grammatically complex sentences (Clark, 1995), or naming (Zabar et al., 1998) and short-term auditory atten-
material requiring inference (Fried-Oken et al., 2000) or tion, also being identified as distinguishing cognitive fac-
abstraction (Haak, 2002). Poor memory and executive func- tors between HS and AD.
tioning in AD increase conversational repetition, and reduce
topic maintenance and adherence to turn-taking conven- 20.2.4 VASCULAR DEMENTIA (VAD)
tions (Watson et al., 1999). Oral reading, reading compre-
hension, spelling and writing are often relatively spared, but Rates of cerebrovascular pathology increase with age. In
written passage generation is poor (Bayles et al., 1992; Haak, some individuals the degree of pathology is significant and
2002). On standard language tests, individuals with early AD causes notable cognitive impairment. When these changes
exhibit similar but more pronounced difficulties than those are progressive, resulting in cognitive impairments that
with MCI, especially in the areas of verbal fluency, naming, significantly affect functioning, VaD may be diagnosed.
comprehension (auditory, reading and oral spelling) and Autopsy series have indicated, however, that VaD as a ­solitary
narrative output (Carter et al., 2012; Tsantali et al., 2013). pathology is less common, and usually occurs comorbidly
As AD advances, communicative intent and general with other neurodegenerative conditions, particularly AD
message frameworks are still evident in the moderate (Barker et al., 2002). In VaD, individual deficit profiles vary,
stages; however, utterances are depleted of content words depending on infarct locations, and are less uniform across
(Haak, 2002) and are less concise with reference errors language areas than with AD (Hopper and Bayles, 2001).
abounding (Bayles and Tomoeda, 2007). Due to pervasive Nonetheless, Vuorinen et al. (2000) found little difference in
lexical retrieval difficulty, generic labels (e.g. ‘thing’) often moderate AD and VaD group averages for deficits in com-
replace content words (Bourgeois, 2002) and the person prehension, naming and description tasks. While, Powell
may withdraw or speak excessively (Clark, 1995). Automatic et al. (1988) reported greater motoric speech involvement in
and social phrases may be retained, but people often forget VaD (e.g. dysarthria and intonation changes).
what they wish to say (Haak, 2002) or repeatedly ask the
same questions (Byrne and Orange, 2005). Paraphasias, 20.2.5 DEMENTIA WITH LEWY BODIES
confabulations and non-words appear and discourse break- (DLB)
down increases with poor propositional topic development,
difficulty changing topic and maintaining discourse flow The primary clinical features of DLB are progressive c­ ognitive
(Mentis et al., 1995; Fried-Oken et al., 2000; Bayles and decline, fluctuating attention and alertness, recurrent visual
Tomoeda, 2007). In the very later stages, language output hallucinations and parkinsonian motor features (McKeith
may breakdown altogether, with people eventually becom- et al., 1996). DLB also has a high level of co-mor­bidity with
ing mute (Bourgeois, 2002). Language input processing is AD (McKeith et al., 1995). Impaired language output in
also affected as AD advances. Comprehension of complex DLB is common, but the rates and nature of impairment in
grammar deteriorates and abstract interpretation is lost, ‘pure’ DLB compared to ‘pure’ AD differ across studies. It
although the understanding of one- (Haak, 2002) and is generally ­associated with poor verbal fluency (letter and
sometimes two-stage (Fried-Oken et al., 2000) commands semantic ­category), due to difficulties with attention and set
can remain preserved until very later stages. In these stages, maintenance, variable naming performances and difficulties
reading comprehension and writing are often limited to on semantically based activities (Hamilton et al., 2008). Often,
single words (Haak, 2002) and accuracy is reduced (Bayles mild extrapyramidal features (e.g. dysarthria and dysphagia)
et al., 1992). are also observed (Tomoeda, 2001).
210 Dementia

20.2.6 FRONTOTEMPORAL DEMENTIA when present, have been found to occur with verb naming
(FTD) in the early and later stages, on tasks requiring seman-
tic processing similar to sPPA, and are described below
FTD is an umbrella term for neurodegenerative condi- (Harciarek and Cosentino, 2013).
tions causing language and/or behavioural decline, with The remaining three FTD syndromes are characterized
the underlying neurodegenerative changes localized to the by language decline, with the nature and severity of the
frontal and temporal lobes (Grossman, 2002; Rohrer and aphasia progressing over time. The language features of each
Rosen, 2013). There are four well-described subtypes of this syndrome are described below, and the main distinguishing
condition. These are behavioural variant FTD (bvFTD) and features are summarized in Table 20.1 (where information
the three primary progressive aphasia (PPA) syndromes – has been collated from Gorno-Tempini et al. (2004, 2011)
nonfluent PPA (nfPPA), logopenic PPA (lPPA) and semantic and Harciarek and Cosentino [2013]). Non-fluent PPA is
PPA (sPPA). characterized by halting, non-fluent speech and articula-
Behaviour changes are the most prominent feature tion difficulties. Speech fluency is reduced and problems
of bvFTD. They include marked alterations in personal- with sentence construction and syntax also become appar-
ity, impulsivity, disinhibition, inappropriate or offensive ent. Although not always observed, sentence level auditory
remarks, hyperorality and changes in dietary preferences comprehension deficits have been documented with com-
(Pressman and Miller, 2014). The degree and nature of the plex sequential commands and syntactic constructions
cognitive dysfunction in bvFTD is variable. Many cases (Grossman et al., 1996; Hodges and Patterson, 1996; Blair
show little cognitive impairment in the early stages; how- et al., 2007; cf. Gorno-Tempini et al., 2004). Despite initial
ever, impairments in memory, executive function and lan- divergent presentations, evidence suggests that nfPPA pro-
guage domains are seen at times, particularly as the disease gresses towards eventual mutism at a more rapid pace than
progresses (Piguet et al., 2011). Specific language difficulties, in AD (Kertesz et al., 2003).

Table 20.1 Language features of the primary progressive aphasias by subtype


Subtype
Areas of Deficit nfPPA sPPA lPPA
Apraxia of speech ✓ – –
Agrammatism in ✓ – –
spontaneous speech
Speech fluency ✓ – ✓
Sentence construction ✓ – –
Syntax ✓ – –
Pragmatics of language – ✓ –
Phonologic paraphasias – – ✓
Repetition of single/short ✓ – –
words
Repetition of sentences ✓ – ✓
Letter fluency ✓ ✓ ✓
Semantic fluency ✓ ✓ ✓
Confrontation naming ✓ ✓ ✓
Object knowledge – ✓ –
Semantic matching – ✓ –
Auditory word recognition – ✓ –
Sequential commands ✓ – ✓
Syntactic comprehension ✓ – ✓
Single-word comprehension – ✓ –
Surface dyslexia – ✓ –
Surface dysgraphia – ✓ –
Sources: Gorno-Tempini, M.L. et al., Neurology, 76, 2011; Harciarek, M. and Cosentino, S. International Review of Psychiatry, 25, 2013;
Gorno-Tempini, M.L. et al, Annals of Neurology, 55, 2004.
Abbreviations: lppa, logopenic primary progressive aphasia; nfPPA, nonfluent primary progressive aphasia; sPPA, semantic primary progres-
sive aphasia.
✓ Represents commonly found areas of deficit (mild, moderate or severe).

✓ An area of difficulty that is a defining feature of the disorder.

– An area where deficits are not generally observed.


Speech and language therapy in dementia assessment and management 211

sPPA is characterized by impaired functioning on all linguistic expression, verbal memory, visuospatial skills
activities that are dependent on semantic knowledge (see and mental status. Although developed in the United States,
Table 20.1). Confrontation picture naming and semantic ABCD norms have also been found appropriate with UK
category verbal fluency performances are both impaired (Armstrong et al., 1996) and Australian (Moorhouse et al.,
at a more severe level than in nfPPA, lPPA and AD. Poor 1999) samples. The Barnes Language Assessment (BLA)
matching of picture-based semantic associations is seen on (Bryan et al., 2001) assesses mild, moderate and sometimes,
tasks such as the Pyramids and Palm Trees Test (Howard severe dementia. It examines expression, comprehension,
and Patterson, 1992). Repetition is generally well preserved reading and writing, memory and executive functions
along with speech fluency in conversation and articulation, using items adapted from existing tests. A third tool, the
while single-word comprehension is reduced. However, as Progressive Aphasia Severity Scale (PASS) (Sapolsky et al.,
the disease progresses, difficulty with pragmatics, including 2014), quantifies impairments across 13 speech, language,
turn-taking, thematic perseveration and interruptions may pragmatic and functional communication domains. It
develop (Kertesz et al., 2010). is particularly useful for distinguishing PPA subtypes
lPPA is characterized by word retrieval deficits in spon- and determining deterioration in language over time and
taneous speech, moderately impaired confrontation nam- requires skilled clinical judgements of language function-
ing abilities and marked difficulty with sentence repetition, ing, based on domain specific language test performances
but relatively spared single-word repetition (see Table 20.1). (discussed below).
Fluency of spontaneous speech in lPPA is affected, due to In addition to general language batteries, SLTs can also
frequent word-finding difficulties, but agrammatism is not utilize individual speech and language tests to compre-
observed. Phonological paraphasias are also frequently hensively examine a language skill in more detail often
noted. using normative data most appropriate to the population
being assessed. Examples include the Boston Naming Test
(BNT) (Kaplan et al., 2001), the Controlled Oral Word
20.3 COGNITIVE-LINGUISTIC Association Test (COWAT) (Strauss et al., 2006), the
ASSESSMENTS USED IN Sentence Production Test (SPT) (Wilshire et al., 2014), the
DEMENTIA Putney Auditory Comprehension Screening Test (PACST)
(Beaumont, 2002), the Psycholinguistic Assessments of
Language Processing in Aphasia (PALPA) (Kay et al., 2009)
In diagnosing the presence and type of dementia, individ- the Pyramid and Palm Trees Test (PPT) (Howard and
ual profiles of cognitive-linguistic functioning ideally form Patterson, 1992) and the Cambridge Semantic Memory
a part of a memory clinic evaluation (Ames et al., 1992). Test Battery (Adlam et al., 2010). Tests of functional and
Accurate early dementia diagnosis is crucial in identifying pragmatic language skills may also be useful, particu-
strengths and weaknesses for optimal informational coun- larly for shaping therapeutic intervention, examples of
selling, some interventions and effective care plan devel-
these include the Functional Linguistic Communication
opment. It also allows for establishment of baselines for
Inventory (FLCI; Bayles and Tomoeda, 1994), the
intervention effects or decline over time (Tomoeda, 2001).
Communication Outcome Measure of Functional
Appropriate cognitive-linguistic normative data help to
Independence (COMFI) (Santo Pietro and Boczko, 1997)
distinguish those with early dementia from the ‘worried
and the Latrobe Communication Questionnaire (LCQ)
well’ (Bryan et al., 2001). Normative data are essential for
(Douglas et al., 2000).
determining whether difficulties represent actual deficits,
or are within the performance distribution seen in nor-
mal ageing. An individual’s academic and occupational
history should also be taken into account, along with esti-
20.4 OPTIMIZING COMMUNICATION
mated premorbid intelligence quotient (IQ) (if known from
neuropsychological assessment). Premorbid IQ can affect
IN DEMENTIA
performance on a variety of language activities, includ-
ing confrontation naming, verbal fluency and token tasks It is useful to conceptualize communication interventions
(Steinberg et al., 2005), while education level can affect the in dementia in terms of the International Classification of
diagnostic utility of verbal fluency tasks in differentiating Functioning, Disability and Health (ICF) (World Health
normals and those with MCI or early dementia (Radanovic Organization [WHO], 2001) (Byrne and Orange, 2005).
et al., 2009). Interventions are described below with reference to this
The best known cognitive-linguistic assessment designed framework. Irrespective of intervention level and the chal-
specifically for dementia assessment is the Arizona Battery lenges involved, every effort should be made to involve
for Communication Disorders of Dementia (ABCD) those with PPA (Croot et al., 2011), PWD and/or their regu-
(Bayles and Tomoeda, 1993). It assesses mild-to-moderate lar communication partners in collaborative goal setting
dementia across five constructs: linguistic comprehension, (see Volkmer, 2013 for detailed discussion).
212 Dementia

20.4.1 ICF, DISABILITY AND HEALTH the promising but limited findings regarding spelling treat-
IMPAIRMENT-DIRECTED ments in lPPA and discussed need for additional research
INTERVENTIONS to elucidate appropriate dysgraphia therapies across PPA
types. Tsapkini et al. (2014) found that phoneme-to-
Impairment-directed approaches to restore communica- grapheme conversion training augmented by non-invasive
tion often involve relearning or maintenance of the abil- transcranial direct current stimulation in nfPPA or lPPA
ity to name specific word groups or learn key information improved spelling on untrained items, which was mostly
in PWD. Spaced retrieval training (SRT) (using recall at maintained at 2-month follow-up.
g radually increasing intervals) (Abrahams and Camp,
­
1993; Brush and Camp, 1998), or coaching on picture 20.4.2 ICF, DISABILITY AND HEALTH
description and word category assignments (Arkin and ACTIVITY AND PARTICIPATION-
Mahendra, 2001) facilitated performance on specific tasks DIRECTED INTERVENTIONS
with some­maintenance over several weeks (Abrahams and
Camp, 1993). More recently, Hopper et al. (2010) exam- These interventions aim primarily to optimize the func-
ined SRT in learning face-name associations primarily in tional communication of PWD. Clark and Witte (1991)
AD. Some participants could learn and relearn semantic advocated adaptive strategy training in early AD – e.g.
information, however, associations faded within 6 weeks. saying ‘Please repeat exactly what you just said’ or ‘Give
Limited recent studies examining specific impairment- me a little time. I’m having trouble finding the exact
directed therapy may reflect an emphasis shift of commu- words’ or ‘I forgot what we were discussing’. They also
nication interventions for more global dementias to other suggested self-cueing strategies (e.g. using circumlo-
ICF levels, where carry over is more likely to be enduring cution or semantically related words) and teaching of a
(discussed below). script strategy for simple storytelling (e.g. theme, char-
Conversely in PPA, recent impairment-directed studies acters, setting and events) to improve cohesion and topic
have proliferated; however, due to rarity of this disorder, maintenance in AD (Clark, 1988 – cited in Clark and
participant numbers in individual studies remain small Witte, 1991). Carers reported maintained or increased
and findings somewhat anecdotal. As for more common functional communication and aphasia test scores that
dementias, therapeutic efficacy may be gauged by stabil- were stable for 6 months.
ity or slowed deterioration as well as improvement (Croot Group programmes using residual communication
et al., 2011). Acquisition and retention patterns may also strengths with tangible stimuli may also be beneficial.
vary according to PPA type (Newhart et al., 2009). Jokel Santo Pietro and Boczko (2001) used procedural memories
and Anderson (2012) found that errorless word relearning to facilitate communication in residents with AD. During
precipitated better performance and maintenance in sPPA. ‘breakfast clubs’ members used greetings, discussed and
Reviews of PPA naming therapy studies also revealed chose food, helped one another prepare ingredients, set
promising immediate and maintenance results; however, the table, made general conversation, cleaned up and said
potential for generalizability requires further research goodbye. SLTs provided visual and semantic cues, paired
(Carthery-Goulart et al., 2013; Jokel et al., 2014). Savage choices and trigger phrases to facilitate choice making.
et al. (2014) reported generalization of relearned names Participants improved significantly on the ABCD and also
on a specific description task in sPPA; however, circum- had more self-initiated on topic comments per session, more
stances leading to conversational generalization remain cross-conversation exchanges and increased procedural ini-
uncertain. In an individual with nfPPA, Henry et al. tiation. Matched controls in a conversation group, showed
(2013) found that after simplification and repetition inter- either no gains or slight declines.
ventions for speech sound and syllabic errors, oral read- Several studies have examined activity/participation-
ing and picture description inaccuracies remained below directed therapies in PPA. Cartwright and Elliot (2009)
pretreatment levels for 6 months or longer. Beeson et al. facilitated comprehension of a half-hour TV documentary
(2011) used graded semantic feature analysis followed by in four people with PPA using techniques including initial
generative naming tasks – an intervention designed to tax topic priming, pausing to allow clinician-facilitated updat-
lexical retrieval on demanding tasks to facilitate retrieval ing of a whiteboard plot summary and a clinician-supported
in more functional contexts in an individual with lPPA. feature analysis guide sheet. Between pre- and post-test (on
They found significantly improved naming post-treatment different episodes with opportunity to independently use
and at 6-month follow-up indicative of both maintenance the guide sheet), participants retold significantly more infor-
and some generalization. Interestingly comparison of mation units, approaching performance of controls. Kindell
functional magnetic resonance imaging (fMRI) before et al. (2013) analysed spontaneous adaptive facial, spatial
and after treatment showed marked activation in the left and tone-of-voice–based enactment in one individual with
dorsolateral prefrontal region (consistent with strategic sPPA. They concluded that examining adaptive strategies
planning and monitoring of l­ exical retrieval), not engaged that facilitate conversational repair may be a fruitful place
during pretreatment scanning. Graham (2014) reviewed to begin therapy. Wong et al. (2009) had previously initiated
Speech and language therapy in dementia assessment and management 213

this type of work and argued, given ultimate progression, (2000) found that 6 months post-training, when planning
that it makes sense to focus on conversational effective- a menu together, FOCUSED trained carers asked signifi-
ness because such strategies remain useful in later stages of cantly more yes/no and choice questions (associated with
sPPA. They targeted use of residual (automatic) verbal and successful communication outcomes) than did controls.
non-verbal abilities along with props in a discourse-based A DVD-based carer training programme titled RECAPS
intervention with an individual with sPPA. The communi- and MESSAGE (acronyms to prompt memory and com-
cation partner was educated to interpret underlying non- munication strategies) was developed by Smith et al. (2011).
verbal messages and facilitate multimodal message delivery It provides information on memory and communication
to both herself and other conversation partners. This indi- necessary in everyday life and commonly preserved and
vidual continued to participate in conversation despite dis- affected abilities across dementia stages. It includes acted
ease progression. vignettes demonstrating strategies and scenarios to prompt
strategy use. Evaluation of efficacy among professional car-
20.4.3 ENVIRONMENTAL SUPPORTS TO ers revealed significant increases in knowledge about sup-
COMMUNICATION port strategies and some increases in carer satisfaction
(Broughton et al., 2011).
Conversation is collaborative. Where one partner has Bourgeois (1992) evaluated use of personalized memory
AD, the other can use strategies to optimize interaction wallets to enhance communication in AD. Wallets con-
(Muller and Wilson, 2008). People with mild AD should tained individualized simple written information, draw-
be ­encouraged to speak (even when having word-finding ings and photographs. After role-playing, carers used
­difficulties) and urged to use spared communicative abilities wallets to facilitate discussion about ‘day’, ‘family’ and
to reinforce self-worth – e.g. reading to a grandchild (Haak, ‘yourself’. During training and maintenance, participants
2002). When optimizing comprehension, the partner should made novel on-topic statements and fewer ambiguous utter-
match language complexity to individuals (Haak, 2002) – ances. Most participants used their wallets spontaneously
initially, using slightly slower speech and reduced informa- to initiate conversations. Haight et al. (2006) evaluated out-
tion (Bourgeois, 2002) and with progression, conversing comes when care staff used similar ‘Life Story Books’ with
about tangible things, ­simplifying vocabulary, using non- 15 PWD. Intervention resulted in significantly improved
verbal cues and restating messages when they are not under- scores compared with controls on depression, mood, mental
stood (Bayles and Tomoeda, 2007) or forgotten (Enderby, status and communication scales. Bourgeois (2013) further
2002). Before s­tarting, it helps to orient the PWD to topic outlines evolution of memory books and their usefulness
and then maintain and extend (Kessler et al., 2001). In later in enhancing conversation, communicating basic needs
dementia, ­questions with alternatives (e.g. ‘do you want fish and reducing challenging behaviours. Because senses of
or chicken?’), short ­utterances and names (rather than pro- smell and taste may be relatively preserved in later demen-
nouns) are useful (Haak, 2002). Demonstration to aid com- tia, stimulation via spice jars, scratch and sniff books, or
prehension, ­avoiding sarcasm and innuendo, use of native ethnic foods (Lubinski, 1991; Bourgeois, 2002) or having a
language and familiar wording (e.g. respectful title or maiden pet or a toy present (Hopper et al., 1998) may also facilitate
name) also promote ­conversation. When confusion occurs, communication.
carers should r­eintroduce themselves (Haak, 2002). Use of Fried-Oken et al. (2012) outlined three treatment goals
­questionnaires (e.g. LCQ; Douglas et al., 2000) r­egarding for Augmentative and Alternative Communication (AAC)
communicative behaviours are useful not only in under- use in PPA – compensating for progressive language loss
standing PWD, but also in ­giving SLTs insights into ­effective (rather than skill recovery), starting early while learning is
interventions for individual c­ ommunication ­partners. These still possible and including key communication partners in
types of ­interventions bear many similarities with ‘Supported training. Initially, SLTs may support usage of appropriate
Conversation’ now widely used in n ­ on-progressive aphasia electronic communication devices or develop individually
(Kagan et al., 2001) and also being introduced for PWD tailored communication books with specific information,
(see Volkmer, 2013 for detailed discussion). Carers are likely questions and text or gestures to reduce communication
to benefit from such training protocols along with more breakdown. As PPA progresses, SLTs should work with
dementia-specific programmes described below. individuals and partners to develop AAC devices such as
Santo Pietro (2002) highlighted SLTs’ role in training talking photo albums or tablets with AAC applications to
trainers or care staff to use effective communication in a express choices, needs and feelings. In later stages, with
‘teaching by modelling’ paradigm, which identifies envi- minimal functional language, partners can support par-
ronmental supports and barriers to effective communica- ticipation by carrying conversation content and presenting
tion. The FOCUSED programme for carers, uses role-play limited choices.
to emphasize strategies based on an interactive discourse Talking Mats are an innovative, accessible and inex-
model – F: face-to-face, O: orient to conversation topic, C: pensive system supporting people with limited communi-
continue the topic, U: unstick communication blocks, S: cation in giving information, making decisions or setting
structure questions, E: exchange conversation and D: direct goals. Structured topics are presented to support compre-
short sentences (Ripich and Wykle, 1996). Ripich et al. hension and expression. Three sets of pictorial symbols
214 Dementia

(topic, options and visual scale) are used. The person places initiation, poor eating environment and limited mealtime-
relevant options under the visual scale (e.g. happy, unsure, assistance. Recently, the issue of bland institutional food has
not happy), which is photographed for further reference. been addressed by some organizations (e.g. Morgan-Jones
Murphy et al. (2005) found Talking Mats assisted seven et al., 2014), but remains a widespread problem. SLTs also
PWD and minimal speech express to views on activ- have a role in ensuring optimal oral hygiene particularly
ity themes by supporting communication and reducing when PWD have significant risk of aspiration pneumonia
memory load. Use of talking mats to discuss problems with and especially when they drink free (unthickened) water
daily living (‘managing’, ‘needing assistance’, ‘not manag- under controlled circumstances (Karagiannis et al., 2011).
ing’ scale) was also more effective than usual communica- SLTs assess oropharyngeal function to reduce choking and
tion modes (Murphy and Oliver, 2013). SLTs have a crucial aspiration risk (e.g. by modifying consistencies or position-
role to play in supporting PWD (particularly those with ing) (Gillick and Mitchell, 2002). Although SLTs may use
disproportionate language decline in PPA) to participate instrumental examinations to investigate dysphagia, PWD
in capacity and decision making assessments. This support often cannot cooperate with such procedures (Gillick and
usually involves tailored use of AAC (see Volkmer, 2013 for Mitchell, 2002). A percutaneous endoscopic gastrostomy
detailed discussion). (PEG) tube is sometimes inserted to administer nutrition
In late dementia, when many individuals become directly into the stomach when PWD can no longer swallow
non-intentional communicators (see Bloomberg et al., safely or reject food or drink (Gillick and Mitchell, 2002).
2009), SLTs can work with carers to make ‘personal Although many consider withholding food cruel, people
­communication ­dictionaries’ (PCDs) (Siegel and Whetherby, appear to experience little thirst or hunger at life’s end
2000). Compilation of PCDs requires careful observation (Bourgeois and Hickey, 2009). PEG limitations include diar-
and ­documentation of consistent non-verbal behaviours – rhoea and nausea in 10% of people, tubes frequently being
‘What the person does’ (e.g. takes your hand and pulls it pulled out (Gillick and Mitchell, 2002) and insufficient evi-
towards his mouth), ‘What it might mean’ (e.g. ‘I’m hungry dence of increased survival rates (Sampson et al., 2009).
or thirsty’), ‘What you should do’ (e.g. give food or drink if Some individuals with an ‘unsafe swallow’ can still tolerate
­appropriate). PCDs are especially useful when staff do not oral intake if optimal assistance and positioning are main-
know the ­person, but may also help familiar carers ­structure tained and consistencies are carefully monitored (Cullen,
observations to best meet individual needs and reduce
­ 2011). If there is no medical living will, guardians should
­problem behaviours. PCDs should be easily accessible for decide based on what they believe the PWD would have
reference and amendment. chosen for themselves. As part of a team, SLTs should pro-
vide family with clear information regarding PEGs versus
20.4.4 PERSONAL AND ENVIRONMENTAL other options (e.g. mouth swabs), so they can make the best
BARRIERS TO COMMUNICATION decision for their relative (Gillick and Mitchell, 2002).

Rejecting optimal communication strategies can cre-


ate unnecessary barriers (Byrne and Orange, 2005).
20.6 CONCLUSIONS
Furthermore, for PWD, Palmer et al. (1999) reported signif-
icantly reduced problem behaviours after hearing aid fitting
while Lemke (2011) outlined strategies to reduce audiologi- In recent years, knowledge about SLTs’ role in assessing and
cal assessment challenges. Vision should also be corrected, managing dementia has increased. There is greater under-
background noise limited, lighting optimized (Enderby, standing of PPA and of AAC to facilitate interactions and
2002) and dignity and cultural sensitivity maintained better use of the ICF framework to promote communica-
(Lydall-Smith et al., 1996). Poor management of medical tive participation. There is also greater awareness of issues
conditions and drug regimens can also hinder communi- around eating in dementia. However, there is an ongoing
cation (Clark, 1995). Although physical space is often lim- need to broaden the evidence base for SLT interventions
ited and opportunities for solitude or intimacy are rare, in PWD. Single case investigations require collection of
socialization can also be minimal (Lubinski, 1991). Some lengthy baseline as well as intervention data, while group
attitudes should also be challenged, e.g. ignoring those with studies require strict selection criteria and well-matched
severe communication issues (Lubinski, 1991). control groups (Lum, 2001).
The upcoming demographic explosion of PWD is well
recognized. Consequently SLTs have developed policy posi-
tion papers, submissions to governmental enquiries and
20.5 EATING IN DEMENTIA detailed examinations of service provision (e.g. Taylor et al.,
2009; Speech Pathology Australia [SPA], 2012; Torresi et al.,
Mid-to-late dementia individuals often develop malnutri- 2012; Royal College of Speech and Language Therapists
tion, weight loss and/or dehydration. Bourgeois and Hickey [RCSLT], 2014). These publications reflect increasing
(2009) stress that SLTs must consider dysphagia along with acknowledgement of the specialist role SLTs have to play in
other precipitating factors, e.g. distractibility, reduced direct service provision and workforce training. Recently,
Speech and language therapy in dementia assessment and management 215

such services have improved in the United Kingdom; how- Beeson, P.M., King, R.M., Bonakdarpour, B. et al. (2011).
ever, issues regarding equity of access persist (RCSLT, 2014). Positive effects of language treatment for the logope-
In Australia, denial of optimal communication intervention nic variant of primary progressive aphasia. Journal of
for many older Australians living in care facilities remains Molecular Neuroscience, 45: 724–736.
an issue (SPA, 2014). As such, there is an ongoing need for Belleville, S., Gauthier, S., Lepage, E. et al. (2014).
professional associations to use research findings to further Predicting decline in mild cognitive impairment: A
advocate for equity of access to SLT services for those living prospective cognitive study. Neuropsychology, 28:
with dementia in all its forms. 643–652.
Blair, M., Marczinski, C. A., Davis-Faroque, N. and Kertesz,
REFERENCES A. (2007). A longitudinal study of language decline
in Alzheimer’s disease and frontotemporal demen-
Abrahams, J.P. and Camp, C.J. (1993). Maintenance and tia. Journal of the International Neuropsychological
generalization of object naming training in anomia Society, 13: 237–245.
associated with degenerative dementia. Clinical Bloomberg, K., West, D., Johnson, H. and Iacono, T.
Gerontologist, 12: 57–72. (2009). The Triple C: Checklist of Communicative
Adlam, A.L.R., Patterson, K., Bozeat, S. and Hodges, J.R. Competencies (Revised). Box Hill, Australia: Scope
(2010). The Cambridge Semantic Memory Test Battery: (Vic).
Detection of semantic deficits in semantic dementia Bourgeois, M.S. (1992). Evaluating memory wallets in
and Alzheimer’s disease. Neurocase, 16: 193–207. conversations with persons with dementia. Journal of
Ames, D., Flicker, L. and Helme, R. (1992). A memory clinic Speech and Hearing Research, 35: 1344–1357.
at a geriatric hospital: Rationale, routine and results Bourgeois, M.S. (2002). Where is my wife and when am
from the first 100 patients. The Medical Journal of I going home? The challenge of communicating with
Australia, 156: 618–622. persons with dementia. Alzheimer’s Care Quarterly, 3:
Arkin, S. and Mahendra, N. (2001). Discourse analysis 132–144.
of Alzheimer’s patients before and after interven- Bourgeois, M.S. (2013). Memory and Communication
tion: Methodology and outcomes. Aphasiology, 15: Aids for People with Dementia. Baltimore, MD: Health
533–569. Professions Press, U.S.
Armstrong, L., Bayles, K., Borthwick, S. and Tomoeda, C. Bourgeois, M.S. and Hickey, E.M. (2009). Dementia: From
(1996). Use of the Arizona battery for communication Diagnosis to Management: A Functional Approach.
disorders of dementia in the UK. European Journal of Hove, England: Taylor and Francis.
Disorders of Communication, 31: 171–180. Brandt, J. and Manning, K.J. (2009). Patterns of word-
Au, R., Joung, P., Nicholas, M. et al. (1995). Naming ability list generation in mild cognitive impairment and
across the adult life span. Aging, Neuropsychology Alzheimer’s disease. The Clinical Neuropsychologist,
and Cognition: A Journal on Normal and Dysfunctional 23: 870–879.
Development, 2: 300–311. Broughton, M., Smith, E.R., Baker, R. et al. (2011).
Barker, W.W., Luis, C.A., Kashuba, A. et al. (2002). Relative Evaluation of a caregiver education program to sup-
frequencies of Alzheimer disease, Lewy body, vascu- port memory and communication in dementia: A
lar and frontotemporal dementia, and hippocampal controlled pretest-posttest study with nursing home
sclerosis in the State of Florida Brain Bank. Alzheimer staff. International Journal of Nursing Studies, 48:
Disease and Associated Disorders, 16: 203–212. 1436–1444.
Bayles, K.A. and Tomoeda, C.K. (1993). The Arizona Brush, J.A. and Camp, C.G. (1998). Using spaced retrieval
Battery for Communication Disorders of Dementia. as an intervention during speech-language therapy.
Austin, TX: Pro-Ed. Clinical Gerontologist, 19: 51–64.
Bayles, K.A. and Tomoeda, C.K. (1994). Functional Bryan, K., Binder, J., Dann, C. et al. (2001). Development
Linguistic Communication Inventory. Austin, TX: of a screening instrument for language in older
Pro-Ed. people. Ageing and Mental Health, 5: 371–378.
Bayles, K.A. and Tomoeda, C.K. (2007). Cognitive- Byrne, K. and Orange, J.B. (2005). Conceptualizing
Communication Disorders of Dementia. San Diego, communication enhancement in dementia for family
CA: Plural Publishing, Inc. caregivers using the WHO-ICF Framework. Advances
Bayles, K.A., Tomoeda, C.K. and Trosset, M.W. (1992). in Speech-Language Pathology, 7: 187–202.
Relation of linguistic communication abilities of Carter, S.F., Caine, D., Burns, A. et al. (2012). Staging of
Alzheimer’s patients to stage of disease. Brain and the cognitive decline in Alzheimer’s disease: Insights
Language, 42: 454–472. from a detailed neuropsychological investigation of
Beaumont, J.G. (2002). PACST: Putney Auditory mild cognitive impairment and mild Alzheimer’s dis-
Comprehension Screening Test. Bury St. Edmunds, ease. International Journal of Geriatric Psychiatry, 27:
England: Thames Valley Test Company. 423–432.
216 Dementia

Carthery-Goulart, M.T., da Costa da Silveira, A., Machado, Gillick, M.R. and Mitchell, S.L. (2002). Facing eating
T.H. et al. (2013). Nonpharmacological interventions difficulties in end stage dementia. Alzheimer’s Care
for cognitive impairments following primary progres- Quarterly, 3: 227–232.
sive aphasia. Dementia and Neuropsychologia, 7: Gorno-Tempini, M.L., Dronkers, N.F., Rankin, K.P. et al.
122–131. (2004). Cognition and anatomy in three variants of
Cartwright, J. and Elliott, K. A. E. (2009). Promoting stra- primary progressive aphasia. Annals of Neurology, 55:
tegic television viewing in the context of progressive 335–346.
language impairment. Aphasiology, 23: 266–285. Gorno-Tempini, M.L., Hillis, A.E., Weintraub, S. et al.
Clark, L.W. (1988). Enhancement of Communication (2011). Classification of primary progressive aphasia
Adequacy in Early Stage Alzheimers’s Disease Patients. and its variants. Neurology, 76: 1006–1014.
Unpublished research paper. New York, NY: Hunter Graham, N. (2014). Dysgraphia in primary progressive
College of CUNY. aphasia: Characterisation of impairments and therapy
Clark, L.W. (1995). Interventions for persons with options. Aphasiology, 28: 1092–1111.
Alzheimer’s disease: Strategies for maintaining and Grossman, M. (2002). Frontotemporal dementia: A review.
enhancing communicative success. Topics in Language Journal of the International Neuropsychological
Disorders, 15: 47–65. Society, 8: 566–583.
Clark, L.W. and Witte, K. (1991). Nature and efficacy of Grossman, M., Mickanin, J., Onishi, K. et al. (1996).
communication management in Alzheimer’s disease. Progressive nonfluent aphasia: Language, cogni-
In Lubinski, R. (ed.), Dementia and Communication. tive, and PET measures contrasted with prob-
Philadelphia, PA: B.C. Decker Inc., pp. 238–256. able Alzheimer’s disease. Journal of Cognitive
Corey-Bloom, J., Sabbagh, M. N., Bondi, M. W. et al. Neuroscience, 8: 135–154.
(1997). Hippocampal sclerosis contributes to dementia Haak, N.J. (2002). Maintaining connections:
in the elderly. Neurology, 48: 154–160. Understanding communication from the perspective
Croot, K., Taylor, C. and Nickels, L. (2011). What’s the evi- of persons with dementia. Alzheimer’s Care Quarterly,
dence? Evidence for speech, language, and communi- 3 (2): 116–131.
cation interventions in progressive aphasia. Australian Haight, B.K., Gibson, F. and Michel, Y. (2006). The
Communication Quarterly, 13: 37–40. Northern Ireland life review/life storybook project for
Cullen, C. (2011). Gastrostomy tube feeding in adults: The people with dementia. Alzheimer’s and Dementia, 2:
risks, benefits and alternatives. Proceedings of the 56–58.
Nutrition Society, 70: 293–298. Hamilton, J.M., Salmon, D.P., Galasko, D. et al. (2008).
Douglas, J.M., O’Flaherty, C.A. and Snow, P.C. (2000). Visuospatial deficits predict rate of cognitive decline
Measuring perception of communicative ability: The in autopsy-verified dementia with Lewy bodies.
development and evaluation of the La Trobe communi- Neuropsychology, 22: 729–737.
cation questionnaire. Aphasiology, 14: 251–268. Harciarek, M. and Cosentino, S. (2013). Language, execu-
Dubois, B., Feldman, H.H., Jacova, C. et al. (2010). tive function and social cognition in the diagnosis of
Revising the definition of Alzheimer’s disease: A new frontotemporal dementia syndromes. International
lexicon. The Lancet Neurology, 9: 1118–1127. Review of Psychiatry, 25: 178–196.
Enderby, P. (2002). Promoting communication skills with Harris, J.L., Kiran, S., Marquardt, T.P. and Fleming,
people who have dementia. In Stokes, G. and Goudie, V.B. (2008). Communication wellness check up©:
F. (eds.), The Essential Dementia Care Handbook. Age related changes in communicative abilities.
Bicester, Oxfordshire: Speechmark, pp. 102–108. Aphasiology, 22: 813–825.
Fleming, V.B. (2014). Early detection of cognitive-linguistic Henry, M.L., Meese, M.V., Truong, S. et al. (2013).
change associated with mild cognitive impairment. Treatment for apraxia of speech in nonfluent variant
Communication Disorders Quarterly, 35: 146–157. primary progressive aphasia. Behavioral Neurology,
Fried-Oken, M., Beukelman, D.R. and Hux, K. (2012). 26: 77–88.
Current and future AAC research considerations for Hodges, J.R. and Patterson, K. (1996). Nonfluent progres-
adults with acquired cognitive and communication sive aphasia and semantic dementia: A comparative
impairments. Assistive Technology, 24: 56–66. neuropsychological study. Journal of the International
Fried-Oken, M., Rau, M.T. and Oken, B. S. (2000). AAC Neuropsychological Society, 2: 511–524.
and dementia. In Beukleman, D., Yorkston, K. and Hopper, T. and Bayles, K.A. (2001). Management
Reichle, J. (eds.), Augmentative and Alternative of neurogenic communication disorders associ-
Communication for Adults with Acquired Neurological ated with dementia. In Chapey, R. (ed.), Language
Disorders. Baltimore, MD: Paul H Brooks, pp. Intervention Strategies in Aphasia and Related
375–405. Neurogenic Communication Disorders, Fourth Edition.
Gauthier, S., Reisberg, B., Zaudig, M. et al. (2006). Mild Philadelphia, PA: Lippincott, Williams and Wilkins,
cognitive impairment. The Lancet, 367: 1262–1270. pp. 829–846.
Speech and language therapy in dementia assessment and management 217

Hopper, T., Bayles, K.A. and Tomoeda, C.K. (1998). Using Lubinski, R. (1991). Environmental considerations for
toys to facilitate communicative function in individuals elderly patients. In Lubinski, R. (ed.), Dementia and
with Alzheimer’s disease. Journal of Medical Speech Communication. Philadelphia, PA: B.C. Decker Inc.,
Language Pathology, 6: 73–80. pp. 257–273.
Hopper, T., Drefs, S.J., Bayles, K.A. et al. (2010). The Lum, C. (2001). Scientific Thinking in Speech and
effects of modified spaced retrieval training on Language Therapy. London, England: Lawrence
learning and retention of face–name associations Erlbaum.
by individuals with dementia. Neuropsychological Lydall-Smith, S., Moorhouse, B. and Gilchrist, J. (1996).
Rehabilitation, 20: 81–102. Culturally Appropriate Dementia Assessment.
Howard, D. and Patterson, K. (1992). Pyramids and Palm Trees. Canberra, Australia: Commonwealth of Australia.
Bury St. Edmonds, England: Thames Valley Test Company. McKeith, I.G., Galasko, D., Kosaka, K. et al. (1996).
Jokel, R. and Anderson, N.D. (2012). Quest for the best: Consensus guidelines for the clinical and pathologic
Effects of errorless and active encoding on word re- diagnosis of dementia with Lewy bodies (DLB): Report
learning in semantic dementia. Neuropsychological of the consortium on DLB international workshop.
Rehabilitation, 22: 187–214. Neurology, 47: 1113–1124.
Jokel, R., Graham, N.L., Rochon, E. and Leonard, C. (2014). McKeith, I.G., Galasko, G.K., Wilcock, G.K. and Byrne, E.J.
Word retrieval therapies in primary progressive apha- (1995). Lewy body dementia – Diagnosis and treat-
sia. Aphasiology, 28: 1038–1068. ment. British Journal of Psychiatry, 167: 709–717.
Kagan, A., Black, S.E., Duchan, F.J. et al. (2001). Training Machulda, M., Pankratz, V., Christianson, T. et al.
volunteers as conversation partners using “Supported (2013). Practice effects and longitudinal cogni-
Conversation for Adults with Aphasia” (SCA): A con- tive change in normal aging versus incident mild
trolled trial. Journal of Speech Language and Hearing cognitive ­impairment and dementia in the Mayo
Research, 44: 624–638. Clinic Study of Aging. Alzheimer’s and Dementia,
Kaplan, E., Goodglass, H. and Weintraub, S. (2001). 9 (Suppl. 4): S793.
Boston Naming Test, Second Edition. Philadelphia, PA: Mentis, M., Briggs-Whittaker, J. and Gramigna, G.D.
Lippincott Williams & Wilkins. (1995). Discourse topic management in senile demen-
Karagiannis, M.J., Chivers, L. and Karagiannis, T.C. (2011). tia of the Alzheimer type. Journal of Speech and
Effects of oral intake of water in for patients with oro- Hearing Research, 38: 1054–1066.
pharyngeal dysphagia. BMC Geriatrics, 11: 9. Available Moorhouse, B., Douglas, J. Pannacio, J. and Steel, G.
at http://www.biomedcentral.com/1471–2318/11/9. (1999). Use of the Arizona battery for communication
Accessed 18 October 2014 disorders of dementia in an Australian context. Asia
Kay, J., Lesser, R. and Coltheart, M. (2009). PALPA: Pacific Journal of Speech, Language and Hearing, 4:
Psycholinguistic Assessments of Language Processing 93–107.
in Aphasia. Hove, England: Psychology Press. Morgan-Jones, P., Colombage, E., McIntosh, D. and
Kertesz, A., Davidson, W., McCabe, P. et al. (2003). Ellis, P. (2014). Don’t Give Me Eggs that Bounce: 118
Primary progressive aphasia: Diagnosis, varieties, evo- Cracking Recipes for People with Alzheimer’s. Sydney,
lution. Journal of the International Neuropsychological Australia: Hammondcare.
Society, 9: 710–719. Muller, N and Wilson, B. (2008). Collaborative role
Kertesz, A., Jesso, S., Harciarek, M. et al. (2010). What is construction in a conversation with dementia: An
semantic dementia? A cohort study of diagnostic fea- application of systemic functional linguistics. Clinical
tures and clinical boundaries. Archives of Neurology, Linguistics and Phonetics, 22: 767–774.
67: 483–489. Murphy, J. and Oliver, T. (2013). The use of talking mats
Kessler, T., Croot, K. and Togher, L. (2001). Strategies to to support people with dementia and their carers to
facilitate communication with a person with demen- make decisions together. Health and Social Care in the
tia of the Alzheimer type. In Wilson, L. and Hewat, Community, 21: 171–180.
S. (eds.), Proceedings of the 2001 Speech Pathology Murphy, J., Tester, S., Hubbard, G. and Downs, M. (2005).
Australia National Conference, Melbourne, Australia, Enabling frail older people with a communication dif-
May 20–23, 2001, pp. 27–34. ficulty to express their views: The use of Talking Mats
Kindell, K., Sage, J., Keady, J. and Wilkinson, R. (2013). as an interview tool. Health and Social Care in the
Adapting to conversation with semantic dementia: Community, 13: 95–107.
Using enactment as a compensatory strategy in Nelson, P. T., Schmitt, F. A., Lin, Y. et al. (2011).
everyday social interaction. International Journal of Hippocampal sclerosis in advanced age: Clinical and
Language and Communication Disorders, 48: 497–507. pathological features. Brain, 134: 1506–1518.
Lemke, U. (2011). Hearing impairment in dementia – How Newhart, M., Davis, C., Kannan, V. et al. (2009). Therapy
to reconcile two intertwined challenges in diagnostic for naming deficits in two variants of primary progres-
screening. Audiology Research, 1 (e15): 58–60. sive aphasia. Aphasiology, 23: 823–834.
218 Dementia

Palmer, C., Adams, S., Bourgeois, M. et al. (1999). Savage, S., Piguet, O. and Hodges, J.R. (2014). Giving
Reduction in caregiver-identified problem behaviours words new life: Generalisation of word retraining out-
in patients with Alzheimer disease post hearing aid comes in semantic dementia. Journal of Alzheimer’s
fitting. Journal of Speech and Hearing Research, 42: Disease, 40: 309–317.
312–328. Savundranayagam, M.Y., Hummert, M.L. and
Petersen, R.C. and Negash, S. (2008). Mild cognitive Montgomery, R.J. (2005). Investigating the effects
impairment: An overview. CNS Spectrums, 13: 45–53. of communication problems on caregiver burden.
Piguet, O., Hornberger, M., Mioshi, E. and Hodges, J.R. The Journals of Gerontology Series B: Psychological
(2011). Behavioural-variant frontotemporal demen- Sciences and Social Sciences, 60: S48–S55.
tia: Diagnosis, clinical staging and management. The Siegel, E. and Wetherby, A. (2000). Nonsymbolic commu-
Lancet Neurology, 10: 162–172. nication. In Snell, M. and Brown, F. (eds.), Instruction of
Powell, A.L., Cummings, J.L., Hill, M.A. and Benson, D.F. Students with Severe Disabilities, Fifth Edition. Upper
(1988). Speech and language alterations in multi- Saddle River, NJ: Prentice Hall, pp. 409–451.
infarct dementia. Neurology, 38: 717–719. Smith, E.R., Broughton, M., Baker, R. et al. (2011).
Pressman, P.S. and Miller, B.L. (2014). Diagnosis and man- Memory and communication support in dementia:
agement of behavioral variant frontotemporal demen- Research based strategies for caregivers. International
tia. Biological Psychiatry, 75: 574–581. Psychogeriatrics, 23: 256–263.
Radanovic, M., Diniz, B.S., Mirandez, R.M. et al. (2009). Speech Pathology Australia (SPA) (2012). Submission 074
Verbal fluency in the detection of mild cognitive to the House of Representatives Standing Committee
impairment and Alzheimer’s disease among Brazilian on Health and Ageing: An Inquiry into Dementia Early
Portuguese speakers: The influence of education. Diagnosis and Intervention. Available at www.aphref.
International Psychogeriatrics, 21: 1081–1087. aph.gov.au-house-committee-haa-dementia-subs-
Ripich, D.N. and Wykle, M.L. (1996). Alzheimer’s Disease sub074-speech pathology australia-02 may 2012.pdf.
Communication Guide: The FOCUSED Programme Accessed 12 October 2014.
for Caregivers. San Antonio, CA: The Psychological Speech Pathology Australia (SPA) (2014). Submission to
Corporation. the Inquiry into the Prevalence of Different Types of
Ripich, D.N., Ziol, E., Fritsch, T. and Durand, E.J. Speech, Language and Communication Disorders
(2000). Training Alzheimer’s disease caregivers for and Speech Pathology Services in Australia: Senate
successful communication. Clinical Gerontologist, Community Affairs Reference Committee, February,
21: 37–56. 2014. Available at www.speechpathologyaustralia.
Rohrer, J.D. and Rosen, H.J. (2013). Neuroimaging in org.au/library/lobbying/20140221%20%20Senate%20
frontotemporal dementia. International Review of Inquiry%20Submission%20FINAL.pdf.
Psychiatry, 25: 221–229. Steinberg, B.A., Bieliauskas, L.A., Smith, G.E. and Ivnik,
Royal College of Speech and Language Therapists R.J. (2005). Mayo’s older Americans normative studies:
(RCSLT) (2014). Speech and Language Therapy Age-and IQ-adjusted norms for the trail-making test,
Provision for People with Dementia. London: RCSLT. the Stroop test, and MAE controlled oral word associa-
RCSLT Position Paper 2014. Available at http://www. tion test. The Clinical Neuropsychologist, 19: 329–377.
rcslt.org/members/publications/publications2/demen- Strauss, E., Sherman, E.M. and Spreen, O. (2006).
tia_position_paper2014. Accessed 6 October 2014 A Compendium of Neuropsychological Tests:
Sampson, E.L., Candy, B. and Jones, L. (2009). Enteral Administration, Norms, and Commentary, Third
Tube Feeding for Older People with Advanced Edition. New York, NY: Oxford University Press.
Dementia (Review). The Cochrane Collaboration. Stuart-Hamilton, I. and Rabbitt, P. (1997). Age related
Chichester, England: John Wiley and Sons, Ltd. decline in spelling ability: A link with fluid intelligence?
Santo Pietro, M.J. (2002). Training nursing assistants to Educational Gerontology, 23: 437–441.
communicate effectively with people with Alzheimer’s Taler, V. and Phillips, N.A. (2008). Language performance
disease: A call for action. Alzheimer’s Care Quarterly, in Alzheimer’s disease and mild cognitive impair-
3: 157–164. ment: A comparative review. Journal of Clinical and
Santo Pietro, M.J. and Boczko, F. (1997). Communication Experimental Neuropsychology, 30: 501–556.
Outcome Measure of Functional Independence Taylor, C. Kingma, R.M., Croot. K. and Nickels, L. (2009).
(COMFI Scale). Vero Beach, FL: Speech Bin. Speech pathology services for primary progressive
Santo Pietro, M.J. and Boczko, F. (2001). The breakfast aphasia: Exploring an emerging area of practice.
club. Alzheimer’s Care Quarterly, 2: 56–60. Aphasiology, 23: 161–174.
Sapolsky, D., Domoto-Reilly, K. and Dickerson, B.C. (2014). Testa, J.A., Ivnik, R.J., Boeve, B. et al. (2004). Confrontation
Use of the Progressive Aphasia Severity Scale (PASS) naming does not add incremental diagnostic util-
in monitoring speech and language status in PPA. ity in MCI and Alzheimer’s disease. Journal of the
Aphasiology, 28(8–9): 993–1003. International Neuropsychological Society, 10: 504–512.
Speech and language therapy in dementia assessment and management 219

Tomoeda, C.K. (2001). Comprehensive assessment for Watson, C.M., Chenery, H.J. and Carter, M.S. (1999). An
dementia: A necessity for differential diagnosis and analysis of trouble and repair in the natural conversa-
management. Seminars in Speech and Language, 22: tions of people with dementia of the Alzheimer’s type.
275–289. Aphasiology, 13: 195–218.
Torresi, K., Moorhouse, B. and Scott, A. (2012). Submission Weakley, A., Schmitter-Edgecombe, M. and Anderson,
098 to the House of Representatives Standing J. (2013). Analysis of verbal fluency ability in amnestic
Committee on Health and Ageing: An Inquiry into and non-amnestic mild cognitive impairment. Archives
Dementia Early Diagnosis and Intervention. Available at of Clinical Neuropsychology, 28: 721–731.
www.aphref.aph.gov.au-house-committee-haa-dementia- Wilshire, C.E., Lukkien, C.C. and Burmester, B.R.
subs-sub098 - kym torresi browyn moorhouse amanda (2014). The sentence production test for aphasia.
scott - 20 july 2012.pdf. Accessed 19 October 2014. Aphasiology, 28: 658–691.
Tsantali, E., Economidis, D. and Tsolaki, M. (2013). Could Wong, S.B., Anand, R., Chapman, S.B. et al. (2009).
language deficits really differentiate Mild Cognitive When nouns and verbs degrade: Facilitating
Impairment (MCI) from mild Alzheimer’s disease? ­communication in semantic dementia. Aphasiology,
Archives of Gerontology and Geriatrics, 57: 263–270. 23: 286–301.
Tsapkini, K., Frangakis, C., Gomez, Y. et al. (2014). World Health Organization (WHO) (2001). International
Augmentation of spelling therapy with transcranial Classification of Functioning, Disability and Health.
direct current stimulation in primary progressive apha- Available at www3.who.int/icf/icftemplate.cfm.
sia: Preliminary results and challenges. Aphasiology, 28: Accessed 15 October 2014.
1112–1130. Zabar, Y., Carson, K.A., Troncoso, J.C. and Kawas, C.H.
Volkmer, A. (2013). Assessment and Therapy for Language (1998). Dementia due to hippocampal sclerosis:
and Cognitive Communication Difficulties in Dementia Clinical features and comparison to Alzheimer’s dis-
and Other Progressive Diseases. Guildford, England: J ease. Neurology, 50: A59–A60.
and R Press Ltd.
Vuorinen, E., Laine, M. and Rinne, J. (2000). Common
pattern of language impairment in vascular dementia
and in Alzheimer’s disease. Alzheimer Disease and
Associated Disorders, 14: 81–86.
21
Role of the physiotherapist in the management
of dementia

SUE LORD AND LYNN ROCHESTER

the evidence for physiotherapy in dementia, we have referred


21.1 INTRODUCTION to current literature where appropriate.

Physiotherapy (and exercise) plays a key role in preserva-


tion of movement in people with cognitive impairment and
21.2 ROLE OF THE PHYSIOTHERAPIST
dementia, with evidence to suggest that exercise may also
be protective of future decline in memory. Over the past
decade, understanding of the role of cognition in motor Physiotherapy has its roots in movement science and views
control has increased with consequences for independent movement as central to health and well-being. The role of
and safe mobility and this knowledge provides a strong the physiotherapist is to maximize independence and par-
rationale for physiotherapy intervention. Recognition of the ticipation in work, family and society activities through
trajectory of cognitive impairment assists in understanding preservation of motor and cognitive functions. What is
the role of physiotherapy across the continuum, which is becoming clear increasingly is that physiotherapists should
adapted to suit personal factors such as age, ethnicity, pre- be involved in the management of people with cognitive
existing comorbidities and environment – all of which may impairment right at the very outset of signs, rather than
act as barriers or facilitators for the patient. restrict input to the later stages of the disease, which has
This chapter outlines physiotherapy treatment for people been the traditional approach. Often, the role of physiother-
with cognitive impairment and dementia and provides a apy is one of consultation with reference to external service-­
model to guide implementation of the same. First, we pro- providers and education is a key focus. Identification of
vide an overview of physiotherapy in dementia manage- services and personnel in the early stages is likely to include
ment and then outline specific physiotherapy interventions. gym referrals, personal trainers and community classes.
A clinical framework for physiotherapy is presented and a Over time, these will be supplanted by physiotherapy assis-
problem-oriented approach to key impairments is discussed. tants, dementia ‘buddies’, carers and support workers. In
The term ‘physiotherapy’ is extended to include ‘exercise’ the early stages there is less need for ‘hands-on’ physiother-
and ‘activity’ and the term ‘dementia’ is used throughout apy but this changes as expert handling and facilitation is
to denote Alzheimer’s disease (AD), Parkinson’s disease required to ensure optimal positioning, safe transfers and
with dementia (PDD), Lewy body dementia, frontotem- mobility and comfort. Behavioural changes, altered mood,
poral dementia and vascular dementia. General principles loss of motivation and poor compliance impact on the
of physiotherapy are broadly applicable to all dementias, receptivity of physiotherapy as disease progresses and its
however, we also highlight specific techniques. We consider utility is questioned. Priorities of management will lie else-
cognitive impairment along with a continuum of severity, where. Throughout the disease, physiotherapy must be rel-
from mild cognitive impairment to dementia and whilst it is evant and suited to an individual’s context and needs, skill
beyond the scope of this chapter to systematically examine and experience is required to optimize input. Figure 21.1

220
Role of the physiotherapist in the management of dementia 221

MCI/Mild Moderate Severe

Maintain physical Continue with MCI/mild Reduce carer burden


capacity aims and add Reduce falls risk
Strength Reduce carer Delay
Balance burden institutionalisation
Endurance Reduce falls risk Reduce secondary
Flexibility Manage risks of immobility
Maintain cognitive behavioural change Train carer/staff in
capacity effective moving and
Maintain mobility handling
Maintain independent
ADL
Maintain leisure
activity

Figure 21.1 Broad aims of physiotherapy across the stages of dementia.

outlines the broad aims of physiotherapy across the spec- outcomes have been reported for a range of gait outcomes
trum of cognitive decline and Figure 21.2 projects how these and mobility, strength and power, balance and activities
aims might be achieved. of daily living (for reviews see Pitkala et al., 2003; Forbes
et al., 2013). However, the translation from improvement in
impairment to gains in function and levels of activity is not
automatic (Chin et al., 2008) and physiotherapists need to
21.3 INTERVENTION APPROACHES be aware of personal, contextual and environmental factors
FOR DEMENTIA that impede translation of gains to daily functions. Early
intervention is likely to yield greater benefits before demen-
21.3.1 EXERCISE tia is established because the potential for motor learning
is greater and compensation more effective. Targeting gait
The mainstay of physiotherapy management is exercise, dysfunction and falls risk in mild cognitive impairment and
delivered in a variety of modes and combinations to suit the treating this aspect as a prodromal stage for dementia may
personal preference of the individual, the stages of demen- ultimately yield to greatest benefits.
tia and the context. The aims of exercise are to maintain
physical capacity, particularly, strength and balance, which 21.3.1.2 Exercise: prevention of secondary
are integral to effective locomotion. The benefits of exer- deconditioning and secondary
cise are threefold. First, exercise helps to maintain physi- age-related risk factors
cal and cognitive capacity to maintain/optimize physical
performance in activities of daily living (ADL), and thereby, The protective effect of sustained physical activity over
promote independence and quality of life. Second, exercise the life span is well recognized, even in older adults who
helps avoid the secondary consequences of decondition- participate in physical activity at a later stage in life.
­
ing associated with reduced mobility and a sedentary life- Guidelines advocate that people living with chronic dis-
style. Third, an emerging body of evidence suggests that in eases remain active to mitigate multi-system deconditioning
the initial stages of dementia (mild cognitive impairment brought about by increased sedentary behavior (Thompson
[MCI] and mild dementia) exercise may have an important et al., 2003), which is an independent risk factor for poor
role to play to maintain cognitive function or capacity. Each health outcomes. The definition of ‘activity’ in the guidelines
of these features is considered and outlined in detail in the is broad and includes walking, dancing, exercise classes,
following. circuit training etc. Counter-intuitively, sedentary behav-
iour and activity yield quite different cellular and molecular
21.3.1.1 Exercise: maintenance of motor responses. One is not the inverse of the other and the impact
performance of sedentary lifestyle on vascular integrity, cardiovascular
health and diabetes has been reported often (Manns, 2012).
Almost undisputedly, exercise improves physical and Implications from this body of research are that preservation
cognitive performance in older adults. Most exercise pro- of activity is critical for as long as possible. Beyond subtle
grammes, whether of low or high intensity, general or spe- physiological responses, increased sedentary behaviour also
cific, are able to show a positive physiological response. For impacts on muscle tissue and loss of joint integrity. Over
people with cognitive impairment or dementia, improved time, there is an inability to carry out previously rehearsed
222 Dementia

Disease stage Possible interventions


Activities Exercise Functional training Compensatory strategies
MCI • Dancing • Strength • Transfers • Cueing
• Endurance • Complex sequences • Prompting
• Balance and coordination • Diary reminders
• Flexibility
• Complex motor/cognitive
training
Mild • Computer games • As above • As above • As above
– with family
Moderate • Walking groups • As above with adaptations • Training carers to cue and
and supervision (e.g. seated prompt the patient
exercise for postural
instability)
Severe • Gym or NA • Training care staff needed • Training care staff needed in
community in moving and handling moving and handling the
exercise groups the patient patient

Figure 21.2 Examples of possible interventions to address physiotherapy aims with respect to the stages of dementia.
Abbreviation: MCI, mild cognitive impairment.

sequences of movement simply because the action is not between higher levels of physical activity and reduced risk
repeated often enough. of cognitive decline and dementia, with a recent meta-anal-
Several questions remain unanswered with respect to ysis reporting an 18% reduction in risk of cognitive decline
exercise, most notably the dosage, intensity and timing of and dementia due to physical activity (Blondell et al., 2014).
it. Provision of a standardized exercise protocol is almost There is evidence to suggest cognitive retraining alone has
impossible given the range of variables that impact the out- a positive impact on selected features of cognition in cog-
come of their implementation on the patient. As a guide, nitively impaired older adults (Forbes et al., 2013) and a
exercise three times a week for 30 minutes, over 6–8 weeks review of 15 studies reported a benefit of cognitive train-
should yield benefits and if improvement is not evident after ing for people with dementia over and above any medica-
this, then it is unlikely to be worth extending the interven- tion effect (Woods et al., 2012). More recently, arguments
tion. Securing a behavioural change is the ultimate goal of have been put forward for combining motor and cognitive
any exercise intervention, but this is ultimately challenging retraining strategies (see the following). Given the close
for people with cognitive impairment who may also have relationship between cognition and gait and the compen-
associated mood disorders and reduced motivation. The satory role of cognition in maintaining gait performance
potential to ensure carry over from exercise to daily, func- improved benefits seem plausible, however, further studies
tional tasks must be recognized and encouraged, so that are required to evaluate single component versus multicom-
regular practise ensues. Intensity must be sufficient to chal- ponent interventions.
lenge motor performance. For example, although exercise
is overall protective for first fall in dementia (Allan et al.,
21.3.2 COMPENSATORY STRATEGIES
2009), Sherrington (Sherrington et al., 2008) reported that
it was not effective in reducing falls in people with demen- Compensatory strategies develop in response to motor
tia who were living in care homes, possibly because the impairment and reflect versatility of the motor system to
exercises were mostly in sitting position, which has limited adapt effectively (Shumway-Cook and Woollacott, 2007).
effect on postural mechanisms. With respect to timing, the Use of compensatory strategies to bypass deficits in motor
earlier the intervention, the greater the gains, which in turn control is a feature of this versatility. However, timing of
are more likely to be sustained in future. Physiotherapy is introduction of compensatory strategies is critical because
often administered too late, when deconditioning and attri- once the new motor pattern is established it will become the
tion of habitual motor patterns impair function. default pattern and this expediency does not always produce
the most efficient movement. Movement patterns become
21.3.1.3 Exercise: protection of ingrained often prior to retraining or relearning has been
cognitive function attempted. A simple illustration is the use of hand support to
attain sit to stand/stand to sit, which becomes increasingly
Guidelines for healthy ageing emphatically include exer- difficult with age but is amenable to ­retraining. The goal is
cise as a protective agent for cognitive decline, which has achieved at the expense of decreased concentric and e­ ccentric
been extended to include people with cognitive impairment muscle work, abnormal postural control ­strategies, reduced
and dementia. Longitudinal studies suggest a causal link preparedness for safe walking. People with ­dementia show
Role of the physiotherapist in the management of dementia 223

retained capacity for implicit motor learning ­(van Halteren- attention) is used (Verlinden et al., 2014). A seminal study
van Tilborg et al., 2007) and also through explicit ­learning by Lundin-Olsson (1997) showed an increased risk of falls
techniques, via observational learning and ­learning through for older, cognitively impaired adults who stopped walk-
guidance (van Tilborg et al., 2011). Cognition is used to ing whilst talking compared with those people who could
enhance use of ­compensatory strategies and is used to good successfully carry out both tasks simultaneously. Some
effect via attentional ­mechanisms or via use of overt cues or features of motor control are selectively associated with
prompts. Evidence supports the use of visual and auditory cognitive impairment. There is, for example, a functional
cues to enhance gait in Parkinson’s disease (PD) (Nieuwboer relationship between the hippocampus, which degenerates
et al., 2007) and has the potential to be ­effective even as early in amnestic pathology (evidenced by loss of short-term
­cognitive i­mpairment advances – especially if ­cueing skills memory) and spatial disorientation via visual, vestibular
are embedded and taught earlier. Identifying the thresh- and proprioceptive loss, which leads to gait disturbance
old of cognitive impairment from which learning can be (Scherder et al., 2007). Because of this close relationship,
achieved is difficult and compounded by associated features gait has predictive value for future cognitive decline, as evi-
such as reduced motivation, anxiety, depression and reduced denced by large community cohort studies and a threshold
self-efficacy (Burn et al., 2006). for pathological gait (gait speed below 1 m/s) has been iden-
tified from the predictive models (Studenski et al., 2011).
21.3.3 NOVEL INTERVENTIONS Recent evidence supports the sensitivity of a motoric risk
syndrome (combined motor and cognitive outcomes) to
Current interventions reflect the contemporary view predict future cognitive decline (Verghese et al., 2014). The
­outlined in the following that motor control (especially relative contribution of cognitive function to gait, balance
gait and balance) is underpinned by neural substrates, and falls in dementia is likely to be higher than contribu-
which are partly shared by cognitive networks (Thom tion from other neural systems such as sensorimotor func-
and Clare, 2011). Approaches vary from specific dual task tion, however, this has not been examined yet. Figure 21.3
retraining programmes to general activities, with a recent contrasts traditional and current views of the association
­systematic review reporting benefit of a combined approach between motor and cognitive function and its consequences
(Lee et al., 2015). Schwenk reported greater improve- and provides a theoretical basis to development of novel
ment in gait p ­ erformance from cognitive and gait tasks interventions that challenge motor and cognitive systems
(dual-task t­raining) compared to a programme of exer- simultaneously (see the following).
cises only (Schwenk et al., 2010). Dance is gaining momen-
tum as an intervention (Kattenstroth et al., 2013) and
although, ­evidence of efficacy for both dance and music
21.5 A PROBLEM-ORIENTED
as therapy in dementia is lacking (Vink et al., 2004) there
is some ­support for a positive impact on behaviour and
APPROACH TO PHYSIOTHERAPY
­socialization (Guzman-Garcia et al., 2013). Technology is
used to good effect. In a recent review of physical and cog- We consider the main problems that physiotherapists
nitive ­interventions in ageing, Bamidis et al. (2014) argued address: gait and postural control, falls, transfers and seat-
for the use of exergaming (computer games that require ing. We take an evidence-based approach to inform man-
intense physical activity), which may be appropriate in the agement of these problems and consider the underlying
very early stages of cognitive decline. mechanisms of motor impairment in cognitive decline.
We then complete this section by considering some of the
­complications that arise with dementia that are likely to
21.4 A CLINICAL FRAMEWORK FOR impact physiotherapy. For a summary of selected resources,
PHYSIOTHERAPY: INTERPLAY see Table 21.1.
BETWEEN COGNITIVE AND
MOTOR SYSTEMS
21.6 GAIT AND POSTURAL CONTROL
Recent evidence in the field of motor control provides a
strong rationale for physiotherapy intervention. Neural Gait impairment is common in older adults, with 35% pre-
systems that underpin cognitive function are also impli- senting a clinically observable deficit (Studenski, 2012).
cated in the control of movement, thus, strengthening the Global features of gait (such as gait speed) are sensitive to all
link between cognition, gait and falls in dementia. This ‘top dementias and also sensitive to severity of cognitive impair-
down’ control is evident even in cognitively healthy older ment. Goldman et al. (1999) reported a slower gait veloc-
adults (Lord et al., 2014). Gait is not an automatic motor ity for people with AD and moderate dementia (Clinical
task, independent of cognition, as previously believed. Use Dementia Rating (CDR) Scale: CDR stage 1) compared with
of a dual task research paradigm (e.g. walking whilst reciting mild dementia (CDR stage 0.5). The prevalence and sever-
numbers) reveals the extent to which cognition (primarily ity of gait disorders is also dependent upon the dementia
224 Dementia

(a) Traditional view

Slow gait velocity


Instability Falls - fractures

MCI
Cognitive impairment Dementia

(b) Alternative, emerging view

Slow gait velocity


Instability Falls - fractures

Cognitive impairment MCI


Executive function–working memory
Dementia

Figure 21.3 Traditional and contemporary views of the decline in cognition and mobility and their consequences.
(a) Traditional view. (b) Alternative, emerging view. (From Montero-Odasso, M. et al.: Gait and cognition: A comple-
mentary approach to understanding brain function and the risk of falling. Journal of the American Geriatrics Society, 60,
2127–2136, 2012. Copyright Wiley-VCH Verlag GmbH & Co. KGaA. Reproduced with permission.)

Table 21.1 Resources for physiotherapists


Reviews Dementia and physical therapy (Blondell, et al., 2014)
Cognitive stimulation to improve cognitive functioning in people with dementia (Woods et al., 2012)
Exercise programmes for people with dementia (Forbes, et al., 2013)
Music therapy for people with dementia (Vink et al., 2004)
Effects of combined cognitive and exercise interventions on cognition in older adults with and without
cognitive impairment: A systematic review (Law et al., 2014)
Efficacy of physical exercise intervention on mobility and physical functioning in older people with
dementia: A systematic review (Pitkala et al., 2013)
Educational CSP Physiotherapy Works: Dementia Care
resources http://www.csp.org.uk/publications/physiotherapy-works-dementia-care
Living well with dementia: A national dementia strategy
https://www.gov.uk/government/publications/living-well-with-dementia-a-national-dementia-strategy

subtype (Allan et al., 2005) (see Table 21.2), with it being the associated with frontal–subcortical lesions and atrophy
greatest in Lewy body dementia (LBD), especially, PDD and and hypometabolism of the hippocampus (Annweiler et al.,
the lowest in AD. Other features of gait are specific to under- 2012). It is important to keep these features in mind when
lying pathophysiology and can be used to discriminate state assessing gait and using gait as a tool for differential diag-
of diseases. For example, reduced step length and start hesi- nosis of different forms of dementia. Physiotherapists have a
tation is a sign of a frontally mediated ‘cautious’ gait (Nutt key role to play in informing diagnosis and disease progres-
et al., 1993), whereas increased step asymmetry and reduced sion in this way.
step length is an indicator of PD gait. However, the aetiol- Although physiotherapy has a positive effect on a
ogy of gait disorder in 15%–30% of older adults diagnosed range of balance and gait deficits in dementia (Shaw et al.,
with gait impairment is unclear (Kafri et al., 2013) and 2003; Christofoletti et al., 2008), the preservation of ‘nor-
the broad classification ‘high-level gait disorder’ (HLGD), mal’ gait and amelioration of specific gait deficit is not a
which exemplifies a shared gait and cognitive pathology, is primary focus for intervention even in the early stages.
most often applied. An added complication to dementia is Instead, a more global approach is required. The empha-
the presence of white matter lesions (WML) and cerebro- sis is on functional mobility such as encouraging out-
vascular disease (CVD), both of which contribute further door walking or community exercise classes. As a result
to gait disturbance, balance and falls (Inzitari et al., 2013). of these interventions, improvement in at least some gait
A systematic review found that in AD gait impairment was characteristics is likely to occur without them being tar-
associated with a higher burden of WML with changes geted at, specifically.
Role of the physiotherapist in the management of dementia 225

Table 21.2 Annual prevalence and incidence of falls (falls/1000 persons) and prevalence of gait and balance disorders in
different dementia subtypes
Controls AD VAD DLB PDD PD
Prevalence (%) 36 47 47 77 90 61
Incidence 1,023 2,486 3,135 9,087 19,000 4,617
(falls/1000
people)
Gait disorder, 3/42 (7) 10/40 (25) 31/39 (79) 43/46 (93) 24/32 (75) 20/46 (43)
N (%)
Source: Allan, L.M. et al., PloS One, 4, e5521, 2009; Allan, L.M. et al., Journal of the American Geriatrics Society, 53, 1681–1687, 2005.)

Gait and balance disturbances are important risk Parkinson’s disease are identified as risk factors for falls in
factors for falls with a higher prevalence in demen- patients with dementia. A second risk factor relates to medi-
tia compared with non-demented older adults (Tinetti cation with polypharmacy, identified as a risk factor for falls
and Williams, 1998; Shaw, 2002; Allan et al., 2009). in older adults (Neutel et al., 2002). Psychotropic medica-
Preservation of effective balance is critical but challeng- tions increase somnolence, confusion and loss of awareness
ing. Use of tai chi and other balance-specific interven- of surroundings and antidepressants are also associated
tions have been reported to improve postural control in with falls risk. Treatment of falls in dementia is challeng-
older adults (Logghe et al., 2010), although to our knowl- ing. There is no convincing evidence to mitigate falls risk
edge this has not been replicated in people with cogni- in dementia, however, current evidence points to the need
tive decline and dementia. Improvement in balance, gait for targeted selection of participants and individualized
and functional mobility is likely to be a positive spin-off therapies. A recent pilot study reported 32.6% reduction in
from general exercise and activity and maximum gains falls and decreased agitation in people with dementia living
may be achieved by taking a broader approach. What is in a residential care home through the use of personalized
evident is the added burden apparently placed by gait and interventions based on behavioural and cognitive profiles
balance disorders on falls risk in dementia, highlighting (Bharwani et al., 2012).
the complex relationship between gait, balance, cognition
and falls.

21.8 TRANSFERS AND POSITIONING

21.7 FALLS As disease progresses even simple functional tasks such as


sit to stand, bed mobility and transfers from bed to upright
The consequence of falls in older adults with dementia is standing position is challenging. Independent mobility
drastic and includes an increased risk of serious injury effectively ceases and supervision and assistance is required
(Kallin et al., 2005), institutionalization (Morris et al., 1987) to facilitate movement. Loss of movement is due to cognitive
and mortality (Morrison et al., 2000). Remaining mobile rather than pure motor decline. Effective movement is more
and free from falls is a therapeutic priority in older adults likely to occur if complex sequences of movement are bro-
and those with dementia and understanding falls in demen- ken down into a series of sub-movements, which are carried
tia is of critical importance to mitigate the burden caused out in a consistent manner by all staff handling the patient,
not only to the patient, but also the carer (Lowery et al., with repetition critical to success. In the earlier stages, men-
2000). People with dementia are two to three times more tal preparation of movements prior to their execution may
likely to fall than age-matched healthy counterparts, with also be helpful. Physiotherapy in the later stages is centred
institutionalized adults also at a higher risk compared with on ensuring safe mobility and 24-hour comfort via optimal
community living adults (Kropelin et al., 2013). Figures positioning. Vital to successful physiotherapy at this stage
vary, but overall suggests a prevalence of around 40% for is input from multidisciplinary team members who provide
community dwelling adults (Allan et al., 2009) and 60% expertise and options for seating. Contoured chairs and
for people living in residential care (Eriksson et al., 2008). low beds may be useful and semi-recumbent positioning
Falls risk is highly dependent upon the type of dementia, in a wheelchair may promote better head control and pos-
with highest incidence and prevalence occurring in people ture. A sensitive approach is required to balance the need
with dementia associated with Lewy body dementias (LBD for restraint with comfort and freedom, with evidence to
and PDD) (Table 21.2) (Allan et al., 2009). Fall-related inju- suggest a key role of education in appropriate application of
ries are also greater in LBD while Lewy body disease and restraint in late-stage dementia (Testad et al., 2005).
226 Dementia

REFERENCES
21.9 COMPLICATIONS IMPACTING ON
PHYSIOTHERAPY Allan, L.M., Ballard, C.G., Burn, D.J. et al. (2005).
Prevalence and severity of gait disorders in
Behavioural disturbances in the later stages are challenging Alzheimer’s and non-Alzheimer’s dementias. Journal of
to work with. Aggression, frustration, diminished commu- the American Geriatrics Society, 53 (10): 1681–1687.
nication and mood disturbances (depression, anxiety) are Allan, L.M., Ballard, C.G., Rowan, E.N. et al. (2009).
problematic to deal with and can result in futile encoun- Incidence and prediction of falls in dementia:
ters. In future, attention will be paid to how physiotherapy A ­prospective study in older people. PloS One,
is administered – choosing a suitable environment, use of 4 (5): e5521.
language, command and facilitation. Identifying sources Annweiler, C., Beauchet, O., Celle, S. et al. (2012).
of discomfort and pain is also challenging and it is not Contribution of brain imaging to the understanding
always apparent how much to encourage a movement when of gait disorders in Alzheimer’s disease: A systematic
it appears to cause distress. Use of a standardized assess- review. American Journal of Alzheimer’s Disease and
ment of pain and discomfort (Kovach et al., 2001) may help Other Dementias, 27 (6): 371–380.
clarify the situation, but this remains an important feature Bamidis, P.D., Vivas, A.B., Styliadis, C. et al. (2014). A
of dementia management. review of physical and cognitive interventions in aging.
Neuroscince and Biobehavioral Reviews, 44: 206–220.
Bharwani, G., Parikh, P.J., Lawhorne, L.W. et al. (2012).
Individualized behavior management program for
Alzheimer’s/dementia residents using behavior-based
21.10 MEASUREMENT ergonomic therapies. American Journal of Alzheimer’s
Disease and Other Dementias, 27 (3): 188–195.
Because dementia is a progressive disorder, use of out- Blondell, S.J., Hammersley-Mather, R. and Veerman, J.L.
come measures is not routine. It may be required at (2014). Does physical activity prevent cognitive decline
discrete time points, for example, to monitor change in and dementia?: A systematic review and meta-analysis
performance over a predefined period of time for a spe- of longitudinal studies. BMC Public Health, 14: 510.
cific reason, or to measure response to an intervention. Burn, D.J., Rowan, E.N., Allan, L.M. et al. (2006). Motor
Estimation of ‘true change’ over and above measurement subtype and cognitive decline in Parkinson’s disease,
error is difficult, given the natural course of the disease. Parkinson’s disease with dementia, and dementia with
The repertoire of standardized, generic measures com- Lewy bodies. Journal of Neurology, Neurosurgery, and
monly used in physiotherapy can be applied to people Psychiatry, 77 (5): 585–589.
with dementia, such as gait and balance measures, func- Chin, A.P.M.J., van Uffelen, J.G., Riphagen, I. et al. (2008).
tional mobility outcomes, activities of daily living scales The functional effects of physical exercise training
etc. Measurement at the level of activity and participation in frail older people: A systematic review. Sports
rather than impairment is advised. Measurement of carer Medicine, 38 (9): 781–793.
burden may be appropriate alongside quality of life mea- Christofoletti, G., Oliani, M.M., Gobbi, S. et al. (2008).
sures. If the intervention focusses on improving cogni- A controlled clinical trial on the effects of motor inter-
tive performance, measurement of cognitive function is vention on balance and cognition in institutionalized
relevant. elderly patients with dementia. Clinical Rehabilitation,
22 (7): 618–626.
Eriksson, S., Gustafson, Y. and Lundin-Olsson, L. (2008).
Risk factors for falls in people with and without a
diagnose of dementia living in residential care facili-
21.11 SUMMARY ties: A prospective study. Archives of Gerontology and
Geriatrics, 46 (3): 293–306.
Emerging evidence supports physiotherapy in the man- Forbes, D., Thiessen, E.J., Blake, C.M. et al. (2013). Exercise
agement of dementia throughout the disease process with programs for people with dementia. The Cochrane
research to support efficacy even in advanced stages of the Database of Systematic Reviews, 12: CD006489.
disease. Physiotherapy is adapted to suit the individual at all Goldman, W.P., Baty, J.D., Buckles, V.D. et al. (1999). Motor
stages of dementia with a strong focus on maintenance of dysfunction in mildly demented AD individuals without
physical activity in the early stages where exercise is consid- extrapyramidal signs. Neurology, 53 (5): 956–962.
ered to have a neuroprotective effect. The role of the physio- Guzman-Garcia, A., Hughes, J.C., James, I.A. et al. (2013).
therapist throughout the disease is mostly consultative and Dancing as a psychosocial intervention in care homes:
effective liaison with all personnel involved in the care of A systematic review of the literature. International
the person with dementia is essential. Journal of Geriatric Psychiatry, 28 (9): 914–924.
Role of the physiotherapist in the management of dementia 227

Inzitari, M., Gine-Garriga, M., Martinez, B. et al. (2013). Manns, P. (2012). In people aged over 45, increased time
Cerebrovascular disease and gait and balance spent sitting daily is associated with increased risk of
impairment in mild to moderate Alzheimer’s disease. all-cause mortality independent of physical activity
The Journal of Nutrition, Health and Aging, 17 (1): level. Evidence-Based Nursing, 15 (4): 120–121.
45–48. Montero-Odasso, M., Verghese, J., Beauchet, O. et al.
Kafri, M., Sasson, E., Assaf, Y. et al. (2013). High-level (2012). Gait and cognition: A complementary approach
gait disorder: Associations with specific white to understanding brain function and the risk of falling.
matter changes observed on advanced diffu- Journal of the American Geriatrics Society, 60 (11):
sion ­imaging. Journal of Neuroimaging: Official 2127–2136.
Journal of the American Society of Neuroimaging, Morris, J.C., Rubin, E.H., Morris, E.J. et al. (1987). Senile
23 (1): 39–46. dementia of the Alzheimer’s type: An important risk
Kallin, K., Gustafson, Y., Sandman, P.O. et al. (2005). factor for serious falls. Journal of Gerontology, 42 (4):
Factors associated with falls among older, cogni- 412–417.
tively impaired people in geriatric care settings: Morrison, R.S. and Siu, A.L. (2000). Mortality from pneu-
A population-based study. The American Journal of monia and hip fractures in patients with advanced
Geriatric Psychiatry: Official Journal of The American dementia. The Journal of the American Medical
Association for Geriatric Psychiatry, 13 (6): 501–509. Association, 284 (19):2447–2448.
Kattenstroth, J.C., Kalisch, T., Holt, S. et al. (2013). Six Neutel, C.I., Perry, S. and Maxwell, C. (2002). Medication
months of dance intervention enhances postural, sen- use and risk of falls. Pharmacoepidemiology and Drug
sorimotor, and cognitive performance in elderly with- Safety, 11 (2): 97–104.
out affecting cardio-respiratory functions. Frontiers in Nieuwboer, A., Kwakkel, G., Rochester, L. et al. (2007).
Aging Neuroscience, 5: 5. Cueing training in the home improves gait-related
Kovach, C.R., Noonan, P.E., Griffie, J. et al. (2001). Use of mobility in Parkinson’s disease: The RESCUE trial.
the assessment of discomfort in dementia protocol. Journal of Neurology, Neurosurgery, and Psychiatry, 78
Applied Nursing Research, 14 (4): 193–200. (2): 134–140.
Kropelin, T.F., Neyens, J.C., Halfens, R.J. et al. (2013). Nutt, J.G., Marsden, C.D. and Thompson, P.D. (1993).
Fall determinants in older long-term care residents Human walking and higher-level gait disorders, par-
with dementia: A systematic review. International ticularly in the elderly. Neurology, 43 (2): 268–279.
Psychogeriatrics/IPA, 25 (4): 549–563. Pitkala, K., Savikko, N., Poysti, M. et al. (2013). Efficacy of
Law, L.L., Barnett, F., Yau, M.K. et al. (2014). Effects of physical exercise intervention on mobility and physical
combined cognitive and exercise interventions on functioning in older people with dementia: A system-
cognition in older adults with and without cognitive atic review. Experimental Gerontology, 48 (1): 85–93.
impairment: A systematic review. Ageing Research Scherder, E., Eggermont, L., Swaab, D. et al. (2007). Gait
Reviews, 15: 61–75. in ageing and associated dementias; Its relation-
Lee, J.S., Kim, C.G., Seo, T.B. et al. (2015). Effects of ship with cognition. Neuroscience and Biobehavioral
8-week combined training on body composition, Reviews, 31 (4): 485–497.
isokinetic strength, and cardiovascular disease risk fac- Schwenk, M., Zieschang, T., Oster, P. et al. (2010). Dual-
tors in older women. Aging Clinical and Experimental task performances can be improved in patients with
Research, 27: 179–186. dementia: A randomized controlled trial. Neurology,
Logghe I.H., Verhagen A.P., Rademaker A.C. et al. (2010). 74 (24): 1961–1968.
The effects of Tai Chi on fall prevention, fear of fall- Shaw, F.E. (2002). Falls in cognitive impairment and
ing and balance in older people: A meta-analysis. dementia. Clinics in Geriatric Medicine, 18 (2):
Preventive Medicine;51(3–4):222–227. 159–173.
Lord, S., Galna, B., Coleman, S. et al. (2014). Cognition and Shaw, F.E., Bond, J., Richardson, D.A. et al. (2003).
gait show a selective pattern of association dominated Multifactorial intervention after a fall in older people
by phenotype in incident Parkinson’s disease. Frontiers with cognitive impairment and dementia presenting to
in Aging Neuroscience, 6 (249): 1–9. the accident and emergency department: Randomised
Lowery, K., Mynt, P., Aisbett, J. et al. (2000). Depression controlled trial. BMJ, 326 (7380): 73.
in the carers of dementia sufferers: A comparison Sherrington, C., Whitney, J.C., Lord, S.R. et al. (2008).
of the carers of patients suffering from dementia Effective exercise for the prevention of falls: A system-
with Lewy bodies and the carers of patients with atic review and meta-analysis. Journal of the American
Alzheimer’s disease. Journal of Affective Disorders, Geriatrics Society, 56 (12): 2234–2243.
59 (1): 61–65. Shumway-Cook, A. and Woollacott, M.H. (2007). Motor
Lundin-Olsson, L., Nyberg, L. and Gustafson, Y. (1997). Control: Translating Research into Clinical Practice,
“Stops walking when talking” as a predictor of falls in Third Edition. Baltimore, MD: Lippincott Williams &
elderly people. Lancet, 349 (9052): 617. Wilkins.
228 Dementia

Studenski, S., Perera, S., Patel, K. et al. (2011). Gait older persons. The Journals of Gerontology Series
speed and survival in older adults. The Journal of the A, Biological Sciences and Medical Sciences, 53 (2):
American Medical Association, 305 (1): 50–58. M112–M119.
Studenski, S.A. (2012). Gait speed in hospitalized older van Halteren-van Tilborg, I.A., Scherder, E.J., Hulstijn,
people: Comment on “assessing gait speed in acutely W. (2007). Motor-skill learning in Alzheimer’s dis-
ill older patients admitted to an acute care for elders ease: A review with an eye to the clinical practice.
hospital unit”. Archives of Internal Medicine, 172 (4): Neuropsychology Review, 17 (3): 203–212.
358–359. van Tilborg, I.A., Kessels, R.P. and Hulstijn, W. (2011).
Testad, I., Aasland, A.M. and Aarsland, D. (2005). The Learning by observation and guidance in patients with
effect of staff training on the use of restraint in Alzheimer’s dementia. NeuroRehabilitation, 29 (3):
dementia: A single-blind randomised controlled trial. 295–304.
International Journal of Geriatric Psychiatry, 20 (6): Verghese, J., Ayers, E., Barzilai, N. et al. (2014). Motoric
587–590. cognitive risk syndrome: Multicenter incidence study.
Thom, J.M. and Clare, L. (2011). Rationale for combined Neurology, 83: 2278–2284.
exercise and cognition-focused interventions to Verlinden, V.J., van der Geest, J.N., Hofman, A. et al.
improve functional independence in people with (2014). Cognition and gait show a distinct pattern of
dementia. Gerontology, 57 (3): 265–275. association in the general population. Alzheimer’s and
Thompson, P.D., Buchner, D., Pina, I.L. et al. (2003). Dementia: The Journal of the Alzheimer’s Association,
Exercise and physical activity in the preven- 10 (3): 328–335.
tion and treatment of atherosclerotic cardiovas- Vink, A.C., Birks, J.S., Bruinsma, M.S. et al. (2004). Music
cular disease: A statement from the Council on therapy for people with dementia. The Cochrane
Clinical Cardiology (Subcommittee on Exercise, Database of Systematic Reviews, 3: CD003477.
Rehabilitation, and Prevention) and the Council Woods, B., Aguirre, E., Spector, A.E. et al. (2012).
on Nutrition, Physical Activity, and Metabolism Cognitive stimulation to improve cognitive functioning
(Subcommittee on Physical Activity). Circulation, 107 in people with dementia. The Cochrane Database of
(24): 3109–3116. Systematic Reviews, 2: CD005562.
Tinetti, M.E. and Williams, C.S. (1998). The effect of falls
and fall injuries on functioning in community-dwelling
22
Therapeutic effects of music in persons with
dementia

LINDA A. GERDNER AND RUTH REMINGTON

Awake, arise, listen to the song of life … At first all may seem confusion … but wait, strive to catch the deeper melody, for the
song its there.

Albert J. Atkins
A Song of Life, 1905

22.1 BEHAVIOURAL AND 22.2 CLASSICAL MUSIC OR MUSIC


PSYCHOLOGICAL SYMPTOMS BELIEVED TO BE CALMING/
OF DEMENTIA RELAXING

The presence of agitation or behavioural and psychologi- Calming music (CM) has been shown to reduce agitation
cal symptoms has been reported as high as 90% in persons in nursing home residents (Remington, 2002; Hicks-Moore,
with Alzheimer’s disease or related dementias (ADRD) 2005). The response to calming or relaxing music is related
(Fernández et al., 2010). Agitation interferes with care to the qualities of the musical sound that produce a cogni-
delivery and social interaction, ultimately having a nega- tive and physiological response in the participant, whereas
tive impact on the person’s quality of life (Léger et al., 2002; familiar music relies on the emotional response to the music
Sloane et al., 2004; Samus et al., 2005). Management of these to achieve the effect (Remington, 2002). In research, music
behaviours has been identified as a major stressor to health- chosen for its calming qualities has been referred to as an
care professionals (Brodaty et al., 2003). For this reason experimenter-centred approach, minimizing response
much of the research that has evaluated the effects of music bias in studies (Elliot et al., 2011). Characteristics of music
in persons with dementia (PWD) has focused on agitation that have been shown to be relaxing include a slow tempo,
as the primary outcome variable. 80–100 beats per minute (Elliot et al., 2011). It has also been
The presentation of music can be grouped into the fol- suggested that a tempo less than the resting heart rate is
lowing three categories: classical music or music believed more calming (Watkins, 1997), whereas a faster tempo
to be calming or relaxing, individualized or preferred heightens arousal (Ziv and Dolev, 2013). A constant, subtle
music, and music activities implemented by a certified rhythm with a regular beat played at a constant volume and
music therapist. in a major key was found to be relaxing (Elliot et al., 2011).

229
230 Dementia

Piano and strings are more calming than percussion or intervention and again 1 hour following the intervention.
brass instrumentation. Vocals are more likely to induce Findings report a statistically significant reduction in agi-
memories and arousal (Remington, 2002; Ziv and Dolev, tated behaviours across all three experimental groups when
2013). compared with the controls.
Goddaer and Abraham (1994) hypothesized that ‘relax- Using a repeated measures design, Thompson et al.
ing’ music would buffer the ‘general noise level’ found in (2005) compared the effect of music (Vivaldi’s Four Seasons,
dining rooms in long-term care facilities, thus exerting a Winter) on language fluency in healthy older adults (n = 16)
calming effect that would reduce the frequency of agitated and older adults with dementia (n = 16). Subjects were asked
behaviours in 29 persons with severe cognitive impairment. to name as many examples of items in a category as they
Baseline data were collected during week 1. Music was could within 1 minute. Each subject was tested both with
played in the dining room daily during the noon meal in music playing and without. Both the healthy subjects and
week 2. Music was removed during week 3 and reintroduced the subjects with dementia demonstrated a similar small
during week 4. A modified version of the Cohen-Mansfield but significant improvement in performance when music
Agitation inventory (CMAI) was used to measure the was played.
dependent variable. Significant reductions were observed A reduction in agitation during mealtime was reported
on the cumulative incidence of total agitated behaviours in a quasi-experimental study involving 33 residents with
(63.4%) as well as the cumulative incidence of physically dementia in a special care unit. Residents were observed for
nonaggressive behaviours (56.3%) and verbally agitated the number of agitated behaviours exhibited during the eve-
behaviours (74.5%). ning meal for 4 weeks. During weeks 1 and 3 no music was
The findings of this study were replicated using a smaller played and during weeks 2 and 4 CM was played. The inci-
sample of subjects (n = 10) residing in a special care unit dence of agitated behaviours was decreased in weeks when
(Denny, 1997). Classical music was played from 11:45 am music was played compared with weeks when no music was
to 1:15 pm daily during weeks 2 and 4 in the dining room played (Hicks-Moore, 2005).
during the noon meal. A modified version of the CMAI was In an experimental study of the effect of a group music
used to measure the dependent variable. intervention on agitated behaviours, Lin et al. (2011) found
Tabloski et al. (1995) used a quasi-experimental design a significant decrease in total agitation in the treatment
with a convenience sample of 20 cognitively impaired nurs- group. One hundred and four residents with dementia par-
ing home residents to measure the effects of Kobialka’s ticipated in the study. The treatment group participated in
recording of Pachelbel’s Canon in D on the frequency of a 30-minute group music intervention twice a week for 6
agitation. The dependent variable was measured using the weeks while the control group continued their usual daily
Agitated Behavior Scale (Corrigan, 1989). Agitation was activities. The decrease in agitation was seen in each of
assessed 15 minutes prior to the intervention, during the the four categories of agitated behaviours; physically non-
15-minute presentation of music and throughout the 15 aggressive, physically aggressive, verbally non-aggressive
minutes following the presentation of music. The inter- and physically aggressive. The effect of the intervention was
vention and assessment process were repeated approxi- sustained at 1 month after the intervention (Lin et al., 2011).
mately 1 week later. Repeated measure analysis of variance The CMAI or a modified CMAI was used to measure agi-
(R-ANOVA) revealed a statistically significant reduction in tated behaviours in most of the music intervention studies.
agitated behaviours during (p < 0.01) and after the music The instrument and modification have demonstrated reli-
intervention (p < 0.05). ability and validity and enable comparison across studies.
Remington (2002) conducted a randomized controlled
trial to compare the effects of hand massage (HM) and
CM on the frequency of agitation in PWD. Agitated nurs-
ing homes residents were randomly assigned to one of four
22.3 INDIVIDUALIZED MUSIC
groups (n = 17 each): (1) HM, (2) CM, (3) simultaneous
implementation of CM and HM or (4) control. A new age Individualized music is defined as music that has been
arrangement of Pachelbel’s Canon in D was selected for integrated into the person’s life and is based on personal
its calming qualities, such as a slow tempo of 52 beats per preference (Gerdner, 1992). Extensive clinical experience
minute (instead of 88–108 beats per minute in the original in combination with research findings led to the develop-
orchestral arrangement), soft dynamic levels and repetitive ment of a mid-range theory of individualized music inter-
themes. The music did not contain recognizable lyrics or vention for agitation (IMIA) (Gerdner, 1997). Elements
melodies that might evoke emotional responses. Each group of the mid-range theory include cognitive impairment,
received 10 minutes of the identified intervention with the progressively lowered stress threshold, agitation and
control receiving no intervention. The number of occur- individualized music. Cognitive impairment results in a
rences (frequency) and types of agitated behaviours were decreased ability to receive and process stimuli, resulting
recorded for 10 minutes on four occasions using a modi- in a progressive decline in the person’s stress threshold
fied version of the CMAI immediately before the interven- (Hall and Buckwalter, 1987). Dysfunctional behaviours
tion, during the intervention, immediately following the such as agitation occur when the stress threshold is
Therapeutic effects of music in persons with dementia 231

exceeded (Hall and Buckwalter, 1987). Music may be used was a significant reduction in stress index as measured
as a means of communicating with this population even by a salivary chromogranin A (CgA) following session
in the advanced stages of dementia when the person 16. The authors concluded, ‘the changes in CgA levels
has difficulty or an inability to understand verbal lan- supported Gerdner’s mid-range theory’. No significant
guage and has a decreased ability to interpret environ- findings occurred in the control group across outcome
mental ­stimuli. It is theorized that the presentation of measures.
­individualized music (carefully selected music, based on Given its efficacy when implemented by research staff,
personal preference) will provide an opportunity to stim- it is important to evaluate the effectiveness of individual-
ulate remote memory. This changes the focus of atten- ized music when implemented by trained staff and family
tion and provides an interpretable stimulus, overriding members. As a beginning, Gerdner (2005) conducted a pilot
stimuli in the environment that is meaningless or con- study by using a mixed methodology that included both
fusing. The ­elicitation of memories associated with posi- quantitative and qualitative measures. After appropriate
tive feelings will have a soothing effect on the person with training, staff and family played individualized music for
dementia, which in turn will prevent or alleviate agitation eight PWD living in an LTCF. The intervention was imple-
(Gerdner, 1997). mented over a 4-week period. Individualized music was
Primary propositions of this theory were tested using played daily for 30 minutes at a prescribed time (prior to the
an experimental repeated measures pre-test–post-test estimated ‘peak level’ of agitation). The mean rate of compli-
crossover design that compared the immediate and resid- ance was 86.3%. In addition, staff administered music on an
ual effects of individualized music to classical ‘relaxation’ as needed basis (when the PWD first began exhibiting signs
music relative to baseline on the frequency of agitated of agitation). Agitation was measured using a modified ver-
behaviours in PWD. Thirty-nine subjects were recruited sion of the CMAI. A statistically significant reduction in
from six long-term care facilities (LTCFs). An interview agitation was found during the immediate presentation of
guide was used to obtain specific information on the sub- music, with an overall reduction in agitation found on day
ject’s musical preference and to identify the importance of shift during weeks 1–8 and on evening shift during weeks
music in the subject’s life prior to the onset of dementia. 5–8. Staff and family interviews provided convergent valid-
If cognitive impairment precluded the subject from com- ity to quantitative findings. In addition, staff and family
pleting the form, a family member provided the informa- reported that individualized music provided a catalyst for
tion. Group A (n = 16) received individualized music for 6 meaningful interaction between the person with dementia
weeks followed by a 2-week ‘washout’ period and 6 weeks and others.
of classical relaxation music, Group B (n = 23) received Researchers in Taiwan evaluated nursing staff (n = 17)
the same protocol but in reverse order. Music interven- knowledge and adherence to the evidence-based guideline
tions were presented for 30 minutes, two times per week. for individualized music. Initial training included an inter-
A modified version of the CMAI was used to measure the active educational programme. Ongoing reminders, a local
dependent variable. An R-ANOVA with Bonferroni post opinion leader and an audit checklist were used to facili-
hoc test showed a significant reduction in agitation dur- tate and monitor continued adherence to the intervention
ing and following the presentation of individualized music protocol. Comparison of pre- and post-test scores found a
compared with classical music (Gerdner, 2000). statistically significant improvement (p < 0.001) in knowl-
This seminal work served as the impetus for an expand- edge of the intervention following the training session with
ing body of research that supports the use of individu- a mean compliance of 72% (Sung et al., 2008). In addition,
alized music for the management of agitation in PWD this study evaluated residents’ responses to individualized
(Ragneskog et al., 2001; Janelli et al., 2002; Suzuki et al., music as measured by the CMAI (Sung et al., 2006). An
2004; Sung et al., 2006, 2008, 2010; Park and Pringle-Specht, experimental group (n = 32) received individualized music
2009; Gallagher, 2011). for 30 minutes, twice per week over 6 weeks, while the con-
Japanese researchers (Suzuki et al., 2004) expanded trol group (n = 25) received usual care with no music. The
these efforts by adding functional and biophysiological experimental group had a statistically significant reduction
measures as well as behavioural outcome measures. The in overall agitation (t = −2.19, p < 0.05) and physically non-
study included 10 subjects with dementia who received aggressive behaviours (t = −3.75, p < 0.0001) compared to
individualized or preferred music twice per week for 8 the control group.
weeks. During the corresponding time period, 13 sub- Park and Pringle-Specht (2009) trained 15 family mem-
jects participated in a comparison intervention (games, bers, caring for an older adult with dementia in the home,
drawing, pasting pictures). Analysis, comparing baseline on the use of the evidence-based guideline for individual-
with 1-week post-intervention scores, found that subjects ized music (Gerdner 2007). Outcome measures included the
in the experimental group had a statistically significant modified CMAI. A quasi-experimental design was used in
improvement in the ‘language’ subscale of the Mini- which individualized music was implemented twice a week
mental State Examination (MMSE) and a statistically for 2 weeks. There was a significant reduction in agitation
significant reduction in ‘irritability’ as measured by the during the intervention period (p < 0.05) compared with
Multidimensional Observational Scale. In addition, there baseline and post-intervention periods.
232 Dementia

increased positive social interaction exhibited by the sub-


22.4 EVIDENCE-BASED GUIDELINE jects supports Kitwood’s (1997) theory of personhood.
A group MT intervention reduced agitation in nurs-
An evidence-based guideline for the implementation of ing home residents with dementia. Twenty subjects were
individualized music was originally developed in 1996, was assigned to receive either 50 minutes of MT three times per
refined over time and is now in the fifth edition (Gerdner, week for 15 weeks, or no experimental intervention. The
2013). An evidence grade schema accompanies this guide- experimental group demonstrated a significant reduction
line. The schema is used to assign a specific grade based on in agitated behaviours (Choi et al., 2009).
the strength and type of evidence for each recommendation In a randomized clinical trial, Guetin et al. (2009) inves-
within the guideline. An abridged version of the protocol tigated the effect of individualized MT sessions on anxiety
was originally developed for consumers in 2001 and has and depression in adults with mild to moderate dementia.
been revised to accompany the most recent version of the Subjects were randomly assigned to the treatment group
evidence-based protocol (Gerdner, 2015a). (n = 15) receiving weekly sessions of individualized MT or
The guideline describes the assessment criteria for use of the control group receiving weekly reading sessions for 16
pre-recorded music for the purpose of alleviating agitation in weeks. The individualized MT sessions used music of a style
PWD. The assessment addresses the importance of ethnicity chosen by each subject. Subjects in the experimental group
in the identification of musical preferences. The importance were exposed to a 20-minute sequence with a progressive
of ethnicity is underscored in a published case example of reduction in rhythm, frequency and volume followed by a
a Mexican-American man who preferred guitar music that ‘re-enlivening’ phase. Subjects were assessed for measures
reflected his ethnic heritage (Gerdner, 2015b). The guideline of anxiety (Hamilton Scale) and depression (Geriatric
also includes a recommendation for assessing the temporal Depression Scale) at weeks 1, 4, 16 and 24. Results indicated
patterning of agitation so that the timing of the intervention that anxiety was significantly improved in the MT group at
can be tailored to maximize the effects and benefits of the week 4 (p = 0.002) and week 16 (p = <0.001). The effect was
intervention. sustained for 8 weeks after withdrawal of the intervention
Importantly, the Oslo Resource Center for Dementia (p = 0.0002). A non-significant difference in depression was
and Psychiatric Care of the Elderly, under the direction reported for weeks 4 and 8, however, a significant reduction
of Dr. Audun Myskja, implemented a previous version of in depression from baseline was observed in the MT group
the evidence-based protocol for individualized music in by week 24 (p = 0.03). Although the sample appeared small,
three nursing homes in Oslo, Sweden, and has incorpo- it was determined by power analysis to be adequate for a
rated the intervention into a complementary therapy mod- level of significance of 0.05 and power of 0.9.
ule designed with academic credits for a master’s degree at Ziv et al. (2007) examined the effect of background
Buskerud University College (GERIA, n.d.). music on both positive and negative behaviours in a sample
of 28 nursing home residents with dementia. Positive, nega-
tive and neutral behaviours were recorded on an investiga-
tor developed scoring sheet for two 17-minute periods, one
22.5 MUSIC THERAPY with music and one without music. The music interven-
tion consisted of popular music from 1964, assumed to be
Although the music interventions described above can be familiar to the subjects. Results indicated that more positive
utilized by professional and lay caregivers, music therapy behaviours and fewer negative behaviours were exhibited
(MT) is a discipline in its own right. A music therapist while music was played (p = 0.001). There was no difference
receives advanced training in music, psychology, physiol- in the frequency of neutral behaviours.
ogy and MT techniques and completes a clinical intern- In a pilot study, Brotons and Marti (2003) examined
ship in a college degree programme (Parker, 2009). Music the effect of MT in 11 patients with probable Alzheimer’s
therapy is presented to individuals or groups. Social inter- disease and their caregiver. In a residential setting, sub-
action becomes an important part of the music session. The jects (patients) received 10 MT sessions over 12 days, four
therapist interacts with group members through singing, alone and seven with their caregiver. Caregivers perceived
eye contact and conversation between songs. Participants improved social behaviours (50%), emotional state (67%)
benefit from the social stimulation from others in the group. and physical/motor activity (33%). Most caregivers (66.7%)
Familiar group singing during the sundowning period, reported feeling more relaxed after MT sessions. Anecdotal
late afternoon and early evening, improved mood and social observations reported include improved expressive lan-
behaviour in four nursing home residents with moderate guage, improved short-term memory and spontaneous
dementia who exhibited sundowning syndrome (Lesta and social interaction following MT.
Pelocz, 2006). Subjects’ mood and social behaviour were Ueda et al. (2013) conducted a systematic review and
assessed before, during and after a 30-minute group singing meta-analysis on the effects of MT on behaviour and psy-
intervention, using a facility constructed behaviour assess- chological symptoms in dementia (BPSD). Twenty studies
ment chart and a mood and social behaviour tool devel- were selected including randomized controlled trials, con-
oped by the investigators. The authors concluded that the trolled clinical trials, cohort studies and controlled trials
Therapeutic effects of music in persons with dementia 233

and conducted a meta-analysis using standardized mean Gerdner, L.A. (1997). An individualized music interven-
differences (SMD). Findings showed that MT had small tion for agitation. Journal of the American Psychiatric
effects on BPSD (SMD, –0.49; 95% confidence interval, Nurses Association, 3: 177–184.
–0.82 to –0.17, p = 0.02). Gerdner, L.A. (2000). Effects of individualized vs. clas-
sical "relaxation" music on the frequency of agita-
tion in elderly persons with Alzheimer’s disease and
related disorders. International Psychogeriatrics,
22.6 CONCLUSIONS 12: 49–65.
Gerdner, L.A. (2005). Use of individualized music by
Most research evaluating the effects of music in PWD has trained staff and family: Translating research into prac-
been conducted in LTCFs. There is an obvious need to tice. Journal of Gerontological Nursing, 31: 22–30.
expand these research efforts to other healthcare environ- Gerdner, L.A. (2007). Evidence–Based Guideline:
ments. Outcome measures have focused on the observa- Individualized Music for Elders with Dementia, Fourth
tion of agitated behaviours. There is also need to expand edition. In M.G. Titler (series ed.) series on Evidence–
these efforts through biophysiological measures. With the Based Practice for Older Adults. Iowa City, IA: The
emphasis on a reduction in negative behaviours there is also University of Iowa College of Nursing Gerontological
need to develop ways to quantify the positive behavioural Nursing Intervention Research Center, Research
response that has been noted anecdotally. Finally, feasibil- Translation and Dissemination Core.
ity studies are required to evaluate the cost effectiveness of Gerdner, L.A. (2013). Evidence-Based Guideline:
implementing music in PWD. Individualized Music for Elders with Dementia, Fifth
Edition. In M.G. Titler (series ed.), series on Evidence–
REFERENCES Based Practice for Older Adults. Ann Arbor, MI:
National Nursing Practice Network, University of
Brodaty, H., Draper, B. and Low, L.-F. (2003). Nursing Michigan, School of Nursing.
home staff attitudes towards residents with dementia: Gerdner, L.A. (2015a). Individualized Music Quick
Strain and satisfaction with work. Journal of Advanced Reference Guide. In M.G. Titler (series ed.), series on
Nursing, 44: 583–590. Evidence–Based Practice for Older Adults. Ann Arbor,
Brotons, M. and Marti, P. (2003). Music therapy with MI: National Nursing Practice Network: University of
Alzheimer’s patients and their family caregivers: Michigan School of Nursing.
A pilot project. Journal of Music Therapy, 40: 138–150. Gerdner, L.A. (2015b). Ethnicity an inherent criteria for
Choi, A.N., Lee, M.S., Cheong, K.J. and Lee, J.S. (2009). assessment of individualized music for persons with
Effects of group music intervention on behavioral and Alzheimer’s disease. Clinical Gerontologist, 38 (2):
psychological symptoms in patients with dementia: 179–186.
A pilot-controlled trial. The International Journal of GERIA – The City of Oslo Resource Center for Dementia
Neuroscience, 119: 471–481. and Psychiatric Care (n.d.). Individualized Music
Corrigan, J.D. (1989). Development of a scale for assess- Training Program course description. Available at
ment of agitation following traumatic brain injury. http://www.helse-og-velferdsetaten.oslo.kommune.
Journal of Clinical and Experimental Neuropsychology, no/getfile.php/helse-ogvelferdsetaten (HEV)/Internett
1: 261–277. (HEV)/GERIA/GERIaintro eng.pdf. Accessed 15 July
Denny, A. (1997). Quiet music an intervention for meal- 2011.
time agitation. Journal of Gerontological Nursing, 23: Goddaer, J. and Abraham, I.L. (1994). Effects of relaxing
16–23. music on agitation during meals among nursing home
Elliot, D., Polman, R. and McGregor, R. (2011). Relaxing residents with severe cognitive impairment. Archives
music for anxiety control. Journal of Music Therapy, of Psychiatric Nursing, 8: 150–158.
48: 264–288. Guetin, S., Portet, F., Picot, M.C. et al. (2009). Effect of
Fernández, M., Gobartt, A.L., Balañá, M. and the music therapy on anxiety and depression in patients
COOPERA Study Group (2010). Behavioural symptoms with Alzheimer’s type dementia: Randomised, con-
in patients with Alzheimer’s disease and their associa- trolled study. Dementia and Geriatric Cognitive
tion with cognitive impairment. BMC Neurology, 10: Disorders, 28: 36–46.
1–9. doi:10.1186/1471-2377-10-87 Hall, G.R. and Buckwalter, K.C. (1987). Progressively low-
Gallagher, M. (2011). Evaluating a prtotocol to train ered stress threshold: A conceptual model for care of
hospice staff in administering individualized adults with Alzheimer’s disease. Archives of Psychiatric
music. International Journal of Palliative Nursing, Nursing, 1: 399–406.
17: 196–201. Hicks-Moore, S.L. (2005). Relaxing music at mealtime
Gerdner, L.A. (1992). The effects of individualized music in nursing homes: Effects on agitated patients with
on elderly clients who are confused and agitated. dementia. Journal of Gerontological Nursing, 31:
Masters Thesis, The University of Iowa, Canada. 26–32.
234 Dementia

Janelli, L., Kanski, G. and Wu, Y. (2002). Individualized dementia: A randomized, controlled trial. Journal
music – A different approach to the restraint use. of the American Geriatrics Society, 52: 1795–1804.
Rehabilitation Nursing, 27: 221–226. doi:10.1111/j.1532-5415.2004.52501.x
Kitwood, T. (1997). Dementia Reconsidered: The Sung, H., Chang, A.M. and Abbey, J. (2006). The effects
Person Comes First. New York, NY: Open of preferred music on agitation of older people with
University Press. dementia in Taiwan. International Journal of Geriatric
Léger, J., Moulias, R., Robert, P. et al. (2002). Agitation Psychiatry, 21: 999–1000.
and aggressiveness among the elderly population Sung, H.-C., Chang, A.M. and Abbey, J. (2008). An
living in nursing or retirement homes in France. implementation programme to improve nursing home
International Psychogeriatrics, 14: 405–416. staff’s knowledge of and adherence to an individu-
Lesta, B. and Petocz, P. (2006). Familiar group singing: alized music protocol. Journal of Clinical Nursing,
Addressing mood and social behavior of residents with 17: 2573–2579.
dementia displaying sundowning. Australian Journal of Sung, H.-C., Chang, A.M. and Lee, W.L. (2010). A
Music Therapy, 17: 2–17. preferred music listening intervention to reduce
Lin, Y., Chu, H., Yang, C. et al. (2011). Effectiveness of anxiety in older adults with dementia in nursing
group music intervention against agitated behavior in homes. Journal of Clinical Nursing 19: 1056–1064.
elderly persons with dementia. International Journal of doi:10.111/j.1365-2702.2009.0316.x
Geriatric Psychiatry, 26: 670–678. Suzuki, M., Kanamori, M., Watanabe, M. et al. (2004).
Park, H. and Pringle-Specht, J.K. (2009). Effect of individu- Behavioral and endocrinological evaluation of music
alized music on agitation in individuals with dementia therapy for elderly patients with dementia. Nursing
who live at home. Journal of Gerontological Nursing, and Health Sciences, 6: 11–18.
35: 47–55. Tabloski, P.A., McKinnon-Howe, L. and Remington,
Parker, A.B. (2009). Music therapy clarifications. Journal R. (1995). Effects of calming music on the level of
of Psychosocial Nursing and Mental Health Services, ­agitation in cognitively impaired nursing home resi-
47: 15. dents. The American Journal of Alzheimer’s Care and
Ragneskog, H., Asplund, K., Kihlgren, M. and Norberg, Related Disorders and Research, 10 (1): 10–15.
A. (2001). Individualized music played for agitated Thompson, R.G., Moulin, C.J.A., Hayre, S. and Jones,
patients with dementia: Analysis of video-recorded R.W. (2005). Music enhances category fluency in
sessions. International Journal of Nursing Practice, healthy older adults and Alzheimer’s disease patients.
7: 146–155. Experimental Aging Research, 31: 91–99.
Remington, R. (2002). Calming music and hand mas- Ueda, T., Suzukamo, Y., Sato, M. and Isumi, S.-I. (2013).
sage with agitated elderly. Nursing Research, Effects of music therapy on behavioral and psycho-
51: 317–323. logical symptoms of dementia: A systematic review of
Samus, Q.M., Robsenblatt, A., Steele, C. et al. (2005). meta-analysis. Ageing Research Reviews, 12: 628–641.
The association of neuropsychiatric symptoms and Watkins, G.R. (1997). Music therapy: Proposed physiologi-
environment with quality of life in assisted living cal mechanisms and clinical implications. Clinical Nurse
­residents with dementia. Gerontologist (Special Specialist, 11: 43–50.
Issue), 45 (Suppl. 1), 19–26. doi:10.1093/geront/45. Ziv, N. and Dolev, E. (2013). The effect of background
suppl_1.19 music on bullying: A pilot study. Children and Schools,
Sloane, P.D., Hoeffer, B., Mitchell, C.M. et al. (2004). 35: 83–90.
Effect of person-centered showering and the towel Ziv, N., Granot, A., Hai, S. et al. (2007). The effect of back-
bath on bathing-associated aggression, agita- ground stimulative music on behavior in Alzheimer’s
tion and discomfort in nursing home residents with patients. Journal of Music Therapy, 44: 329–343.
23
Psychological, behavioural and psychosocial
interventions for neuropsychiatric symptoms
in dementia: What works, what does not and
what needs more evidence?

GILL LIVINGSTON AND CLAUDIA COOPER

different aetiologies (Bruen et al., 2008) and require dif-


23.1 INTRODUCTION ferent management. Neuropsychiatric symptoms are often
treated with psychotropic medications (see Chapter 25).
The neuropsychiatric symptoms of dementia (see Chapters There are major concerns about their safety, particularly
7 and 8) include disturbed perception, thought, mood or antipsychotics (Schneider et al., 2005) and also citalopram
behaviour (Finkel et al., 1996). Effective treatment is a pri- (Porsteinsson et al., 2014), and efficacy (antipsychotics,
ority as clinically significant neuropsychiatric symptoms memantine, carbamazepine and cholinesterase inhibitors)
are distressing to the person living with dementia and con- (Trinh et al., 2003; Schneider et al., 2006; Howard et al.,
tribute significantly to caregiver burden (Coen et al., 2002), 2007; Fox et al., 2012). Guidelines, therefore, recommend
institutionalisation (O’Donnell et al., 2004) and care costs non-pharmacologic strategies as the preferred first line
(Ryu et al., 2005), occur frequently and are ­persistent. The treatment approach (except where there is imminent dan-
prevalence of clinically significant symptoms is around a ger or safety concerns) (National Institute for Clinical
quarter of those with mild cognitive impairment (MCI) Excellence [NICE], 2006; Kales, 2015).
(Lyketsos et al., 2002; Ryu et al., 2011) one-third of people The acronym DICE (Describe, Investigate, Create,
with mild dementia rising to two-thirds with more severe Evaluate) outlines an approach to non-pharmacological
dementia in the community (Lyketsos et al., 2002; Ryu et management of behavioural problems in dementia. This
al., 2005) and around 80% of people with dementia in care is a systematic way of describing the problems, investigat-
homes (Selbaek et al., 2008; Bergh et al., 2011). The symp- ing possible causes, creating a plan and evaluating how well
toms are persistent in each setting: around 75% of people treatments are working and whether there is a need to mod-
with MCI have persistent symptoms (Ryu et al., 2011) and ify the approach (Kales, 2015).
80% of those in the community have symptoms for at least Several previous reviews, including our own (Livingston
6 months (Ryu et al., 2005). In care homes, symptoms such et al., 2005), considered all neuropsychiatric symptoms’
as delusions, agitation, irritability and apathy (Lawlor, management together. A strategy that is successful for
2000; Margallo-Lana et al., 2001) are particularly likely to one symptom may, however, not be right for another. Two
remain a problem with about 75% persistence for at least systematic reviews, including our own, have specifically
a year (Selbaek et al., 2008), making the need for effective considered non-pharmacological management of agita-
treatment a high priority. tion in dementia (Kong et al., 2009; Livingston et al., 2014)
Neuropsychiatric symptoms occur in clusters and the and another for depression and anxiety (Orgeta et al.,
most commonly defined clusters are agitation, psychosis 2014). This chapter examines the evidence for each therapy
and mood disorder (Ballard et al., 2008). They may have and discusses what works, what does not and what needs

235
236 Dementia

more evidence. We draw on evidence from our system- et al., 2001; Huang et al., 2003). Training caregivers in BMT
atic reviews that give more details of the papers reviewed did not help to reduce psychotropic drug use or symptom
(Livingston et al., 2005, 2014). frequency at 1-year ­follow-up (Weiner et al., 2002).
Combined exercise and BMT led to significant improve-
ments in depression at 3 months but not at 2 years (Teri
et al., 2003). In a pilot RCT, caregivers were taught BMT
23.2 BEHAVIOURAL MANAGEMENT
either through written materials or in a home training pro-
AND COGNITIVE THERAPIES
gramme with the objective to reduce response to identified
stressors (Huang et al., 2003). Care recipients in the second
23.2.1 COGNITIVE OR BEHAVIOURAL group were less aggressive, but this finding should be inter-
MANAGEMENT TECHNIQUES preted cautiously, as the study was small and raters were not
(BMTs) WITH PATIENTS blind to the intervention status.
An excellent, adequately powered RCT trained ‘com-
Two randomized controlled trials (RCTs) employed behav- munity consultants’ (primary care health workers) to use
ioural management techniques (BMTs). One used a complex a structured manual-based intervention (the STAR or Staff
intervention (life review, sensory stimulation, single-word Training in Assisted Living Residences-C treatment) (Teri
commands and problem-oriented strategies) and was inef- et al., 2005). They had eight sessions with 95 individual
fective (Beck et al., 2002). The second used a token economy, patient/caregiver dyads at home, teaching them to monitor
which was less effective than a milieu treatment (Mishara, behavioural problems, understand behaviour management
1978). and modify antecedents or consequences. They also con-
In one RCT, 44 participants with dementia received centrated on improving caregiver communication, increas-
manual guided treatment for the patient and caregiver and ing pleasant events and enhancing caregiver support. There
43 received a problem-solving treatment only for the care- were significant improvements, maintained at 6 months, in
giver (Teri et al., 1997). Both interventions were equally suc- caregiver mood and care recipient neuropsychiatric symp-
cessful in improving depressive symptoms immediately and toms. In a small study to evaluate the STAR programme
at 6-month follow-up (Teri et al., 1997). Three smaller RCTs in UK care homes, residents demonstrated significantly
also had positive results (Suhr et al., 1999; Benedict et al., reduced symptoms of depression and behavioural problems
2000; Spector et al., 2015). In one intervention, participants (Goyder et al., 2012).
were taught progressive muscle relaxation and 2 months
later they had significantly fewer neuropsychiatric symp- ●● The evidence that the STAR-C treatment which trains
toms. In the second intervention, the behaviour of patients staff helps neuropsychiatric symptoms is strong.
with multiple sclerosis associated dementia improved with a ●● Teaching BMT’s principles to family caregivers seems
cognitive behaviour intervention. Finally, a pilot RCT, found inefficacious in managing neuropsychiatric symptoms.
that up to 10 Cognitive Behaviour Therapy (CBT) sessions
for people with dementia, with family caregivers supporting
them, may reduce patient depression (Spector et al., 2015). A 23.2.3 TEACHING FAMILY CAREGIVERS
systematic review of psychological therapies for depression TO CHANGE THEIR BEHAVIOUR
in dementia, including two CBT interventions, concluded
that they may help depression but not anxiety (Orgeta et al., Ten studies (including nine RCTs) taught family caregivers
2014). But the studies, while hopeful, were not high quality to change their interactions with relatives with dementia. In
enough to be definitive (Orgeta et al., 2015). an RCT of an educational programme for family caregivers,
comprising supportive counselling, psychoeducation and
●● There is consistent evidence from the RCTs that psycho- training in management strategies, patients’ institutional-
logical therapies are a promising intervention that may ization rates decreased (Eloniemi-Sulkava et al., 2001) for
reduce depressive symptoms and the effects lasted for 3 months but not in 2 years. A smaller study of the same
months but no definitive evidence. intervention with individual families found significant
improvements at 6 months in mood and ideational distur-
23.2.2 TEACHING CAREGIVERS BMT bance (McCallion et al., 1999a). One RCT, with 144 people,
compared a group intervention to treatment as usual and
Several disparate studies trained family caregivers in BMT. the difference in neuropsychiatric symptoms at 16 weeks
Two high-quality studies found (at 3 months, 6 months or approached significance (Herbert et al., 2003). In a fur-
12 months) that training family caregivers in BMT was inef- ther trial, primarily powered for caregiver’s mental health,
fective over 4 or 11 sessions for severe or symptomatic agita- neuropsychiatric symptoms improved immediately after
tion in people with dementia living at home (Teri et al., 2000; 12 weeks training in stress management, dementia educa-
Gormley et al., 2001). Similarly, two studies that trained staff tion and coping skills but not 3 months later (Marriott et al.,
and family caregivers in CBT for people with severe agita- 2000). A pilot RCT, with limited power, involved psycho-
tion found no improvement compared with controls (Wright education, instruction to caregivers on how to change their
Psychological, behavioural and psychosocial interventions for neuropsychiatric symptoms in dementia 237

interactions with the patient or both (Burgener et al., 1998). et al., 2001) showed some positive results on non-cognitive
The difference in patients’ behaviour at 6 months again symptoms, although the studies used different follow-up
approached significance. end points (immediately after to 9 months after therapy).
There were early behaviour improvements, but by 9 months
●● BMT providing psychoeducation to caregivers about no significant difference was found. One study showed
changing their own interactions with patients may help reduced depression (Spector et al., 2001). The final study did
reduce neuropsychiatric symptoms possibly limited not report whether behavioural differences were significant
to ­affective symptoms (National Institute for Clinical (Quayhagen et al., 2000).
Excellence and Social Care Institute for Excellence
[NICE/SCIE], 2006 3/id). ●● There is inconsistent evidence that CST improves
some neuropsychiatric symptoms immediately and for
months afterwards.
23.3 DEMENTIA-SPECIFIC THERAPIES
23.4 EDUCATING STAFF TO CHANGE
23.3.1 REMINISCENCE THERAPY THEIR PERSPECTIVE AND
Reminiscence therapy is a group intervention using m ­ aterial COMMUNICATION
from the past, such as photos or music, to s­ timulate memory
and help create identity and intimacy. There is no evidence Person-centred care, communication skills training and
it benefits neuropsychiatric symptoms (Baines et al., 1987; dementia care all seek to change staffs’ communication
Baillon et al., 2004; Livingston et al., 2005). with and thoughts about people with dementia, encour-
aging them to see and treat people as individuals rather
23.3.2 VALIDATIOIN THERAPY than being task focused. Five RCTs have evaluated whether
these strategies reduce agitation (McCallion et al., 1999a,
Validation therapy is about individual uniqueness in order 1999b; Sloane et al., 2004; Chenoweth et al., 2009; Deudon
to help people with dementia resolve conflicts by validating et al., 2009). All included supervision during training and
their feelings rather than concentrating on facts. There is implementation.
one reasonably sized RCT (n = 88) that compares validation One high-quality study of person-centred care training
therapy to usual care or a social contact group (Toseland, found that severe agitation significantly improved dur-
1997) for neuropsychiatric symptoms. There were no differ- ing the intervention and 8 weeks later (Chenoweth et al.,
ences at 1-year follow-up in nursing time or in psychotropic 2009). Two studies of improving communication skills or
medication and restraint usage. person-centred care, for participants with s­ymptomatic
agitation, found significant improvements compared to
●● There is no good evidence of benefit and there is some of the control group during (McCallion et al., 1999a, 1999b)
lack of efficacy. and up to 6 months after intervention (McCallion et al.,
1999a). A large study including participants without high
23.3.3 REALITY ORIENTATION THERAPY agitation levels found that agitation improved significantly
during 8 weeks person-centred care training and 20 weeks
This is based on the idea that impaired orientation handicaps later (Deudon et al., 2009). One small study, where par-
people with dementia so reminders can improve function- ticipant’s agitation levels were unspecified, showed imme-
ing. There are two RCTs using reality orientation consider- diate improvement in agitation during bathing compared
ing neuropsychiatric symptoms and both do not show any to the control group (Sloane et al., 2004).
significant benefit (Hanley et al., 1981; Baines et al., 1987). One large, high-quality care home study evaluated
dementia care mapping. They observed and assessed each
●● There is no good evidence of benefit and some of lack resident’s behaviour, factors improving well-being and
of efficacy. potential triggers; they explained the results to caregivers
and supported proposed change implementation. Severe
23.3.4 COGNITIVE STIMULATION agitation decreased during the intervention and 4 months
THERAPY afterwards (Chenoweth et al., 2009).
Teaching communication skills also reduced patients’
Cognitive stimulation therapy (CST), derived from reality depression at 6 months (McCallion et al., 1999b). A pilot
orientation, uses information processing though themed study of a nursing assistant implementing behavioural
activities to engage people with dementia while providing methods found improvements in non-affective behav-
a group’s social benefits. Its main purpose is cognitive but it ioural symptoms (Lichtenberg et al., 2005). Staff education
might benefit neuropsychiatric symptoms. Three CST RCTs to implement dementia specific therapies was ineffective
(Quayhagen et al., 1995; Romero and Wenz, 2001; Spector (Schrijnemaekers et al., 2002).
238 Dementia

●● Training paid caregivers in communication skills or Weert et al. (2005) subsequently reported that Snoezelen
person-centred care or dementia care mapping, with over 18 months reduced aggression immediately after
supervision during implementation is significantly treatment. An RCT (n = 120) comparing relaxing mas-
effective in care homes for symptomatic and severe sage therapy and ear acupressure interventions to a no
agitation, immediately and up to 6 months. treatment control found that both active treatments led to
●● These strategies may be effective in alleviating patient improvements in depression and anxiety symptoms dur-
mood disturbance, immediately and over months. ing treatment and for up to 1 month afterwards among
people with dementia living in care homes in Spain
(Rodriguez-Mansilla et al., 2015).
Studies evaluating touch as an intervention have
23.5 PSYCHOSOCIAL INTERVENTIONS found a significant improvement in symptomatic and
clinically significant agitation compared to usual care
23.5.1 SENSORY ENHANCEMENT (Remington, 2002; van Weert et al., 2005; Lin et al., 2009).
Three studies investigated ‘therapeutic touch’, defined
as a healing-based touch intervention focusing on the
23.5.1.1 Music/music therapy whole person (Woods et al., 2005; Hawranik et al., 2008;
Woods et al., 2009). Despite therapeutic touch being
Music can be an activity or part of other activities, such efficacious before and after analyses, therapeutic touch
as mealtimes or bath times. (see also Chapter 22) There tended towards being less efficacious than ordinary mas-
were seven small RCTs of music/music therapy inter- sage or usual treatment between group analyses. An RCT
ventions’ effect on neuropsychiatric symptoms in our meta-analysis found that sensory interventions, defined
systematic review (Livingston et al., 2005). All found as aromatherapy, hand massage and a thermal bath, were
improvements in apathy, agitation or disruptive behav- an effective intervention immediately for agitation (Kong
iour during or immediately after the session with no et al., 2009).
longer-term effects.
Three more recent RCTs, all in care homes, evaluated ●● There is evidence for short-term benefits, but no sus-
music therapy by protocol’s effect on agitation, typically tained effects, of sensory stimulation in agitation.
involving warming up with a well-known song, listen- ●● There are contradictory results about sensory stimula-
ing to and then joining in with music (Cooke et al., 2010; tion effect on depressive and anxiety symptoms.
Lin et al., 2011; Sung et al., 2012). The largest study that
included participants of all agitation levels found that
music therapy, twice a week for 6 weeks, was effective com- 23.5.1.3 Aromatherapy
pared to usual care (Lin et al., 2011). A second found a sig-
nificant effect compared to a reading group (Cooke et al., Two RCTs of aromatherapy in care homes explored effec-
2010) and the third a borderline significant effect (Sung tiveness at reducing agitation (Ballard et al., 2002; Burns
et al., 2012). et al., 2011). One large high-quality blinded study found
no immediate or long-term improvement compared to the
●● In care homes, music therapy by protocol significantly control group for participants with severe agitation (Burns
reduces agitation including in those with symptomatic et al., 2011). The other, a non-blinded study, found signifi-
agitation immediately and not longer term. cant improvements compared to the control group (Ballard
et al., 2002). A Cochrane review of aromatherapy for all
neuropsychiatric symptoms concluded that more research
23.5.1.2 Snoezelen therapy and other is needed (Forrester et al., 2014).
sensory stimulation
●● When assessors are blinded to the intervention, aroma-
Snoezelen therapy/multisensory stimulation (MSS) com-
therapy has not been shown to reduce agitation in care
bines relaxation, lights and sounds in specially designed
homes.
rooms for 30–60 minutes. We found seven RCTs in our
earlier systematic review. The largest RCT recruited 136
patients with moderate to severe dementia randomized to 23.5.1.4 Visually complex environments
MSS or a control activity (Baker et al., 2003). There was
no group difference in behaviour or mood and commu- Nine studies investigated complex or distracting visual pat-
nication/interaction either in the short-term or 1-month terns positioned near or on the exit (Livingston et al., 2005)
post-intervention. This large trial followed two promis- were extremely small without a control group. There are no
ing small trials, which found (as did other studies) that RCTs that attempt to reduce wandering in either domes-
disruptive or agitated behaviour briefly improved in or tic or ward environments (Hermans et al., 2007). There is
outside the treatment setting but there was no effect after a consistent effect that a complex environment that makes
the treatment had stopped (Livingston et al., 2005). Van the exit less clear decreases attempts to leave.
Psychological, behavioural and psychosocial interventions for neuropsychiatric symptoms in dementia 239

23.5.1.5 Light therapy differences between groups in neuropsychiatric symptoms


(Lowery et al., 2014), possibly because of low adherence to
Light therapy is hypothesized to reduce agitation through the intervention. A second RCT found no effects on behav-
manipulating circadian rhythms, typically by 30–60 min- iour when one caregiver walked or walked and talked with
utes daily bright light exposure. Three RCTs, all in care two residents (Cott et al., 2002). A Psychomotor Activation
homes (Ancoli-Israel et al., 2003; Dowling et al., 2007; Program RCT involved 134 residents in 11 ‘group care’
Burns et al., 2009), have investigated its effectiveness in homes (Hopman-Rock et al., 1999) in sporting activities
doing so. Light therapy either increased agitation or did such as ball playing and hockey sitting down. There was a
not improve it. trend for the behaviour to improve, but the high dropout
due to illness considerably reduced power.
●● There is no evidence that light therapy reduces symp-
tomatic or severe agitation in care homes, and it may ●● Exercise therapy does not appear to be effective at
worsen it. improving neuropsychiatric symptoms of dementia,
although it requires to be designed and more evidence
23.5.2 STRUCTURED ACTIVITY as to how to increase adherence to a programme.

23.5.2.1 Therapeutic activity programs 23.6 WHAT WORKS, WHAT DOES


NOT AND WHAT NEEDS MORE
Five reasonably sized RCTs have evaluated group activities
for agitation in care settings; RCTs with standard activi-
EVIDENCE?
ties reduced mean agitation levels and decreased symp-
toms in care homes while they were in place (Buettner Summarizing the evidence, there are therapies for which
et al., 1997; Lin et al., 2009). One very high-quality RCT we have evidence of efficacy in alleviating neuropsychiatric
found no additional effect on agitation of individual- symptoms immediately and over the longer term.
izing activities according to functional level and inter-
est (Kolanowski et al., 2011), although two lower-quality 23.6.1 IMMEDIATE AND LONGER-TERM
RCTs did (Kovach et al., 2003; Cohen-Mansfield and TREATMENT
Jensen, 2006). All s­ tudies were in care homes, except one,
where some participants attended a day centre and others Overall, there is excellent evidence that training staff in care
lived in a care home (Cohen-Mansfield et al., 2006). None homes about dementia, respecting the person as an individ-
specified a significant degree of agitation for inclusion. ual, communication and sometimes behavioural skills alle-
Only one study measured agitation after the intervention viate mood disturbance, agitation and aggression in people
was stopped and it did not show effects at 1- and 4-week living with dementia, immediately and over months. There
follow-up (Kolanowski et al., 2011). is also evidence that for people with dementia living in care
In a recent cluster-randomized trial in 18 nursing homes homes activities and music therapy by protocol decrease
in Berlin, which included 10 months of brief activities, pro- overall agitation, and sensory intervention decreases clini-
vided once a week, there were significant group differences cally significant agitation immediately but not over the lon-
in apathy during the 10-month intervention period, which ger term.
reflected an increase in apathy in the control but not in the Teaching family caregivers how to react to neuropsy-
intervention group. These differences did not persist after chiatric symptoms, coping strategies that ensure that they
the intervention stopped (Treusch et al., 2015). have emotional support and the principles of BMT are
effective in managing some neuropsychiatric symptoms.
●● Activities in care homes reduce levels of agitation sig- It is, however, unclear whether the whole package is essen-
nificantly while in place but there is no evidence regard- tial. In contrast, teaching the principles of BMT to caregiv-
ing longer-term effect. There is no evidence for activities ers as a sole intervention is of doubtful efficacy. However,
in severe agitation, or outside care homes. professionals (as opposed to families) using principles of
●● Evidence for other neuropsychiatric symptoms is incon- BMT with people with dementia may decrease depressive
sistent and inconclusive. symptoms.

23.5.2.2 Exercise 23.6.2 WHAT DOES NOT WORK


One large RCT evaluated an exercise regimen for people Aromatherapy, exercise and light therapy have not demon-
with dementia and their family caregivers (individually strated efficacy in adequately powered studies. There is no
tailored walking regimen designed to become progres- evidence that reminiscence therapy, reality orientation and
sively intensive and last between 20 and 30 minutes, validation therapy help neuropsychiatric symptoms and
at least five times per week). There were no significant some evidence that they do not.
240 Dementia

23.6.3 WHAT NEEDS MORE EVIDENCE dementia in Norwegian nursing homes. International


Psychogeriatrics, 23: 1231–1239.
The most promising strategies need more evidence and are Bruen, P.D., McGeown, W.J., Shanks, M.F. and Venneri, A.
about the effect of coping strategies, behaviour manage- (2008). Neuroanatomical correlates of neuropsychi-
ment, including increasing pleasant events and cognitive atric symptoms in Alzheimer’s disease. Brain, 131:
behaviour strategies for people with dementia, depressive 2455–2463.
symptoms and depression. Buettner, L.L. and Ferrario, J. (1997). Therapeutic
­recreation-nursing team: A therapeutic intervention
for nursing home residents with dementia. Annual in
Therapeutic Recreation, 7: 21.
23.7 CONCLUSIONS Burgener, S.C., Bakas, T., Murray, C. et al. (1998). Effective
caregiving approaches for patients with Alzheimer’s
Overall, we judge that effective tools are available to treat disease. Geriatric Nursing, 19: 121–126.
neuropsychiatric symptoms in dementia but the systems are Burns, A., Allen, H., Tomenson, B. et al. (2009). Bright
not yet in place where they can be routinely available. Finding light therapy for agitation in dementia: A randomized
ways in which management strategies can be effectively, effi- controlled trial. International Psychogeriatrics,
ciently and consistently delivered is now a high priority. 21: 711–721.
Burns, A., Perry, E., Holmes, C. et al. (2011). A double-
REFERENCES blind placebo-controlled randomized trial of Melissa
officinalis oil and donepezil for the treatment of agita-
Ancoli-Israel, S., Martin, J.L., Gehrman, P. et al. (2003). tion in Alzheimer’s disease. Dementia and Geriatric
Effect of light on agitation in institutionalized patients Cognitive Disorders, 31: 158–164.
with severe Alzheimer disease. American Journal of Chenoweth, L., King, M.T., Jeon, Y.H. et al. (2009). Caring
Geriatric Psychiatry, 11: 194–203. for Aged Dementia Care Resident Study (CADRES)
Baillon, S., Van Diepen, E., Prettyman, R. et al. (2004). of person-centred care, dementia-care mapping, and
A comparison of the effects of Snoezelen and reminis- usual care in dementia: A cluster-randomised trial.
cence therapy on the agitated behaviour of patients Lancet Neurology, 8: 317–325.
with dementia. International Journal of Geriatric Coen, R.F., O’Boyle, C.A., Coakley, D. and Lawlor, B.A.
Psychiatry, 19: 1047–1052. (2002). Individual quality of life factors distinguishing
Baines, S., Saxby, P. and Ehlert K. (1987). Reality orienta- low-burden and high-burden caregivers of dementia
tion and reminescence therapy. A controlled cross- patients. Dementia and Geriatric Cognitive Disorders,
over study of elderly confused people. British Journal 13: 164–170.
of Psychiatry, 151: 222–231. Cohen-Mansfield, J. and Jensen, B. (2006). Do interven-
Baker, R., Holloway, J., Holtkamp, C.C.M. et al. (2003). tions bringing current self-care practices into greater
Effects of multi-sensory stimulation for people correspondence with those performed premorbidly
with dementia. Journal of Advanced Nursing, benefit the person with dementia? A pilot study.
43: 465–477. American Journal of Alzheimer’s Disease and Other
Ballard, C., Day, S., Sharp, S. et al. (2008). Neuropsychiatric Dementias, 21: 312–317.
symptoms in dementia: Importance and treatment Cohen-Mansfield, J., Parpura-Gill, A. and Golander, H.
considerations. International Review of Psychiatry, 20: (2006). Utilization of self-identity roles for ­designing
396–404. interventions for persons with dementia. Journals
Ballard, C.G., O’Brien, J.T., Reichelt, K. and Perry, E.K. of Gerontology Series B: Psychological Sciences and
(2002). Aromatherapy as a safe and effective treatment Social Sciences, 61B: 202–212.
for the management of agitation in severe dementia: Cott, C.A., Dawson, P., Sidani, S. and Wells, D. (2002).
The results of a double-blind, ­placebo-­controlled The effects of a walking/talking program on commu-
trial with Melissa. Journal of Clinical Psychiatry, 63: nication, ambulation, and functional status in resi-
553–558. dents with Alzheimer disease. Alzheimer Disease and
Beck, C.K., Vogelpohl, T.S., Rasin, J.H. et al. (2002). Effects Associated Disorders, 16: 81–87.
of behavioral interventions on disruptive behavior and Cooke, M.L., Moyle, W., Shum, D.H. et al. (2010). A ran-
affect in demented nursing home residents. Nursing domized controlled trial exploring the effect of
Research, 51: 219–228. music on agitated behaviours and anxiety in older
Benedict, R.H.B., Shapiro, A., Priore, R. et al. (2000). Neuro- people with dementia. Aging and Mental Health,
psychological counseling improves social behavior 14: 905–916.
in cognitively-impaired multiple sclerosis patients. Deudon, A., Maubourguet, N., Gervais, X. et al. (2009).
Multiple Sclerosis, 6: 391–396. Non-pharmacological management of behavioural
Bergh, S., Engedal, K., Roen, I. and Selbaek, G. (2011). The symptoms in nursing homes. International Journal of
course of neuropsychiatric symptoms in patients with Geriatric Psychiatry, 24: 1386–1395.
Psychological, behavioural and psychosocial interventions for neuropsychiatric symptoms in dementia 241

Dowling, G.A., Graf, C.L., Hubbard, E.M. and Luxenberg, J.S. for improving caregiver self-efficacy and decreasing
(2007). Light treatment for neuropsychiatric behaviors the behavioral problems of elders with dementia in
in Alzheimer’s disease. Western Journal of Nursing Taiwan. International Journal of Geriatric Psychiatry,
Research, 29: 961–975. 18: 337–345.
Eloniemi-Sulkava, U., Notkola, I.L., Hentinen, M. et al. Kales, H. (2015). Assessment and management of behav-
(2001). Effects of supporting community-living ioral and psychological symptoms of dementia. British
demented patients and their caregivers: A random- Medical Journal, 350: h369.
ized trial. Journal of American Geriatrics Society, Kolanowski, A., Litaker, M., Buettner, L. et al. (2011).
49: 1282–1287. ­A randomized clinical trial of theory-based activities
Finkel, S., Costa e Silva, J., Cohen, G. et al. (1996). for the behavioral symptoms of dementia in nursing
Behavioral and psychological signs and symptoms of home residents. Journal of the American Geriatrics
dementia: A consensus statement on current knowl- Society, 59: 1032–1041.
edge and implications for research and treatment. Kong, E.H., Evans, L.K. and Guevara, J.P. (2009).
International Psychogeriatrics, 8: 497–500. Nonpharmacological intervention for agitation in
Forrester, L.T., Maayan, N., Orrell, M. et al. (2014). dementia: A systematic review and meta-analysis.
Aromatherapy for dementia. The Cochrane Database Aging and Mental Health, 13: 512–520.
of Systematic Reviews (2): CD003150. Kovach, C., Cashin, S., Taneli, Y. et al. (2003).
Fox, C., Crugel, M., Maidment, I. et al. (2012). Efficacy Effects of the BACE intervention on agitation
of memantine for agitation in Alzheimer’s dementia: of demented ­residents in long term care. The
A randomised double-blind placebo controlled trial. Gerontologist, 43: 233.
PLos One, 7: e35185. Lawlor, B. (2000). Managing behavioural and psycho-
Gormley, N., Lyons, D. and Howard, R. (2001). Behav- logical symptoms in dementia. British Journal of
ioural management of aggression in dementia: A ran- Psychiatry, 181: 463–465.
domized controlled trial. Age and Ageing, 30: 141–145. Lichtenberg, P.A., Kemp-Havican, J., MacNeill, S.E. and
Goyder, J., Orrell, M., Wenborn, J. and Spector, A. Johnson, A.S. (2005). Pilot study of behavioral treat-
(2012). Staff training using STAR: A pilot study in ment in dementia care units. Gerontologist, 45:
UK care homes. International Psychogeriatrics, 24: 406–410.
911–920. Livingston, G., Johnston, K., Katona, C. et al. (2005).
Hanley, I.G., McGuire, R.J. and Boyd, W.D. (1981). Reality Systematic review of psychological approaches to
orientation and dementia: A controlled trial of two the management of neuropsychiatric symptoms
approaches. British Journal of Psychiatry, 138: 10–14. of dementia. American Journal of Psychiatry, 162:
Hawranik, P., Johnston, P. and Deatrich, J. (2008). 1996–2021.
Therapeutic touch and agitation in individuals with Livingston, G., Kelly, L., Lewis-Holmes, E. et al. (2014).
Alzheimer’s disease. Western Journal of Nursing A systematic review of the clinical effectiveness and
Research, 30: 417–434. cost-effectiveness of sensory, psychological and
Herbert, R., Levesque, L., Vezina, J. et al. (2003). Efficacy behavioural interventions for managing agitation
of a psychoeducative group program for caregivers in older adults with dementia. Health Technology
of demented persons living at home: A randomized Assessment, 18: 1–226, v–vi.
controlled trial. Journal of Gerontology Series B – Lin, Y., Chu, H., Yang, C.Y. et al. (2011). Effectiveness of
Psychological Sciences and Social Sciences, 58: group music intervention against agitated behavior in
S58–S67. elderly persons with dementia. International Journal of
Hermans, D.G., Htay, U.H. and McShane, R. (2007). Geriatric Psychiatry, 26: 670–678.
Non-pharmacological interventions for wander- Lin, L.C., Yang, M.H., Kao, C.C. et al. (2009). Using acu-
ing of people with dementia in the domestic set- pressure and Montessori-based activities to decrease
ting. Cochrane Database of Systematic Reviews, (1): agitation for residents with dementia: A cross-over
CD005994. trial. Journal of the American Geriatrics Society, 57:
Hopman-Rock, M., Staats, P.G.M., Tak, E.C.P.M. and 1022–1029.
Droes, R.-M. (1999). The effects of a psychomotor Lowery, D., Cerga-Pashoja, A., Iliffe, S. et al. (2014). The
activation programme for use in groups of cognitively effect of exercise on behavioural and psychological
impaired people in homes for the elderly. International symptoms of dementia: The EVIDEM-E randomised
Journal of Geriatric Psychiatry, 14: 633–642. controlled clinical trial. International Journal of
Howard, R.J., Juszczak, E., Ballard, C.G. et al. (2007). Geriatric Psychiatry, 29: 819–827.
Donepezil for the treatment of agitation in Alzheimer’s Lyketsos, C.G., Lopez, O., Jones, B. et al. (2002). Pre-
disease. New England Journal of Medicine, 357: valence of neuropsychiatric symptoms in dementia and
1382–1392. mild cognitive impairment: Results from the cardio-
Huang, H.L., Shyu, Y.I.L., Chen, M.C. et al. 2003. A pilot vascular health study. Journal of the American Medical
study on a home-based caregiver training program Association, 288: 1475–1483.
242 Dementia

Margallo-Lana, M., Reichelt, K., Hayes, P. et al. (2001). Romero, B. and Wenz, M. (2001). Self-maintenance
Longitudinal comparison of depression, coping, and therapy in Alzheimer’s disease. Neuropsychological
turnover among NHS and private sector staff caring Rehabilitation, 11: 333–355.
for people with dementia. British Medical Journal, 322: Ryu, S.H., Ha, J.H., Park, D.H. et al. (2011). Persistence of
769–770. neuropsychiatric symptoms over six months in mild
Marriott, A., Donaldson, C., Tarrier, N. and Burns, ­A . cognitive impairment in community-dwelling Korean
(2000). Effectiveness of cognitive-behavioural family elderly. International Psychogeriatrics, 23: 214–220.
intervention in reducing the burden of care in carers Ryu, S.H., Katona, C., Rive, B. and Livingston, G. (2005).
of patients with Alzheimer’s disease. British Journal of Persistence of and changes in neuropsychiatric
Psychiatry, 176: 557–562. symptoms in Alzheimer disease over 6 months – the
McCallion, P., Toseland, R.W. and Freeman, K. (1999a). An LASER-AD study. American Journal of Geriatric
evaluation of a family visit education program. Journal Psychiatry, 13: 976–983.
of the American Geriatrics Society, 47: 203–214. Schneider, L.S., Dagerman, K.S. and Insel, P. (2005). Risk
McCallion, P., Toseland, R.W., Lacey, D. and Banks, S. of death with atypical antipsychotic drug treatment
(1999b). Educating nursing assistants to communicate for dementia: Meta-analysis of randomized placebo-
more effectively with nursing home residents with controlled trials. Journal of the American Medical
dementia. The Gerontologist, 39: 546–558. Association, 294: 1934–1943.
Mishara, B.L. (1978). Geriatric patients who improve in Schneider, L.S., Dagerman, K. and Insel, P.S. (2006).
token economy and general milieu treatment pro- Efficacy and adverse effects of atypical antipsy-
grams: A multivariate analysis. Journal of Consulting chotics for dementia: Meta-analysis of randomized,
and Clinical Psychology, 46: 1340–1348. placebo-controlled trials. American Journal of Geriatric
National Institute for Clinical Excellence (NICE). (2006). Psychiatry, 14: 191–210.
Dementia CG42. Available at https://www.nice.org.uk/ Schrijnemaekers, V., van Rossum, E., Candel, M. et al.
guidance/CG42. Last updated May 2016. Accessed 6 (2002). Effects of emotion-oriented care on elderly
June 2016. people with cognitive impairment and behavioral
O’Donnell, B., Drachman, D. and Barned, H. (2004). problems. International Journal of Geriatrics Psychiatry,
Incontinence and troublesome behaviour predict 17: 926–937.
institutionalisation in dementia. Journal of Geriatric Selbaek, G., Kirkevold, O. and Engedal, K. (2008). The
Psychiatry and Neurology, 5: 45–52. course of psychiatric and behavioral symptoms and
Orgeta, V., Qazi, A., Spector, A.E. and Orrell, M. (2014). the use of psychotropic medication in patients with
Psychological treatments for depression and anxi- dementia in Norwegian nursing homes – A 12-month
ety in dementia and mild cognitive impairment. follow-up study. The American Journal of Geriatrics
The Cochrane Database of Systematic Reviews, Psychiatry, 16: 528–536.
(1): CD009125. Sloane, P.D., Hoeffer, B., Mitchell, C.M. et al. (2004).
Orgeta, V. Qazi, A., Spector, A. and Orrell, M. (2015). Effect of person-centered showering and the
Psychological treatments for depression and anxiety in towel bath on bathing-associated aggression,
dementia and mild cognitive impairment: Systematic agitation, and discomfort in nursing home resi-
review and meta-analysis. British Journal of Psychiatry, dents with ­dementia: A randomized, controlled
207(4): 293–298. trial. Journal of the American Geriatrics Society,
Porsteinsson, A.P., Drye, L.T., Pollock, B.G. et al. (2014). 52: 1795–1804.
Effect of citalopram on agitation in Alzheimer disease: Spector, A., Charlesworth, G., King, M. et al. (2015).
The CitAD randomized clinical trial. Journal of the Cognitive-behavioural therapy for anxiety in dementia:
American Medical Association, 311: 682–691. Pilot randomised controlled trial. The British Journal of
Quayhagen, M.P., Quayhagen, M., Corbeil, R.R. et al. Psychiatry, 206: 509–516.
(1995). A dyadic remediation program for care recipi- Spector, A., Orrell, M., Davies, S. and Woods, R.T.
ents with dementia. Nursing Research, 44: 153–159. (2001). Can reality orientation be rehabilitated?
Quayhagen, M.P., Quayhagen, M., Corbeil, R.R. et al. Development and piloting of an evidence-based
(2000). Coping with dementia: Evaluation of four programme of cognition-based therapies for people
nonpharmacologic interventions. International with dementia. Neuropsychological Rehabilitation,
Psychogeriatrics, 12: 249–265. 11: 377–379.
Remington, R. (2002). Calming music and hand massage Suhr, J., Anderson, S. and Tranel, D. (1999). Progressive
with agitated elderly. Nursing Research, 51: 317–323. muscle relaxation in the management of behav-
Rodriguez-Mansilla, J., Gonzalez Lopez-Arza, M.V., Varela- ioural disturbance in Alzheimer’s disease.
Donoso, E. et al. (2015). The effects of ear acupres- Neuropsychological Rehabilitation, 9: 31–44.
sure, massage therapy and no therapy on symptoms Sung, H.C., Lee, W.L., Li, T.L. and Watson, R. (2012). A
of dementia: A randomized controlled trial. Clinical group music intervention using percussion instru-
Rehabilitation, 29 (7): 683–693. ments with familiar music to reduce anxiety and
Psychological, behavioural and psychosocial interventions for neuropsychiatric symptoms in dementia 243

agitation of institutionalized older adults with demen- Trinh, N.H., Hoblyn, J., Mohanty, S.U. and Yaffe, K. (2003).
tia. International Journal of Geriatric Psychiatry, 27: Efficacy of cholinesterase inhibitors in the treatment of
621–627. neuropsychiatric symptoms and functional impairment
Teri, L., Gibbons, L., McCurry, S. et al. (2003). Exercise in Alzheimer disease – A meta-analysis. Journal of the
plus behavioral management in patients with American Medical Association, 289: 210–216.
Alzheimer’s disease: A randomised controlled trial. van Weert, J.C.M., van Dulmen, A.M., Spreeuwenberg,
Journal of the American Medical Association, 290: P.M.M. et al. (2005). Behavioral and mood effects of
2015–2022. Snoezelen integrated into 24-hour dementia
Teri, L., Logsdon, R.G., Peskind, E. et al. (2000). Treatment care. Journal of the American Geriatrics Society,
of agitation in AD – A randomized, placebo-controlled 53: 24–33.
clinical trial. Neurology, 55: 1271–1278. Woods, D.L., Beck, C. and Sinha, K. (2009). The effect
Teri, L., Logsdon, R.G., Uomoto, J. and McCurry, S.M. of therapeutic touch on behavioral symptoms and
(1997). Behavioral treatment of depression in demen- cortisol in persons with dementia. Forschende
tia patients: A controlled clinical trial. The Journals Komplementarmedizin, 16: 181–189.
of Gerontology. Series B, Psychological Sciences and Woods, D.L., Craven, R.F. and Whitney, J. (2005). The
Social Sciences, 52: 159–166. effect of therapeutic touch on behavioral symptoms of
Teri, L., McCurry, S.M., Logsdon, R. and Gibbons, L.E. persons with dementia. Alternative Therapies in Health
(2005). Training community consultants to help family and Medicine, 11: 66–74.
members improve dementia care: A ­randomized con- Weiner, M.F., Tractenberg, R.E., Sano, M. et al. (2002). No
trolled trial. The Gerontologist, 45: 802–811. long-term effect of behavioral treatment on psycho-
Toseland, R.W. (1997). The impact of validation group tropic drug use for agitation in Alzheimer’s disease
therapy on nursing home residents with dementia. The patients. Journal of Geriatric Psychiatry and Neurology,
Southern Gerontological Society, 16 (1): 31–50. 15: 95–98.
Treusch, Y., Majic, T., Page, J. et al. (2015). Apathy in Wright, L.K., Litaker, M., Laraia, M.T. and DeAndrade, S.
nursing home residents with dementia: Results from (2001). Continuum of care for Alzheimer’s disease: A
a cluster-randomized controlled trial. European nurse education and counseling program. Issues in
Psychiatry, 30: 251–257. Mental Health Nursing, 22: 231–252.
24
Treating problem behaviours in dementia
by understanding their biological, social and
psychological causes

IAN JAMES AND LOUISA JACKMAN

dementia and the caregivers, often to prevent harm or dis-


24.1 INTRODUCTION tress occurring to someone in the immediate vicinity. The
chapter is in two parts with a theoretical perspective being
Behavioural and psychological symptoms of dementia followed by a more practical section.
(BPSD) is a term used to describe problematic behaviours
displayed by people with progressive cognitive decline.
Until recently, a medicalized perspective was often used for
BPSD and the treatments chiefly involved the use of psycho- 24.2 TREATMENT APPROACHES
tropic medication. Empirical evidence suggests that such
treatments are frequently ineffective and highly problematic 24.2.1 NATURE AND CAUSES OF BC
(Banerjee, 2009; Barnes et al., 2012). This chapter takes a bio-
psychosocial view of BPSD, preferring the term ‘Behaviours From the term BPSD, there was an implication that behav-
that Challenge’ (BC), illustrating that these behaviours are ioural problems were a symptom or defining feature of
products of a number of factors including individualistic, dementia; however, this is not the case. Indeed, the vast
social and organizational features. The emphasis is, there- majority of people with dementia do not regularly engage
fore, on understanding and acting on causal factors rather in problem behaviours even when they are required to
than symptom alleviation. Hence, it is logical that the treat- cope with situations, such as 24-hour care settings, which
ment of BC needs to reflect this aetiology (James, 2011). most of us would find restrictive and challenging (Bird and
A Stepped-Care model of treatment for BC is presented Moniz-Cook, 2008). In order to understand the nature of
(Brechin et al., 2013), which reflects this emphasis and the behaviour, let us first look at the types of behaviours pre-
is intended to encourage the use of evidence-based non- senting a challenge to caregivers and then we shall discuss
pharmacological strategies as alternatives to psychotropics treatment strategies. Table 24.1 is a truncated list of behav-
(Lawrence et al., 2012). Such strategies often emphasize the iours reported as challenging, taken from an audit of the
importance of good communication and interaction skills work of the Newcastle Challenging Behaviour Team, the
from carers, which enable them to improve well-being and full list is provided in James (2011).
reduce the risk of confrontations with people with demen- These behaviours are not unique to dementia as many of
tia. Particular emphasis is placed on caregiver communica- us may have engaged in some of these activities in the past
tion and interaction skills in this chapter because these are under specific settings, conditions and triggers. If we had
the carer activities utilized on a daily basis to cope and deal been observed engaging in such actions and then asked to
with any problem behaviours. Indeed, when a BC occurs justify ‘why?’, we would have probably attempted to explain
medical and psychological treatments may or may not be the extenuating circumstances that had led us to behave in
employed, but in contrast there will always be some form of such a manner (illness, tiredness, alcohol, fear, distraction,
interaction needing to take place between the person with recent argument, etc.). There may even be more than one

244
Treating problem behaviours in dementia by understanding their biological, social and psychological causes 245

Table 24.1 Audit of behaviours that challenge referrals Figure 24.2 illustrates the case example of Mr A, who was
referred to the psychiatric services due to physical aggres-
Aggressive Forms of
sion towards staff on an inpatient ward. On one occasion,
BCs Non-aggressive Forms of BCs
Mr A hid behind a door and attacked a male nurse as he
Hitting Apathy entered the room, using an armlock he had been taught in
Kicking Depression the army. Staff were very frightened of him, attended him
Grabbing Repetitive noise in pairs and rushed any personal care tasks with him in
Pushing Repetitive questions order to complete jobs in the shortest possible time. Mr A
Nipping Making strange noises was extremely sensitive to psychotropic medication owing
Scratching Constant requests for help to his diagnosis of Lewy body dementia (DLB) and thus psy-
Biting Eating/drinking excessively chological approaches were preferred. The psychiatric team
Spitting Overactivity used a conceptual model (i.e. the Newcastle model) similar
Choking Pacing to the Iceberg model to understand the problematic behav-
Hair pulling General agitation iour. When this information was put together with a richer
Tripping someone Following others/trailing description of the BC from information gleaned from the
Throwing objects Inappropriate exposure of parts use of behaviour charts, a clear pattern began to emerge.
Stick prodding of body The attacks seemed to mainly happen in the morning and
Stabbing Masturbating in public areas were chiefly directed at male carers. During the attacks
Swearing Urinating in inappropriate places Mr A would typically say ‘That’s the last time you f*ck with
Screaming Smearing me’; ‘Think you’re big now...eh’.
Shouting Handling things inappropriately The psychiatric team met with staff and collated the
Physical sexual assault Dismantling objects information, leading to a set of hypotheses about the causes
Verbal sexual Hoarding things of his behaviour. Psychologists refer to these tentative
advances Hiding items hypothetical cause–effect relationships as ‘formulations’.
Acts of self-harm Falling intentionally From their discussions the following explanation began to
Eating inappropriate substances emerge: Mr A’s DLB made him prone to falling and poor
Non-compliance mobility. Often when he got up at night to go to the toilet,
Misidentifying he would fall against the wall giving himself bruises. In the
Source: James, I.A., Understanding Behaviour in Dementia that morning, forgetting he had fallen, he would be looking for
Challenges, Jessica Kingsley, London, United Kingdom, 2011. an explanation for his bruises and pain. His assumptions
led him to the belief that he had been assaulted by someone
of these contributing factors influencing our actions. From during the night while he was vulnerable in bed. Further,
a dementia perspective, it, therefore, seems sensible to give as he perceived himself to be a ‘strong’ man, he assumed it
people with dementia the benefit of looking at the circum- must have been done by a male carer because he would have
stances in which their behaviour occurred, before providing been able to fight off any woman.
the blanket explanation that ‘it’s all due to their dementia!’ This case, illustrating the Newcastle model, illustrates
Figure 24.1 outlines some of the common causal features a number of important things a number of important
associated with BC. Thus, when someone is engaging in BC things. First, a person’s BC has a number of moderating
it is helpful to collect information regarding these features factors (personality, physical and mental health, percep-
to explain his/her problem actions. tual issues, etc.). It is important to collect this background
Figure 24.1 is referred to as the Iceberg model; the tri- information in order to examine the context of the BC.
angle represents an iceberg and below the surface are all Second, a detailed description of the behaviour is required
the things about the person we cannot see that influence to understand how the moderating factors actually lead
the behaviour (James, 2011; James and Hope, 2013). These to the BC. Such information is often collected via behav-
would be regarded as moderating (background) features ioural charts; this information provides the where, when,
with respect to the BC. Some of these are age-related prob- who features of the behaviour. Third, although there are
lems, including pain, sensory loss, mobility issues; others numerous moderating factors, the key mediating features
are related to personality traits and attitudes. Above the line for the problematic behaviours are the person’s beliefs
(the surface) is what we can observe, which is the behav- and the resultant emotions that these thoughts produce.
iour (i.e. what we see the person do) and emotional states. As mentioned above, these are the key features of the CBT
Humans are extremely good at identifying and differentiat- cycle (Figure 24.3).
ing between the seven key emotional states (anger, sadness, Figure 24.3 outlines two cycles, Mr A’s and a typical
anxiety, happiness, shame, disgust, surprise) (Ekman, 1973; caregiver response. The cycles highlight the dynamics of
James, 2011). The key aspect mediating between the ‘above the interaction. The nature of the interaction has become
and below’ surface features is the person’s cognition (i.e. dysfunctional leading Mr A to become more unsettled
thoughts and beliefs). To this extent, the Iceberg model is a and confused. In order to resolve this problem, the psychi-
cognitive behavioural therapy (CBT) framework. atric team needed to help the staff understand the causes
246 Dementia

The Observables:
Environmental these features can be
Setting: recorded on a
Structure and carer behavioural chart
interactions Behaviour and
Emotions
Surface

Beliefs: e.g. These f**king carers


are beating me up!

Drug-related issues: Drugs interactions,


side effects

Mental health status: Perceptual difficulties:


Anxiety, mood, psychosis Visual, auditory, tactile

Cognitive and neurological:


Nature of dementia, cognitive
functioning Physical difficulties:
Falls, pain
Metabolic changes: Impacting on
appetite, energy, irritability Premorbid personality: A ‘short-fuse’

Figure 24.1 Iceberg model.

Mental health Life story Social environment


Previous history of depression. Paranoid Dad died when he was four. Mum and big sister relied on Moved into care when wife had a stroke, prior to
thoughts about being attacked, but due him. Mum depressed for many years, later she became that lived with wife and son. Now on top floor of
to memory errors rather than a psychosis. agoraphobic. Mr Smith looked after his mum—visiting her Dementia nursing home with 40 residents.
every evening for 10 years. He worked hard throughout Doors unmarked—difficult to find way around.
life. Passed exams, but responsibilities prevented him No access to garden outside.
Personality
going to college. Married May in his 30s, worked on the
Very independent and single minded.
docks. One son (Mark). Problem in marriage when May
Likes company. Private around self-
admitted to affair, but issues resolved. After this he
care activities—embarrassed easily.
became more independent, more suspicious of others.
Finds it difficult to relax—always Cognitive abilities
keeping busy. Loved learning new DLB—moderately severe. Most confused
things. Proud of the idea he can in mornings, some variability during day.
‘stand-up’ for himself. Prone to falling, particularly in
Loves animals; often watches nature Triggers
evenings/night.
programmes. (Neurospsych assessment could not be
Hobbies—walking dog, clothes Aggression—When getting up in the morning, he
completed). Frontal lobe deficits. Some
shopping. Coped with stress by going shouts and lashes out. Usually triggered by
expressive dysphasia—milder receptive
out for a walk, usually alone with dog. presence of men attending to his needs first thing
dysphasia.
in the morning.

Physical health The aggression less evident later in day. Although Medication
Arthritis; dermatitis. presentation often fluctuates. Mirtazapine; diazepam; paracetamol.
Recent weight gain; diabetes II Psychotropic medication needs to be
high cholesterol. Pain and monitored carefully due to DLB.
discomfort. Prone to falling,
particularly at night.

Situation—Getting out of bed and /or washed in the morning


Emotion: Angry and aggressive.

Behaviour: Lashes out and shouts. Vocal: I will get you ... You f**king
bast*rds

Thought/need: Male carers are coming into my room at night and


beating me up. Need to protect himself and revenge the assault.

Figure 24.2 Newcastle model for Mr A: information from above and below surface factors of iceberg model. (From James,
I.A., Understanding Behaviour in Dementia that Challenges, Jessica Kingsley, London, United Kingdom, 2011.)
Treating problem behaviours in dementia by understanding their biological, social and psychological causes 247

Trigger: Mr Smith experiencing pain first thing in within a treatment ‘tool-box’, particularly in cases of high
the morning and seeing bruises levels of distress and risk (Ballard et al., 2008; Barnes et al.,
PWD 2012), their problematic side effects mean these classes of
Thoughts/understanding of drugs need to be used with great caution (see Chapter 25).
situation: I am being beaten up at In terms of psychosocial approaches, these can be
night by these men who beat me roughly categorized as psychological, environmental and
while I’m in bed
Emotion: carer–client interactions. Psychological treatments (Hulme
Physiological
Anger
arousal et al., 2010; Gitlin et al., 2012; Whitaker et al., 2014) con-
tain a broad church of interventions (cognitive stimulation
therapy, reminiscence, exercise, reality orientation, valida-
Behaviour: Aggression
tion, music, aromatherapy, animal assisted, etc.). With rela-
tively few exceptions their evidence base remains limited,
Interaction and they are often used in preventative capacities rather
than to treat behaviours once they have emerged. In other
Behaviour: Confrontation; words, the use of these approaches makes it less likely that a
rushing interactions; attending in BC will be triggered, rather than treating it once it has been
twos; using male carers triggered.
The second group – environmental strategies – involves
Emotion: Arousal
Apprehension
designing the architecture and locality to reduce levels of
confusion and distress. Simple things such as having access
Thoughts: He’s cunning and
to a garden, using good signage, appropriately designing
evil!; best to get in and out
as quickly as possible; got rooms, equipment and utensils (adaption of shape, colour,
Care giver to use force to protect texture) can reduce levels of agitation markedly (King’s
yourself at times Fund, 2009).
The final set of strategies – optimizing carer approaches
Figure 24.3 Mr A’s cognitive behavioural therapy to the person with dementia – are perhaps the most impor-
formulation. tant in relation to interpersonal relationships between the
person with dementia and caregivers. Further, no mat-
of the aggression, and then assist the nurses to adapt their ter what the cause of the behaviour (biological, social or
approaches to the situation, making their interactions more psychological), appropriate interactions can serve to de-
functional. escalate problematic situations. Indeed, it has long been rec-
Through formulation, key elements to the treatment ognized that highly competent carers are those who are best
were to reduce Mr A’s risk of falling during the night, pro- able interact, connect and engage with people with demen-
vide him with painkillers first thing in the morning, reduce tia (Levy-Storms, 2008). Such caregivers are able to de-
the number of staff attending to his personal care, and in the escalate highly charged and problematic situations, through
morning, ensuring the initial nurses were female that sup- their verbal and non-verbal skills. As such, researchers have
ported his physical and cognitive needs respectively. In the attempted to teach staff, with some success, how to commu-
following section, we will take a look at intervention strate- nicate with people with dementia (Eggenberger et al., 2013).
gies for the treatment of BC, before focusing in more detail Unfortunately to date, while it has been shown that carers
on carer communication. can be taught to communicate better, these improvements
are not reflected in reductions in BC. Owing to the impor-
24.2.2 TREATMENT APPROACHES tance of carer interactions, we will discuss the latter topic
further below in the context of agitation and anger.
There are five main treatment approaches with respect to
BC, two pharmacological and three psychosocial. Some of
these treatments are the foci of chapters within this book 24.2.3 CARER INTERACTIONS AND
therefore are not addressed here in detail. One’s choice of COMMUNICATION WHEN
strategy must be completely aligned with the identified DEALING WITH AGITATION AND
causes of the behaviour. ANGER
The first of the medication regimens involves the iden-
tification and appropriate prescribing of drugs to treat The most frequent BC clinical referrals are related to agita-
common triggers for distress and agitation, such as pain, tion and aggression rather than more passive expressions
constipation, delirium, diabetes, thyroid disorders, anxi- of BC such as refusal to eat, inactivity, and apathy. This is
ety, psychosis, etc. The second pharmacological strategy mainly due to the fact that the latter activities are less dis-
makes use of the sedating, calming and tranquilizing pow- ruptive and therefore less of a challenge to the system in
ers of antipsychotics, benzodiazepines and mood stabilizers which they are performed. The circumstances that pro-
(Ballard and Howard, 2006). While such drugs are crucial duce aggression and agitation in people with dementia are
248 Dementia

similar to those affecting people who do not have dementia. As behaviours can be the consequences of our man-
An example of this was Mr A, whose anger led to an aggres- agement strategies, they can be reduced significantly if
sive response. Beck and colleagues (Beck, 1976; James, 1999; we respond to the person with dementia in a skilful way
James and Hope, 2013) have recognized that anger occurs that takes account of his/her needs, strengths and deficits
when we perceive our rights are being unjustly infringed, (Brechin et al., 2013). The impact that management strate-
such as when we think we are being taken advantage of gies play in the triggering, maintenance and resolution of
(James, 2011). When we have such thoughts, anger will problem behaviours cannot be overstated, and is well repre-
occur in all of us; therefore it is not uniquely a feature of sented in Table 24.2.
dementia. Of course, dementia can reduce people’s ability Figure 24.2 and Table 24.2 demonstrate that problem
to cope with and control anger when executive function- behaviours are not single actions, rather they are dynamic
ing is affected, although another key difference for people interplays. It is important to capture these patterns of action,
with dementia is that in some situations they are more likely in order to understand the causes and develop potential
to perceive their rights are being infringed; the reasons are treatments.
two-fold. We will summarize some of the key issues that we have
First, their view of a situation may sometimes conflict tried to highlight above. First, BCs are not solely features
with that of the caregivers as they are using degraded of cognitive impairment. Indeed, the same fundamen-
information to process their world. For example, owing to tal mechanisms underpinning BC operate for everyone,
memory difficulties, a care home resident may believe she including those without dementia. However, owing to their
needs to go and see her deceased mother immediately. This cognitive difficulties people with dementia can see things
brings her into conflict with her carer who is repeatedly differently, and this can result in disagreements. Further,
attempting to convince her that her mother died 20 years owing to them being vulnerable, risk management strate-
ago. Such a scenario is common and frequently leads to gies that run counter to the person’s ability to exert control
confrontation. Second, it can be a carer’s intended actions over their environment may need to be introduced and this
to keep the person with dementia safe or clean that trig- can also result in confrontations.
ger conflict (e.g. not letting him go out at night; prevent- Observational studies (Levy-Storms, 2008; Eggenberger
ing her driving her car; asking him to get out of a soiled et al., 2013) inform us that skilful carers are those people who
bed). Indeed, because the person with dementia does not are able to communicate with someone who is getting upset in
always share the same reality as his/her caregiver, they a manner that reduces the likelihood of conflict. To optimize
can often perceive such management strategies as restric- the possibility that carers will have sensitive and appropriate
tive practices, which evokes anger and aggression. BCs are interactions with people with dementia who are distressed
sometimes conceptualized as the by-products of strategies and potentially challenging, two types of information are
carers’ employ to manage the lives of people who are con- required that enable them to ‘put themselves in the shoes’ of
fused, impulsive and disorientated. the person with dementia: (1) a detailed understanding of the

Table 24.2 T
 he triggering of problem behaviours through poor management strategies (MSs) of people with
dementia (PWD)

3a. MSii in Response to the


PWD’s Behaviour That
2. Behaviour of PWD Reinforces Problem
1. Carer’s MSi Triggered by MSi Behaviour 3b. Resolution
Locking exit door to prevent Kicking exit door to get out Taking the person to Escorting the person with
the person with dementia another room in the care dementia while they go
from wandering the busy home for a walk outside
street
Increasing volume of TV Shouting and yelling, owing Tell the person with Encourage the residents
owing to care home to high volume of the TV dementia not to be so who have hearing
residents’ hearing selfish. Explaining to her problems to sit nearer the
problems that the other residents TV
need the higher volume
to hear the TV
Trying to get someone to Refusing to agree to wash Tricking the person to go Helping maintain her dignity
wash after 5 days of self, or be assisted to be into the bathroom, and by giving her a flannel and
refusing to do so washed not letting her out until assisting her to wash
she has let you wash her herself under cover of her
dressing gown
Treating problem behaviours in dementia by understanding their biological, social and psychological causes 249

person’s behaviour, its dynamics and likely triggers, and (2) 24.3.1.1.3 PHASE 3. FORMULATION MEETING
knowledge about both the person they are caring for (his- (INFORMATION SHARING SESSION)
tory, likes, dislikes, etc.) and the context/environment. From A specialist clinician who has collated the assessment infor-
a clinical perspective, the value of these two types of infor- mation meets with the people involved in the person’s care
mation is recognized (see the case of Mr A), and assessments (including staff, family and other professionals) to develop a
tools have been devised to capture these details. These assess- shared understanding of why the person presents with BCs.
ment tools are discussed in the next section. The session is intended to refocus the group from problem to
solution-focused through increasing empathy and improv-
ing understanding of the person’s BC. The formulation ses-
24.3 METHODS AND TOOLS sion is the mechanism through which the group comes to
refocus their attention on the needs of the resident/patient
rather than on the challenge they experience. The formu-
Over the last 6 years, many countries have published lation session can be facilitated by a specialist challenging
national dementia strategies, routinely endorsing the use of behaviour clinician or experienced ward or home staff.
a more holistic approach to treating BC. While commend-
able, the dementia strategies have been uniformly poor at
being specific about how to implement evidence-based 24.3.1.1.4 PHASE 4. INTERVENTIONS
alternatives to psychotropic medication. Hence, the follow- Interventions are based on the group’s suggestions, then
ing section operationalizes the conceptual ideas expressed developed and refined during the formulation session with
earlier and describes the assessment tools and protocols the facilitator’s help. Further ongoing support is then pro-
used in clinical settings. vided for 4–6 weeks (negotiated with the individual care
home/service) to ensure appropriate implementation of the
24.3.1 INFORMATION REQUIRED treatment strategies, and tweaking is undertaken where
necessary.
We have already highlighted the need for caregivers to The Newcastle model (James, 1999, 2011) is well
know detailed information about both the behaviour and respected and has received empirical support, being used
the person. However, the rule of parsimony must be used in the Focussed intervention training and support (FITS)
in this area, whereby sufficient information is gained to trial (Fossey et al., 2006; Fossey and James, 2007). The use
guide treatment, but not so much that assessment materi- of this model serves at least two functions, summative and
als become over-burdensome and thereby inefficient. In this formative. The summative function is the establishment of a
section, we will examine the type of information that it is bespoke formulation of the person and his/her presentation.
useful to collect, and tools through which the details can be To obtain this a practitioner typically works with caregivers
collected and conceptualized. to collect personal and contextual information, including
data about physical and mental health status, medication
24.3.1.1 Background of person and social environment. A detailed analysis of the behav-
iour is also undertaken using behavioural charts.
Figure 24.1 outlines some of the background information one The formative function occurs in those caregivers who
can usefully obtain to understand the reason for a person’s BC, participate in the data collection. Their monitoring and care-
and Figure 24.2 outlines how to formulate a case of aggression ful observation facilitates their own pattern recognition and
(Mr A). This framework is taken from the Newcastle concep- hypothesis development with respect to the BC, and can
tualization, which is part of a 10- to 12-week package of care result in the development of a more positive attitude towards
known as the Newcastle model (James, 2011). Adherence to the person with dementia. Thus, the process of collecting
the model involves the following phases. data to inform the conceptual model frequently enhances
carer attitude, leading to a process termed repersonalization
24.3.1.1.1 PHASE 1. BACKGROUND ASSESSMENT (Kitwood, 1997).
The gathering of information relating to the person’s back- There are a number of other conceptual models direct-
ground from friends, family, carergivers and from case files ing the treatment of BC (Kitwood, 1997; Cohen-Mansfield,
and relevant databases. Assessment material can be gath- 2000; Volicer and Hurley, 2003; Aberdeen, 2010) – see James
ered by a specialist worker or can be a task assigned to a key (2011) for a review. The best developed of the latter is the
worker or named nurse. Cohen-Mansfield ‘unmet needs’ perspective, which has a
satisfactory evidence base (Cohen-Mansfield et al., 2007).
24.3.1.1.2 PHASE 2. ASSESSMENT OF TRIGGERS AND In addition to obtaining extensive biographical information
BEHAVIOUR about the person with dementia, this approach suggests that
Information is obtained to determine the events or situa- a BC is typically a poorly executed communication of an
tions that are likely to elicit the behaviours through discus- unresolved ‘need’.
sion and completion of behavioural charts. These charts also Cohen-Mansfield identified three potential links: BCs
provide in-depth information regarding the event itself. aimed at addressing the need (e.g. stealing food to assuage
250 Dementia

hunger), BCs aimed at communicating the need for assis- 24.3.1.2 Behavioural charts
tance (e.g. continuous shouting or buzzing for help) and
BCs when being thwarted at attempting to meet a need (e.g. BC tend to have patterns, made up of contexts and cues
an aggressive response when attempting to maintain one’s that typically trigger the person to act in a certain man-
self-esteem or when being prevented from leaving the build- ner. In the past, antecedents–behaviour–consequence
ing). Once the potential causes of the BCs have been estab- (ABC) charts have been used to analyse BC in detail. In
lished, the next step is to ask how we can help the person to more recent times, these charts have been tailored and
meet their need in a more functional manner. elaborated upon. Such charts are extremely helpful, but
A number of stand-alone tools have also been devised require staff training in order to be completed appropri-
to assist in the collection of personal information, such as ately. Without adequate training and reinforcement the
Dementia Passports (Alzheimer’s Society, 2013). These are charts can be burdensome and misleading. An example
biographic booklets compiled collaboratively by family, of a bespoke chart (Figure 24.4) seeks to gain informa-
with the aid of professionals – detailing personal history, tion about the person with dementia’s emotional states in
preferences, likes and dislikes. The booklets are carried rou- relation to behaviour and context; in keeping with a CBT
tinely by the person with dementia, aiding positive interac- perspective.
tions and facilitating communication.

Behavioural chart for …………………………..

Target behaviour ……………………………………

Please record any episodes of the above behaviour (day/night).

Aim—To record frequency and circumstances of incidents.

Date and time What was the person doing just before the incident? (A—antecedent)

Where did the incident


occurred?

What did you see happen? (B—actual behaviour)

Which staff were


involved (initials)?

What did the person say at the time of the incident?

How did the person appear at the time of the incident? (may be more than one tick)

Angry Frustrated
Anxious Happy
Bored Irritable
Content Physically unwell
Depressed Restless
Despairing Sad
Frightened Worried

How was the situation resolved? (C—consequences)

A—Antecedents are the features happening just prior to the emergence of the behaviour that may have
served to trigger or reinforce it.
B—Behaviours are simply the factual acts witnessed by the staff. The staff are taught not to interpret the
behaviour, rather provide factual details.
C*—Consequences are the responses of others to the behavioural disturbance. An analysis of this aspect
helps to determine what the person might be achieving by acting this way. Also by examining the
consequences, one can check the behaviour is not being inadvertently reinforced.

* Answers in this column give therapists ideas for interventions, and provide clues about which staff are
dealing with the situation well (or not so well).

Figure 24.4 Behavioural chart.


Treating problem behaviours in dementia by understanding their biological, social and psychological causes 251

24.3.1.3 Behavioural analytical tools approach is both a guide and a literature review of inter-
ventions (Brechin et al., 2013). It outlines four steps and
A Cochrane review (Moniz-Cook et al., 2012) recently dem- details when and where to use various physical, psycho-
onstrated the positive value of analysing the behaviours in logical and social approaches. It also provides guidance on
detail to help determine causes and then derive treatments. when comprehensive formulation strategies should be used
Three useful behavioural analytical tools are Dementia in contrast to less intensive methods of intervening. All of
Care Mapping (DCM) (Kitwood, 1997), Bird’s Treatment the steps invite practitioners to develop their assessments to
Protocol (McCabe et al., 2015) and Cohen-Mansfield’s look at the ‘under the surface’ elements of the Iceberg model
Treatment Route Exploratory Agitation (TREA) (Cohen- (Figure 24.1).
Mansfield, 2000). DCM is a multipurpose observational
tool, useful for assessment and training purposes. It links Step 1: The Stepped-Care model suggests that, as a matter
negative and positive care practices with levels of client of course, the initial approach should be recognizing
well-being. Bird’s protocol consists of an eight-page ques- common physical causes for changes in behaviour. For
tionnaire designed to be used with an informant. Questions example, the person should be screened for infection,
are asked about the person with dementia’s physical health, diabetes, constipation, thyroid disturbance and so on.
setting, triggers, strengths, weakness and guides the carer This may provide options for quick and targeted treat-
in the direction of the likely causes of the BC. ments while those caring for the person monitor their
In contrast, TREA was developed through a statistical physical condition and behaviour, and remain vigilant
methodology. It is used in the treatment of different types to risk.
of ‘agitation’ (verbal agitation [VA], physical non-aggres- Step 2: At this stage, emphasis is placed on examining
sion and aggressive behaviours) and suggests that these and changing environmental and caregiver practices.
behaviours have different aetiologies reflecting underlying Thus, the interventions develop from an assessment of
‘needs’, requiring different treatment strategies. For exam- possible environmental and management causes for
ple, Cohen-Mansfield’s (2000) empirical data suggest that the behaviour. Typical assessments tools would include
the needs that typically underlie VA are related to personal the use of behavioural charts and diaries such as those
discomfort, lack of social contacts and physical pain; with in Figure 24.4.
inactivity and depression also playing a role (Figure 24.5). Step 3: At this stage, specially trained health and social
The TREA approach utilizes a decision tree to arrive at care staff should be working with caregivers to assess
the most likely cause of the BC based on assessment of the for potential causal factors. Assessments might
behaviour, the environment and information about the include the use of specific dementia-related observa-
individual’s past preferences and needs. The decision tree is tional tools such as DCM and would always include
used to guide the caregiver to ascertain the need that is most systematic information gathering about the person’s
likely to be contributing to the behaviour. personal context and current behaviour and a review
of previous interventions and outcomes. The use of
24.3.2 TREATMENT PROTOCOLS decision trees, such as Cohen-Mansfield’s (2000)
TREA model (see Figure 24.5), and Bird’s Treatment
In 2013, Brechin et al. reviewed the above literature, and Protocol (McCabe et al., 2015), may lead to new and
other work relating to the treatment of BC, and developed more specifically targeted interventions. At this point,
a Stepped-Care approach for the treatment of problem- ‘as required’ medication may be considered if there is
atic behaviours. The document produced to support the high risk to others.

Verbal agitation

Need for social Need for stimulation?


Need for control?
Causes interaction? Boredom?

Identify meaningful Offer choices; provide


Try social interaction:
Intervention activities/physical tasks that allow for
real or taped
exercise control

Figure 24.5 Examples of approaches to the management of verbal agitation, from the Treatment Route Exploratory
Agitation model.
252 Dementia

Step 4: This stage requires the involvement of specialist they are locked. He refuses food and he is becoming distressed
practitioners using psychological formulation models with staff providing care to him in the bathroom, sometimes
to assess the needs of both the person with demen- trying to push them away and swearing at them.
tia and the care staff. This step is a more systemic
approach and the mechanism of change includes alter- Step 1 – Recognition: A physical assessment demonstrated
ations in the attitudes and attributions of care staff as that Mr B was constipated. He was prescribed laxative
well as their caregiving behaviour. All of the elements medication and staff were given advice about improving
in the Iceberg model are brought together in this step his diet and exercise regime. Although Mr B’s constipa-
as the dynamic between the person with dementia and tion resolved over the following few weeks, he contin-
their physical, social and care environment is targeted. ued to display agitation. He started to pace around the
The specialist practitioner will develop a ‘formulation’ lounge and refuse medication. He was verbally hostile
(see Figure 24.2), which is a story or hypothesis about and flicked his tie when people approached him.
the person’s behaviour taking into their personal and Step 2 – Low Intensity Interventions: An assessment of
environmental context. environmental factors associated with Mr B’s behav-
iour showed that he was largely showing this disturbed
The formulation is developed using psychological models behaviour in the lounge area, but was relatively calm
to understand behaviour; such as CBT or systemic therapy elsewhere. Behaviour charts also suggested that there
models. In most cases, the practitioner shares formulations were only a few members of staff who experienced
with the caregivers via a meeting often referred to as an this continued agitation and these caregivers tended
Information Sharing Session/Shared Formulation Session to employ some restricted practices on the unit. For
(James, 2011; Jackman, 2013). example, these staff preferred to have all of the resi-
In such meetings, the caregivers are guided through the dents in one place just before lunch so the medication
person’s difficulties in context and invited to make links to could be easily given out.
the needs their behaviour might represent. The practitioner   Mr B was often so upset around lunch time that
also considers how any current caregiving practices (i.e. his appetite was affected. His family told staff that he
management strategies) might be counterproductive to this was not a sociable person and had problems mixing
task taking into account the dynamic interplays described with other people. They wondered if he might feel
in Table 24.2. The latter processes are designed to foster col- intimidated by the behaviour of the other residents. As
laboration between the caregivers and practitioner to give a a result of the remarks made by the family, Mr B was
sense of ownership regarding the assessment, formulation given a dining table to himself and allowed to eat later
and subsequent interventions. than others, at a time when more assistance could be
Caregivers and/or specialist clinicians would target their given. Here, we see caregiving practices beginning to
interventions at the appropriate step based on the person’s take into account Mr B’s interpretation of his environ-
presentation, with more severe and complex cases entering ment and consequent emotional state.
immediately at Step 4. However, in some cases occasionally   After an initial improvement, Mr B’s behaviour
the nature of someone’s difficulties may lead them to prog- worsened. He started to ‘interfere’ with other people’s
ress through the stages, as outlined for illustration purposes belongings, ‘targeting’ particular staff he did not like.
in the following case study. He was repetitive and perseverative in speech and
swore at visitors. He was often tearful and distressed,
especially in the mornings.
Step 3 – High Intensity Interventions: At this stage, trained
24.4 CASE STUDY health and social care staff were brought in to work
with caregivers to assess for other causal factors. Mr B
The following case study illustrates how the Stepped-Care was reassessed by clinicians who reviewed his previ-
approach helps to define and address causal factors, delivering ous interventions. Bird’s Treatment Protocol was used
interventions more accurately targeted at these causes. At each to help identify potential causes. Via Bird’s checklist
step, the ‘under the surface factors’ – the person’s individual it was thought that some of his symptoms might be
biography, beliefs and preferences (Figure 24.1) – are taken related to an unresolved depression and so an antide-
into account as part of the assessment process. You will notice pressant was prescribed. Alongside this, it was noted
that, unless there are high levels of risk, antipsychotic medica- that Mr B’s memory and general cognitive functioning
tion is not recommended until the higher steps are engaged had declined. He had become dysphasic when at his
(Barnes et al., 2012), unless there is evidence of psychosis. most symptomatic, and it was at these times that he
Mr B is an 83-year-old gentleman with mixed Alzheimer’s/ became most distressed and threatening.
vascular dementia who lives in care. Over the last 3 weeks,   DCM was also used to help understand the envi-
staff working with him have noticed a change in his behav- ronmental impact on his behaviour, and it was fed
iour. They describe him walking around the unit in an agi- back to carers that they were making quite heavy cog-
tated manner trying to open doors and kicking them when nitive demands on him at times. For example, giving
Treating problem behaviours in dementia by understanding their biological, social and psychological causes 253

him complicated instructions such as ‘if you could (James, 2011) and Cohen-Mansfield’s needs-led frame-
wait here a moment, we’ll bring your tablets and then work (Cohen-Mansfield, 2000). In this particular case,
have you seated for your lunch’, which he misunder- the Newcastle model was chosen. Via this approach
stood, leaving him frustrated and embarrassed. On the clinician gathered information about Mr B’s life
reflection the staff noted that he had been an English and experiences and presented this to the caregivers
teacher who was used to being articulate and able, and in an information sharing session (ISS) along with
hypothesized that his communication difficulties may information about his cognitive impairment and
be a further source of frustration. Information about physical health problems. During the presentation, the
dementia was provided to staff in a teaching session to staff were invited to consider the contrast between the
help them make sense of Mr B’s overall presentation. previous life of this independent, proud and articulate
  Despite using the above tools and strategies, man, with his current position where he had little
Mr B continued to display acute high levels of agita- control over his environment and deficits, mean-
tion at least once a day. As such it was felt that a Step 4 ing he could not express this frustration or despair
approach was required, involving the development of appropriately. The caregivers were able to agree that
a bespoke formulation. This approach usually requires his ­agitation was probably caused by his need to regain
the assistance of specialist clinicians, who adhere to some control over his life, his need to express his
specific treatment protocols, resulting in the develop- distress and his need to have some private and quiet
ment of formulation-led targeted interventions. time. As a result, successful care plans were devised
Step 4 – Specialist Interventions: Mr B was reviewed by that recognized his individual needs (Table 24.3) but
a specialist clinician trained in the use of evidenced- which also reflected the high level of risk he could pose
based protocols, such as the Newcastle model when distressed.

Table 24.3 ‘Traffic Light’ care plans for Mr B


Well-being care plan Mr B is prone to infection, which can leave him feeling tired and out of sorts. Staff should
(green) regularly check his urine markers for infection.
Mr B suffers from constipation. A bowel movement chart should be kept. He should be offered
more high fibre foods at times when he is constipated. He does not like high fibre foods but
will recognize the need if you explain that he is constipated.
Mr B used to describe himself as a loner and does not like to eat with others. Mr B should be offered
a quiet area to eat his meals although accommodated in the main dining room if he prefers.
Mr B often feels low in mood especially in the mornings when he misses his family. His mood lifts
if he has access to activities he enjoys. As he has always enjoyed domestic tasks, staff should
engage Mr B in clearing breakfast away and setting tables for lunch. Mr B is then to have an
opportunity to have a drink with staff after helping.
Mr B has some difficulty understanding instructions and conversation. All staff should use
simplified language with Mr B and allow him time to respond to any requests or questions. NB.
DO NOT USE CHILDISH EXPRESSION OR TONE.
Mr B has always had control over his own life and this is difficult for him now. Staff should provide
opportunities wherever possible for Mr B to be involved in decision making.
Address escalating Mr B shows us he is feeling frustrated or crowded by flicking his tie and making growling noises.
problems (amber) At these times, staff should monitor the business of the environment and offer Mr B the
opportunity to move somewhere quieter. If he refuses, staff should offer others in the area an
alternative place to spend time.
Mr B may enter other people’s rooms and take their belongings. This is a sign that he is lost and
searching for familiar objects. This usually happens on days when his understanding is not so
good. Staff should adapt requests or tasks to his lowered level of functioning at these times.
For example, distract him from other people’s rooms by offering alternative objects rather than
by giving instruction.
Consider using an ‘as required’ dose of a Benzodiazepine. Ensure this is recorded monitored and
reported to the senior member of staff and family.
Risk management (red) At times when Mr B has become extremely distressed, it may be difficult to calm him. Provide 1:1
observation from a distance until the distressed period is over. Then approach and offer
reassurance.
If Mr B seems distressed by specific residents, ensure that another member of staff moves with
them to an alternative environment.
Mr B sometimes responds well to his son’s visits. Consider telephoning Mr B’s family for support.
254 Dementia

Teams using the Newcastle model have found it help- conducted into residential care settings, none employed
ful to develop specific care plans to meet well-being needs CBT, despite all aiming to change staff attitudes (Proctor
(thereby avoiding the person’s need to communicate dis- et al., 1999; Teri et al., 2005b; Fossey et al., 2006; Visser et al.,
tress) to recognize the onset of an episode of distress (and 2008; Chenoweth et al., 2009).
to take immediate opportunities to intervene on the envi- There has also been a number of systematic reviews
ronment or through different levels of care) and to manage regarding caregiver training and support programmes
highly risky situations. Sells and Shirley (2010) refer to these (Sörensen et al., 2002; Cooper et al., 2006). Despite Cooper’s
as ‘Traffic Light’ care plans. An example for Mr B appears review examining only anxiety in caregivers of people with
in Table 24.3. dementia, her categorization of the types of interventions
Cases frequently do not present themselves in such is a helpful summary of the approaches used. Here are the
an orderly and progressive fashion, but the case has been types of projects she identified, together with the num-
used to clarify the various steps. Importantly, the four- ber of trials she found under each heading: five CBT trials
step framework encourages people providing interventions (Gendron et al., 1986, 1996; Wilkins et al., 1999; Hébert
to consider some basic strategies first to address the most et al., 2003; Akkerman and Ostwald, 2004); four coping
common causes of BC. The later steps continue to incorpo- strategy/professional support; four alternative placements;
rate these basic physical and environmental causes as well three behavioural interventions; three respite; two relax-
as looking more closely at the meaning the person with ation/yoga; two exercise; one group counselling and one
dementia might be making of their environment. information technology support.
Her conclusion from the review was somewhat disap-
pointing with only Akkerman and Ostwald’s CBT study
showing significant effects in terms of anxiety. This was
24.5 TRAINING a 9-week CBT intervention that reduced anxiety in fam-
ily carers of people with dementia. Their approach also
In the United Kingdom, two-thirds of people with dementia included training in relaxation when appropriate.
are cared for at home, in many other countries the figure is The picture around the efficacy of training is compli-
far higher. Hence, supporting family carers is an essential cated due to the potential mismatch between attendee
part of any treatment regimen in BC. By keeping carers fit reports of the benefits and the actual improvements in out-
and mentally healthy, they can deal better with the tasks of comes for residents (McCabe et al., 2007). The growth in
supporting and caring for the person through any challeng- dementia-related training, following an increase in interest
ing period. Moniz-Cook et al.’s recent Cochrane review on from governments around the world, also raises problems
the use of behaviour therapy (functional analysis [FA]) of for measurement of effectiveness due to the lack of stan-
BC has shown that 66% of the 15 quality studies have been dardization of the programmes. Further, we lack clarity
conducted within ‘own-home’ settings. The overall findings around what mechanism is responsible for translating staff
of this systematic review demonstrated that the use of FA training to a change in the behaviour of the person with
behavioural methodologies can significantly reduce care- dementia.
giver burden. The approach we have described in this chapter suggests
Carer interventions take various forms. The psychother- that enhanced knowledge about dementia, coupled with a
apeutic approaches may use either a formal 1:1 therapy or greater understanding of the individual and their needs,
a group format, while education and training approaches enables staff in care homes to more effectively (1) establish
can be used to enhance skills, improve knowledge and atti- appropriate interventions to address the cause of the per-
tudes (Mittelman et al., 2004, 2007; McCabe et al., 2007). son’s behaviour and (2) to recognize their own role in main-
It is relevant to note each of the approaches has a different tenance cycles. Measurement of this change in focus of care
goal. The formal therapeutic approaches tend to be used staff is difficult as there are no well-established measures
to tackle psychiatric disorders, such as carer depression of attributional change for people working with this client
and anxiety. The teaching methods enhance coping abili- group (Shirley, 2005).
ties, and help the carers to develop practical caring skills. Finally, the evidence for the benefits of training aimed
One of the few approaches to have an empirically validated at reducing BC has repeatedly demonstrated that train-
manual for the treatment of BC is Teri’s Staff Training in ing on its own is not sufficient to produce lasting effects.
Assisted Residences (STAR) programme (Teri et al., 2005a, In a recent randomized control trial across Australian and
2005b, 2012). STAR teaches staff to understand and modify New Zealand care facilities, McCabe et al. (2014) found
their interactions with residents by identifying ABCs. The that improvements in levels of BC after training were only
programme also involves teaching on dementia, communi- maintained for the group receiving post-training clinical
cation skills and use of activities. Teri has conducted two support. Likewise, when community nurses were trained
Randomized Controlled Trials using STAR; one for families to support families caring for people with dementia whose
and one in a 24-hour residential setting (Teri et al., 2005a, behaviour challenged, the effect relied on extensive train-
2005b). Both of these studies are included in Moniz-Cooks’ ing and ongoing clinical supervisions and support (Moniz-
Cochrane, which shows that of the five quality studies Cook et al., 2008).
Treating problem behaviours in dementia by understanding their biological, social and psychological causes 255

REFERENCES Cooper, C., Katona, C., Orrell, M. and Livingston,


G. (2006). Coping strategies and anxiety in caregivers
Aberdeen, S. (2010). Concept mapping to improve of people with Alzheimer’s disease: The LASER-AD
Victorian residential aged care team learning and study. Journal of Affective Disorders, 90: 15–20.
problem solving for clients with behavioural and Eggenberger, E., Heimerl, K. and Bennett, M. (2013).
psychological symptoms of dementia. Unpublished Communication skills training in dementia care:
doctoral thesis. Australia: School of Public Health, La A systematic review of effectiveness, training con-
Trobe University. tent, and didactic methods in different care settings.
Akkerman, R. and Ostwald, S.K. (2004). Reducing anxiety International Psychogeriatrics, 25: 345–358.
in Alzheimer’s disease family caregivers: Effectiveness Ekman, P. (1973). Cross cultural studies of facial expres-
of a nine-week cognitive behavioural intervention. sion. In: P. Ekman (ed.), Darwin and Facial Expressions:
American Journal of Alzheimer’s Disease and Other A Century of Research in Review. New York, NY:
Disorders, 19: 117–123. Academic Press.
Alzheimer’s Society (2013). This Is Me. London, United Fossey, J., Ballard, C., Juszczak, E. et al. (2006). Effect
Kingdom: Alzheimer’s Society. Available at C:/Users/ of enhanced psychosocial care on antipsychotic use
User/Downloads/Alzheimer_s_Society_This_is_me_ in nursing home residents with severe dementia:
booklet_onlinev.3.pdf. Accessed 10 June 2016. Cluster randomised trial. British Medical Journal, 332:
Ballard, C., Day, S., Sharp, S. et al. (2008). 756–758.
Neuropsychiatric symptoms in dementia: Importance Fossey, J. and James, I.A. (2007). Evidence-Based
and treatment considerations. International Review of Approaches for Improving Dementia Care in Care
Psychiatry, 20: 396–404. Homes. London, United Kingdom: Alzheimer’s
Ballard, C. and Howard, R. (2006). Neuroleptic drugs Society.
in dementia: Benefits and harm. Nature Reviews Gendron, C., Poitras, L. and Dastoor, D.P. (1996).
Neuroscience, 7: 492–500. Cognitive-behavioural group intervention for spouse
Banerjee, S. (2009). The Use of Antipsychotic Medication caregivers: Findings and clinical considerations.
for People with Dementia: Time for Action. London, Clinical Gerontology, 17: 3–19.
United Kingdom: Department of Health. Gendron, C., Poitras, L., Engels, M. et al. (1986). Skills
Barnes, T. Banerjee, S. Collins, N. et al. (2012). training with supporters of the demented. Journal of
Antipsychotics in dementia: Prevalence and quality the American Geriatrics Society, 30: 875–880.
of antipsychotic drug prescribing in UK mental health Gitlin, L., Kales, H. and Lyketsos, C. (2012).
services. British Journal of Psychiatry, 210: 221–226. Nonpharmacologic management of behavioral
Beck, A.T. (1976). Cognitive Therapy and the Emotional symptoms in dementia. The Journal of the American
Disorders. New York, NY: International University Press. Medical Association, 308: 2020–2029.
Bird, M. and Moniz-Cook, E. (2008). Challenging behav- Hébert, R., Lévesque, L., Vézina, J. et al. (2003). Efficacy
iour in dementia: A psychosocial approach to inter- of a psychoeducative group program for caregivers
vention. In B. Woods and L. Clare (eds.), Handbook of demented persons living at home: A random-
of the Clinical Psychology of Ageing, Second Edition. ized controlled trial. Journals of Gerontology, Series
London, United Kingdom: Wiley. B: Psychological Sciences and Social Sciences, 58:
Brechin, D., Murphy. G., James I. and Codner, J. (2013). S58–S67.
Alternatives to Antipsychotic Medication: Psychological Hulme, C., Wright, J., Crocker, T. et al. (2010). Non-
Approaches in Managing Psychological and pharmacological approaches for dementia that
Behavioural Distress in People with Dementia, Vol. 207. informal carers might try to access: A systematic
London, United Kingdom: British Psychological Society review. International Journal of Geriatric Psychiatry, 25:
Publications, pp. 1–38. www.bps.org.uk. 756–763.
Chenoweth, L., King, M.T., Jeon, Y.H. et al. (2009). Caring Jackman, L. (2013). When, how and why does indirect
for Aged Dementia Care Resident Study (CADRES) formulation work? Proposing a ‘shared formulation’
of person-centred care, dementia-care mapping, and framework. Clinical Psychology Forum, 245: 25–31.
usual care in dementia: A cluster-randomised trial. James, I.A. (1999). Using a cognitive rationale to concep-
Lancet Neurology, 8: 317–325. tualise anxiety in people with dementia. Behavioural
Cohen-Mansfield, J. (2000). Nonpharmacological manage- and Cognitive Psychotherapy, 27: 345–351.
ment of behavioural problems in persons with demen- James, I.A. (2011). Understanding Behaviour in
tia: The TREA model. Alzheimer Care Quarterly 1 (4): 22. Dementia That Challenges. London, United Kingdom:
Cohen-Mansfield, J., Libin, A. and Marx, M. (2007). Jessica Kingsley.
Nonpharmacological treatment of agitation: A con- James, I.A. and Hope, A. (2013). Relevance of emo-
trolled trial of systematic individualized interven- tions and beliefs in the treatment of behaviours that
tion. Journal of Gerontology Medical Sciences, 62A: challenge in dementia patients. Neurodegenerative
906–918. Disease Management, 3: 575–588.
256 Dementia

King’s Fund (2009). Improving the Patient Experience: Moniz-Cook, E., Roberts-Stride, K., James, I. et al. (2012).
Developing Supportive Design for People with Functional analysis-based interventions in challeng-
Dementia. The King’s Fund’s Enhancing the Healing ing behavior in dementia. Cochrane Database, (1):
Environment Programme (2009–2012). London: King’s CD006929.
Fund. Proctor, R., Burns, A., Powell, H. et al. (1999). Behavioural
Kitwood, T. (1997). Dementia Reconsidered. Buckingham, management in nursing and residential homes: A ran-
United Kingdom: Open University. domized controlled trial. Lancet, 354: 26–29.
Lawrence, V. Fossey, J. Ballard, C. et al. (2012). Improving Sells, D. and Shirley, L. (2010). Person-centred risk man-
quality of life for people with dementia in care homes: agement: The traffic light approach. The Journal of
Making psychosocial interventions work. British Dementia Care, 18 (5): 21–23.
Journal of Psychiatry, 201: 344–351. Shirley, L.J. (2005). The development of a tool to measure
Levy-Storms, L. (2008) Therapeutic communication train- attributional processes in dementia care settings.
ing in long-term care institutions: Recommendations Clinical Psychology Forum, 154: 21–25.
for future research. Patient Education and Counselling, Sörensen, S., Pinquart, M. and Duberstein, P. (2002).
73: 8–21. How effective are interventions with caregivers?
McCabe, M.P., Bird, M., Davison, T.E. et al. (2014). An RCT An updated meta-analysis. The Gerontologist,
to evaluate the utility of a clinical protocol for staff 42: 356–372.
in the management of behavioral and psychological Teri, L., Huda, P., Gibbons, L. et al. (2005b). STAR:
symptoms of dementia in residential aged-care set- A dementia-specific training program for staff in
tings. Aging and Mental Health, 16: 1–9. assisted living residences. The Gerontologist, 45:
McCabe, M., Bird, M., Davison, T., Mellor, D. et al. (2015). 686–693.
An RCT to evaluate the utility of a clinical protocol for Teri, L., McCurry, S., Logsdon, R. and Gibbons, L. (2005a).
staff in the management of behavioral and psychologi- Training community consultants to help family mem-
cal symptoms of dementia in residential aged-care bers improve dementia care: A randomised controlled
settings. Aging Mental Health, 19(9): 799–807. trial. The Gerontologist, 45: 802–811.
McCabe, M.P., Davison, T.E. and George, K. (2007). Teri, L., McKenzie, G., Logsdon, R.G. et al. (2012).
Effectiveness of staff training programs for behav- Translation of two evidence-based programs for train-
ioural problems among older people with dementia. ing families to improve care of persons with dementia.
Aging and Mental Health, 11: 505–519. The Gerontologist, 52: 352–459.
Mittelman, M. S., Roth, D., Clay, O. and Haley, W. (2007). Visser, S., McCabe, M., Hudgson, C. et al. (2008).
Preserving the health of Alzheimer’s caregivers: Managing behavioural symptoms of dementia:
Impact of a spouse caregiver intervention. American Effectiveness of staff education and peer support.
Journal of Geriatric Psychiatry, 15: 780–789. Aging and Mental Health, 1: 47–55.
Mittelman, M.S., Roth, D., Haley, W. and Zarit, S. (2004). Volicer, L. and Hurley, A. (2003). Management of behav-
Effects of caregiver intervention on negative caregiver ioural symptoms in progressive degenerative demen-
appraisals of behaviour problems in patients with tias. Journal of Gerontology, 58A: 837–845.
Alzheimer’s disease: Results of a randomised trial. Whitaker, R., Fossey, J., Ballard, C. et al. (2014). Improving
Journal of Gerontology, 59B: 27–34. well-being and health for people with dementia
Moniz-Cook, E., Elston, C., Gardiner, E. et al. (2008). Can (WHELD): Study protocol for a randomised controlled
training community mental health nurses to support trial. Trials, 15: 28.
family carers reduce behavioural problems in demen- Wilkins, S., Castle, S., Heck, E. et al. (1999). Immune func-
tia? An exploratory pragmatic randomised controlled tion, mood, and perceived burden among caregivers
trial. International Journal of Geriatric Psychiatry, 23: participating in a psychoeducational intervention.
185–191. Psychiatric Services, 50: 747–749.
25
Treatments for behavioural and psychological
symptoms in Alzheimer’s disease and other
dementias

ANNA BURKE, WILLIAM J. BURKE AND PIERRE N. TARIOT

●● Aggressive (e.g. physical, such as hitting or slapping, or


25.1 INTRODUCTION verbal, such as obscenities or name calling)

Merriam et al. (1988) asserted that Alzheimer’s disease (AD) As Table 25.1 indicates, half or more of patients with
is ‘the most widely encountered cause of psychiatric pathol- dementia will show some feature of agitation at some point
ogy associated with a specific neuropathological substrate’. in the course of illness.
This is consistent with phenomenological studies and litera-
ture reviews, which generally converge on the conclusion Table 25.1 Summary of literature regarding
that roughly 90% of patients with dementia will develop psychopathology of dementia (per cent of patients
significant behavioural problems at some point in their affected among studies reviewed)
­illness (Tariot and Blazina, 1993). The ‘behavioural and
­psychological signs and symptoms of dementia’ (Finkel et al., Range Median
1996) do not typically meet criteria for discrete ­psychiatric 1. Disturbed affect/mood 0–86 19
disorders. They do, however, tend to occur in clusters of 2. Disturbed ideation 10–73 33.5
signs and symptoms that may vary among patients and over 3. Altered perception
time (Tariot and Blazina, 1993). Examples of these clusters Hallucinations 21–49 28
are provided in Table 25.1, based upon a review of published Misperceptions 1–49 23
studies regarding psychopathological changes in dementia 4. Agitation
that was performed in 1993. With experience, these clus- Global 10–90 44
ters of signs and symptoms can be both predicted and rec-
Wandering 0–50 18
ognized, and can be used as guideposts in the selection of
5. Aggression
appropriate therapy.
Verbal 11–51 24
This chapter will focus on agitation, which Cohen-
Mansfield and Billig (1986) defined as ‘inappropriate Physical 0–46 14.3
verbal, vocal, or motor activity unexplained by apparent Resistive/uncooperative 27–65 44
needs or confusion’. Cohen-Mansfield and Billig (1986) 6. Anxiety 0–50 31.8
empirically categorized agitated behaviours into three key 7. Withdrawn/passive behaviour 21–88 61
groups: 8. Vegetative behaviours
Sleep 0–47 27
●● Disruptive but not aggressive (e.g. attempts to leave, Diet/appetite 12.5–77 34
robes and disrobes, repeated complaints or questions) Source: Tariot, P.N. and Blazina, L. In J. Morris (ed.), Handbook of
●● Socially inappropriate (e.g. disrobing or urinating in Dementing Illnesses, New York, Marcel Dekker Inc.,
public) pp. 461–475, 1993.

257
258 Dementia

be trained to identify common triggers as well as con-


25.2 A RATIONAL APPROACH sequences of agitated behaviour and work with health
professionals to attempt to minimize the triggers and
This chapter proposes a systematic general approach to the maximize more positive circumstances.
evaluation and management of agitation that is based on
an elaboration of prior work (Leibovici and Tariot, 1988; If the behavioural disturbance is grossly aggressive or
Tariot, 1996, 1999); key aspects are described as follows: disruptive, and frankly dangerous, it may be necessary to
use antipsychotics on an emergency basis, preferably by
Define target symptoms. Review the available evidence mouth but sometimes parenterally. In rare cases, hospital-
and delineate the patient’s behaviour pattern, for ization is needed. Once the emergency is under control, it
example, ‘wanders without an apparent destination, is imperative to pay attention to the basics outlined earlier.
calls out repeatedly, bites and kicks during care’.
Establish or revisit medical diagnoses. It is a truism in
geriatrics that delirium is a frequent cause of incident
25.3 DEVELOPMENT OF THE
agitation and must always be suspected. Common cul-
prits include respiratory or urinary infections, dehy-
PSYCHOBEHAVIOURAL
dration, occult trauma, electrolyte disturbances or side METAPHOR
effects of medications. Such medical problems should
be treated specifically and the patient monitored care- For persisting non-emergent problems where non-­
fully, ideally without the need for other medications. pharmacological interventions have been exhausted, we adopt
Establish or revisit neuropsychiatric diagnoses. It is also an approach that begins with a definition of a target symptom
possible that disturbed behaviour may reflect a long- pattern that is at least roughly analogous to a drug-responsive
standing, recurrent or new-onset discrete psychiatric syndrome. We refer to this as the ‘psychobehavioural meta-
disorder. When a specific disorder is identified, it phor’ (Leibovici and Tariot, 1988). We then match the domi-
should be specifically treated and monitored. nant target symptoms (the metaphor) to the most relevant
Assess and reverse aggravating factors. Examples might drug class. For example, in the case of a verbally and physically
include deficits in hearing or vision, or an environ- agitated patient who is also irritable, negative, has become
ment that is too dark, noisy or cold. Disrupted daily socially withdrawn and appears dysphoric, we might well
routines may present a remediable factor. It is impor- first undertake a trial of an antidepressant. Conversely, if the
tant to look hard for environmental triggers, sleep patient showed ‘agitation’ in the context of increased motor
deprivation or biological triggers such as pain or activity, loud and rapid speech and lability of affect, we might
constipation. consider early use of a mood stabilizer. The ‘aggressive’ patient
Adapt to specific cognitive deficits. Some patients may whose hostile actions are associated with delusions is likely to
be able to respond to verbal reassurance and redirec- be treated first with an antipsychotic. Although this approach
tion, whereas others may not. Pain or distress may to drug selection is face valid, there is limited empirical sup-
need to be inferred from behaviour or facial expres- port for it (Lyketsos et al., 2000; Devanand, 2000; Tariot et al.,
sion. Some may need constant environmental cueing 2001a, 2002a; De Deyn et al., 2003). Thus, the validity of the
to organize their behaviour. It is important to identify approach that we have endorsed for some years, and that is
each patient’s cognitive strengths and weaknesses, reflected in most consensus guidelines (Doody et al., 2001;
attempting to capitalize on residual strengths and Alexopoulos et al., 2005; APA Work Group on Alzheimer’s
avoid weaknesses. Disease and other Dementias, Rabins et al., 2007; Rabins et al.,
Identify relevant psychosocial factors. It may be easy to 2014), has not been established empirically. Furthermore, the
overlook the fact that a patient with dementia may be use of medications to treat any aspect of psychopathology is
suffering anxiety, sadness, denial or fury in response not recognized by the U.S. Food and Drug Administration
to major and minor life events. In some long-term and would thus be considered ‘off-label’.
care facilities, for instance, moving from one unit to The approach adopted in this chapter more or less reflects
another can be associated with perceived loss of status the position of the American Geriatrics Society and American
as well as contact with familiar people. It is easy to Association for Geriatric Psychiatry consensus statement
overlook this as a precipitant of ‘agitation’. (2003) and the American Psychiatric Association (Rabins
Educate caregivers. Caregivers can learn that change in et al., 2007, 2014) with respect to the use of target symptoms
behaviour usually has meaning and that the behav- to guide selection of drug class. We emphasize selection of a
iours can occur in discernible patterns. This helps medication with at least some empirical evidence of efficacy
caregivers interact in a more supportive manner. and with the highest likelihood of tolerability and safety. We
In the process, caregivers can be taught to employ observe some general rules such as starting with low doses
essential behaviour management principles, learn- and escalating slowly, assessing target symptoms as well as
ing, for instance, that their own behaviour is a major toxicity and discontinuing the medication if it is harmful or
determinant of the patient’s behaviour. Caregivers can ineffective. Even when a medication is helpful at sub-toxic
Treatments for behavioural and psychological symptoms in Alzheimer’s disease and other dementias 259

doses, an empirical trial is often performed in reverse and the Table 25.2 Selected current pharmacological treatments
patient monitored for recurrence of the problem. This type of for agitation in dementia
approach is actually mandated in the nursing home setting
Suggesting starting and (maximal)
in the United States by Federal regulations created in 1987.
Drug and class daily dose (mg/day)a
Sometimes several medications need to be tried in series
before a successful one is identified; sometimes combinations Antipsychotics
are warranted; sometimes no medication is found that is Traditional
helpful. We hold the view that best practice includes routine Haloperidol 0.25–0.5 (2–4)
use of cholinesterase inhibitors (ChEIs) and/or memantine Thioridazine 12.5–25 (50–100)
in most patients with AD as well as certain other dementias, Thiothixene 0.5–1 (2–4)
meaning that psychotropic use occurs against this backdrop. Atypical
Evidence suggests that ChEIs and/or memantine may be of Clozapine 6.25–12.5 (25–100)
benefit in reducing less severe agitated or aggressive behav-
Olanzapine 2.5 (5–10)
iours in patients with AD or possibly delaying their emer-
Quetiapine 25 (50–200)
gence (Birks and Flicker, 1998; Trinh et al., 2003; Cummings
Risperidone 0.5 (1–2)
et al., 2006 a,b; Wilcock et al., 2008). The trials conducted
Aripiprazole 5 (15)
with these agents were aimed towards obtaining regulatory
Ziprasidone Unknown
approval and not informing clinical practice regarding issues
such as the advantages or disadvantages of one agent versus Anxiolytics and sedatives
the other for relief of behavioural symptoms, or of using psy- Benzodiazepines
chotropics in combination with these agents. Practice will Alprazolam 0.5 (1–2)
outstrip evidence for the foreseeable future. Lorazepam 0.5 (2–4)
Oxazepam 10 (40–60)
Non-benzodiazepines
25.4 SPECIFIC CLASSES Zolpidem 2.5 (5)
OF PSYCHOTROPICS Buspirone 10 (30–60)
Antidepressants
The remainder of this chapter reviews the most relevant Trazodone 25–50 (200–300)
classes of agents and specific agents within those classes, SSRIs
focusing on information from about the last 5 years and on Citalopram 10 (20)
older studies that remain as crucial guides to practice. Much Fluoxetine 10 (20)
of the older literature was reviewed previously in the last Sertraline 25–50 (100–200)
edition of this textbook (Profenno et al., 2005); this chapter Anticonvulsants
builds on that as well as some of our earlier work (Tariot,
Carbamazepine 50–100 (300–500)
1999; Loy et al., 1999; Rosenquist et al., 2000; Profenno and
Divalproex 125–375 (500–1500)
Tariot, 2004). Table 25.2 provides an overview of individual
β-Blockers
agents and typical starting doses, proposed based upon a
mixture of data and clinical experience. Propranolol 20 (50–100)
Others
Selegiline 10
25.5 CONVENTIONAL Oestradiol/ 0.625/2.5
ANTIPSYCHOTICS progesterone
Source: Loy, R., et al. In I.R. Katz, Oslin, D. and Lawton, M.P. (eds.)
Within the context of the ‘metaphor’ approach, antipsy- Annual Review of Gerontology and Geriatrics: Focus on
Psychopharmacologic Interventions in Late Life. New
chotics would be used first for treatment of agitation that York, NY: Springer, 1999.
included psychotic features. In reality, antipsychotics have a Suggestions are based on published data as well as anecdotal

been used and studied in patients with a wide range of psy- experience, and should be regarded accordingly.
chopathology. There are two main classes of antipsychotics:
so-called conventional or first generation antipsychotics in the Schneider et al. (1990) meta-analysis. These authors
and the newer atypical or second generation agents. Prior also concluded that type and potency of agent did not influ-
reviews of the conventional agents have concluded that the ence response. They reported an average therapeutic effect
effects of these agents were consistent but modest and that (antipsychotic versus placebo) of 26%, with placebo response
no single agent was better than another (Wragg and Jeste, rates ranging from 19% to 50%. Side effects were reported to
1989; Schneider et al., 1990) (Table 25.3). occur more often on drug than placebo (mean difference 25%).
Lanctôt et al. (1998) performed a meta-analysis includ- These meta-analyses did not include five more recent studies
ing some studies that were less rigorous than those included (Devanand et al., 1998; De Deyn et al., 1999; Allain et al., 2000;
260 Dementia

Table 25.3 Conventional antipsychotics


Dose (mg/day), Adverse
Study Diagnosis N Design duration Response effects
Haloperidol
Schneider Dementia 252 (in 7 Meta-analysis 2.5–4.6, Little benefit beyond Not reported
et al. DB, PC of 33 haloperidol, placebo across all
(1990) trials in neuroleptic 3–8 weeks neuroleptics;
primary trials average effect 18%
dementia) greater than
placebo
Devanand AD 71 Randomized 2–3 or 0.5–0.75, Decreased psychosis, Moderate-to-
et al. DB, PC, 6 weeks aggression, severe EPS
(1998) parallel agitation with at high dose
group higher dose only, (20%)
55%–60% response
vs. 25%–35% low
dose response vs.
25%–30% placebo
Christensen Organic 48 Randomized Mean 0.64, 6 No difference None seen at
et al. mental crossover to weeks between this low dose
(1998) syndrome alprazolam treatments on CGI,
SCAG
DeDeyn AD, 344 Randomized Mean 1.2, 13 Significantly greater EPS 22%,
et al. vascular, MC, DB, weeks improvement for somnolence
(1999) mixed parallel haloperidol vs. (18%
group placebo on haloperidol
risperidone BEHAVE-AD and vs. 4%
or placebo CMAI, decreased placebo)
physical and verbal
aggression (72%
risperidone vs. 69%
haloperidol vs. 61%
placebo on
BEHAVE-AD)
Allain AD, VD, 306 Randomized 100–300 tiapride Significantly Less EPS with
(2000) mixed MC, DB, or 2–6 improved CGI and tiapride than
PC, parallel haloperidol, MOSES irritability/ haloperidol
group 21 days aggress-iveness
sub-score for both
drugs compared to
placebo, no
differences
between drugs
Teri (2000) AD 149 Randomized 1.8 haloperidol, Reduced agitation Less EPS with
DB, PC, 200 trazodone not significantly behaviour
parallel or behaviour different between management
group management treated and techniques
techniques, placebo groups
16 weeks
Tariot Probable 284 Randomized, Mean 1.89, 8 No change in EPS, sedation
(2002a) AD with PC, DB vs. weeks psychosis, some
psychosis quetiapine decrease in
(n = 94 on agitation
haloperidol)
Treatments for behavioural and psychological symptoms in Alzheimer’s disease and other dementias 261

Teri et al., 2000; Tariot et al., 2002a). The data from these newer that caution is warranted. For instance, rates of TD are five-
studies generally conform to the results of the meta-analyses. to six-fold greater in older than in younger populations after
Lonergan et al. conducted a pooled analysis of five randomized treatment with conventional agents (Jeste et al., 1995). Recent
trials of haloperidol versus placebo that included two studies studies also indicate higher mortality rates in patients with
not incorporated in the previous meta-analysis, which found dementia receiving conventional antipsychotics versus those
decreased aggression but no significant improvement in agi- receiving atypical agents (Kales et al., 2012; Huybrechts et al.,
tated symptoms overall among the patients treated with halo- 2012). For practical purposes, side effects typically guide
peridol. There was also a noteworthy increase in adverse side selection of these agents when used in patients with demen-
effects such as rigidity and bradykinesia compared to placebo. tia. A clinical issue that is of particular importance is demen-
Reviews regarding conventional agents cite side effects tia with co-occurring extrapyramidal disorders such as
including akathisia, parkinsonism, tardive dyskinesia (TD), dementia with Lewy bodies, where occasional extraordinary
sedation, peripheral and central anticholinergic effects, sensitivity to conventional agents is seen (Ballard et al., 1998).
postural hypotension, cardiac conduction defects and falls For all these reasons, it was hoped that atypical antipsychot-
(Lanctôt et al., 1998; Lonergan et al., 2002). Based on limited ics would have special utility in patients with dementia.
efficacy and a high likelihood of toxicity, Lonergan et al. con-
cluded by recommending that haloperidol be used sparingly,
on a case by case basis, with monitoring of side effects. Most
of these data come from short-term controlled trials; evi- 25.6 ATYPICAL ANTIPSYCHOTICS
dence regarding long-term safety is generally lacking. Data
available from other elderly populations, however, indicate Atypical antipsychotics are listed in Table 25.4.

Table 25.4 Atypical antipsychotics

Dose (mg/day),
Study drug Diagnosis N Design duration Response Adverse effects
Olanzapine
Satterlee et al. AD 238 Randomized, 1–8, 8 weeks No benefit vs. placebo None reported vs.
(1995) MC, PC, DB placebo
Street et al. AD, 206 Randomized, 5, 10 and 15, Decreased agitation, Dose-dependent
(2000) moderate- MC, PC, DB, 6 weeks delusions, sedation,
to-severe parallel group hallucinations on abnormal gait
NPI-NH at 5 and 10
(but not 15) mg/day
vs. placebo
Street et al. AD 105 Open as Flexible 5–15, Significantly improved Somnolence,
(2001) follow-up to 18 weeks agitation and other accidental injury,
previous symptoms by NPI/ rash
6 week DB trial NH and other scales
Meehan et al. AD, VaD or 272 Randomized, intramuscular 5, Decreased agitation at No significant
(2002) mixed with MC, PC, DB 2.5, or 1 2 and 24 h difference
acute lorazepam, between
agitation 2 h and prn placebo and any
×2 within 20 h active treatment
Quetiapine
Tariot et al. Various 184 Open, MC 12.5–800 Significant decreases Somnolence (31%),
(2000a) psychotic (median in BPRS total and accidental injury
disorders 137.5), CGI Severity (24%), dizziness
52 weeks (17%)
Scharre and AD 10 Open 50–150 (mean = Improved NPI for Drowsiness (40%),
Chang, 100), delusions and weight gain
(2002) 12 weeks agitation or (40%)
aggression
Davis and DLB 10 Open 25–150 (50 mg Significant reduction in No significant
Baskys modal), total NPI and increase in EPS
(2002) 12 weeks agitation subscales
of NPI and BPRS
(Continued )
262 Dementia

Table 25.4 (Continued) Atypical antipsychotics

Dose (mg/day),
Study drug Diagnosis N Design duration Response Adverse effects
Tariot et al. Probable AD 284 Randomized, Mean 100, No change in Sedation
(2002a) with DB, PC vs. 8 weeks psychosis, some
psychosis haloperidol decrease in agitation
(n = 91 on
quetiapine)
Ballard et al. AD, dementia 93 Randomized, Mean 100, No change in Cognitive decline
(2005) DB, PC 26 weeks psychosis, some
decrease in agitation
Tariot et al. Possible/ 284 Randomized, Mean 96.9, Improvement in Somnolence
(2006) probable MC, PC, DB 10 weeks agitation and
AD psychosis
Zhong et al. Dementia 333 DB, fixed dose Mean, 10 weeks Clinically greater No significant
(2007) improvements in difference
Positive and between
Negative Syndrome placebo and any
Scale–Excited active treatment
Component CGI-C
and CGI-C response
rates
De Deyn AD 100 Randomized, Mean 143.6, NPI frequency × None reported vs.
et al. (2012) DB, double 142, 6 weeks severity total, placebo
dummy NPI-NH version,
CMAI, CGI-S CGI
scores improved
Risperidone
Herrmann AD, vascular, 22 Case reports Mean 1.5, Improvement CGI (17) EPS (11), sedation
et al. DLB 1 week, (1), drooling (1)
(1998b) 10 months
Lavretsky and AD, vascular, 15 Open 0.5–3, 9 weeks Decreased agitation (in Sedation (5),
Sultzer PD, (concurrent all 15) akathisia (1)
(1998) Progressive psychotropics
Supranuclear allowed)
Palsy, mixed
De Deyn AD, VaD, 344 Randomized MC, Mean 1.1, Decreased physical EPS not different
et al. (1999) mixed DB, parallel 13 weeks and verbal from placebo,
dementia group vs. aggression (72% somnolence
placebo or risperidone vs. 69% (12% risperidone
haloperidol haloperidol vs. 61% vs. 4% placebo)
for placebo)
Katz et al. AD, VaD, 625 Randomized MC, 0.5, 1.0, 2.0, Dose-dependent Somnolence, EPS
(1999) mixed PC, DB, 12 weeks decrease in 21% (on 2 mg/
dementia parallel group aggression and day, compared
(lorazepam, psychosis, significant with 12% on
benzatropine at 1 and 2 (but not placebo), mild
and chloral 0.5 mg/day) peripheral
hydrate oedema in some
allowed PRN)
Brodaty et al. AD, VaD, 345 Randomized, Mean 1.0, Significant reductions Somnolence,
(2003) mixed MC, PC, DB 12 weeks in CMAI total urinary tract
aggression score infection
and BEHAVE-AD
aggressiveness
(Continued)
Treatments for behavioural and psychological symptoms in Alzheimer’s disease and other dementias 263

Table 25.4 (Continued) Atypical antipsychotics

Dose (mg/day),
Study drug Diagnosis N Design duration Response Adverse effects
Schneider AD, 5110 Meta-analysis of Mean 0.97 ± Decreased agitation Somnolence, UTI,
et al. dementia randomized, 0.74 mg, EPS, gait
(2006a, PC, DB, 16 weeks abnormalities
2006b) parallel-
group trials
Devanand AD 180 Open Mean 0.97 mg, In patients with
et al. (2012) 32 weeks psychosis or
agitation that had
responded to
risperidone therapy
for 4 to 8 months,
discontinuation of
risperidone was
associated with an
increased risk of
relapse

Aripiprazole
DeDeyn et al. AD 108 Randomized, Mean 10 mg, No significant drug- Somnolence (in 1%
(2003) PC, DB 10 weeks placebo difference of placebo and
on primary end 8% of drug
point. Some groups)
secondary measures
showed drug-
placebo differences
at some time points.
Breder et al. AD 487 Randomized, Mean 2, 5, Significant drug- No significant
(2004) PC, DB 10 mg, 10 placebo difference differences in
weeks on primary end rates of adverse
point in 10 mg/d events across
group. Significant groups
effects on CGI-S and
total BPRS in
10 mg/d group;
significant effects in
5 and 10 mg/d
groups on CMAI
Streim et al. AD 256 Randomized, Mean 8.6 mg/ No significant drug- Somnolence (4% in
(2008) PC, DB day, 10 weeks placebo difference pla and 14% in
on primary end aripiprazole
points. groups)
Mintzer et al. AD 487 Randomized, Mean 2, 5, 10 mg/day was CVA
(2007) MC, PC, DB 10 mg, efficacious and safe
10 weeks for psychosis
associated with AD,
significantly
improving psychotic
symptoms, agitation,
and clinical global
impression
264 Dementia

25.6.1 CLOZAPINE conducted of any form of treatment for agitation or aggres-


sion associated with dementia. A conservative interpreta-
Only anecdotes and case reports are available; none are tion of the available evidence would be that the efficacy of
new and are the same as mentioned in the previous chap- risperidone is roughly equivalent to that of haloperidol.
ter. Individual patients showed partial improvement in agi- The tolerability of risperidone appears to be better at doses
tation or psychosis, whereas others developed side effects below 2 mg/day, with low-to-moderate risk of dose-related
including sedation and anticholinergic effects. While not parkinsonism and sedation and numerically higher rates of
mentioned specifically in these reports, agranulocyto- peripheral oedema and sedation in patients treated with ris-
sis and seizures have been reported in other populations, peridone versus placebo.
the former requiring regular haematological testing for In 2003, the Food and Drug Administration (FDA) issued
months. Dose–response issues are unresolved. In using this a warning titled ‘Cerebrovascular Adverse Events, Including
medication, we err on the side of caution by starting at 6.25 Stroke, in Elderly Patients with Dementia’ prompting con-
or 12.5 mg/day. Based on the relative lack of motor toxicity cerns regarding the drug’s safety. It referred to CVAEs
encountered with clozapine, a case can be made to select this (stroke, transient ischaemic attack), including fatalities,
agent preferentially in patients with dementia and extrapy- reported in patients (mean age 85 years; age range 73–97)
ramidal disorders. However, in view of the lack of informa- in trials of risperidone in elderly patients with dementia-
tion regarding dose–response, the risk of side effects in the related psychosis and/or agitation. In placebo-controlled
elderly and the lack of controlled data regarding efficacy and trials, a significantly higher incidence of CVAEs was noted
tolerability, most clinicians are likely to choose from among in patients treated with risperidone compared to those
the newer atypical agents first. treated with placebo (Woolerton, 2002). Risperidone was
associated with a threefold increased risk of serious CVAEs
25.6.2 RISPERIDONE compared with placebo. Soon thereafter, a similar warning
was applied to olanzapine and aripiprazole. The increased
The earliest exploratory studies indicated that doses in the risk for CVAEs was determined from research clinical trials
range of 0.5–2 mg/day might be best tolerated, with dose- and is based on unbiased estimates. However, the construct
related risk of motor toxicity (especially in patients with ‘CVAEs’ was defined after the trials were completed and was
extrapyramidal disorders). Sedation, peripheral oedaema based on spontaneously reported adverse events and inter-
and orthostatic hypotension were sometimes reported. pretation of the risk was limited by the fact that the studies
Use of risperidone can result in increased prolactin levels were not designed to investigate a causal link with CVAEs
in other populations, although the clinical significance (Burke and Tariot, 2009).
of this in patients with dementia is uncertain. We do not
routinely check serum prolactin levels and would only 25.6.3 OLANZAPINE
take action in response to elevated levels associated with
symptoms associated with its use (e.g. galactorrhoea in Studies suggest benefit of olanzapine in treating agitation
women, sexual dysfunction in men) or with symptoms in patients with dementia. A 6-week, randomized, parallel-
indicative of a possible hypothalamic tumour (e.g. diplo- group, multicentre study of olanzapine in 206 nursing home
pia). The earlier open trials and case series indicated that residents with dementia and agitation and/or psychosis was
risperidone may decrease agitation, aggression or psychosis completed using fixed doses of 5, 10 and 15 mg/day ver-
in patients with dementia, spawning more definitive tri- sus placebo (Street et al., 2000). In patients receiving 5 and
als. Three large multicentre trials of risperidone have been 10 mg/day, significant improvement in agitation and aggres-
completed in nursing home patients with relatively severe sion compared to placebo was seen, and patients receiving
dementia (Katz et al., 1999; De Deyn et al., 1999; Brodaty 5 mg/day also demonstrated significant improvement com-
et al., 2003) and one in outpatients (Mintzer et al., 2004). pared with placebo in broader assessments of psychopathol-
A meta-analysis of results from these four studies revealed ogy. Sedation and postural instability were observed at all
that risperidone significantly reduced measures of agitation doses in at least 25% of subjects. Some of the patients from
and/or psychosis in comparison with placebo (Schneider this trial received follow-up open-label, flexible-dose treat-
et al., 2006a, 2006b). Risperidone-treated patients displayed ment for 18-weeks; results were consistent with the findings
positive effects on measures of psychosis. In patients with from an earlier paper (Street et al., 2001).
AD who had psychosis or agitation that had responded to Meehan et al. (2002) studied acute treatment of agita-
risperidone therapy for 4 to 8 months, discontinuation of tion with intramuscular olanzapine in 272 inpatients or
risperidone was associated with an increased risk of relapse nursing home residents with AD and/or vascular dementia.
(Devanand et al., 2012). The main side effects in these trials Olanzapine was superior to placebo in treating agitation at
were somnolence, extrapyramidal symptoms (EPS), abnor- 2 and 24 hours. At present, the utility of this formulation
mal gait and peripheral oedema in addition to cerebrovas- appears limited due to restricted distribution and poten-
cular adverse events (CVAEs) and death described later. tially onerous side effect profiles.
In the aggregate, these risperidone studies comprise Additionally, a cautionary note is warranted when
the largest placebo-controlled, multicentre studies ever ­considering using this agent in patients with dementia.
Treatments for behavioural and psychological symptoms in Alzheimer’s disease and other dementias 265

I­n January 2004, Eli Lilly issued a warning of increased abnormalities, including neutropenia and eosinophilia
incidence of CVAEs in patients with dementia treated with (De Deyn et al., 2012). However, confounding factors made
olanzapine. The medication has also been widely criticized it difficult to establish relationship to quetiapine treatment.
for its potential to cause metabolic syndrome, at least in
younger populations, with limited data in the elderly thus 25.6.5 ZIPRASIDONE AND ARIPIPRAZOLE
far which have come chiefly from the Clinical Antipsychotic
Trial of Intervention Effectiveness – Alzheimer’s Disease There are no geriatric-specific studies of ziprasidone and
(CATIE-AD) trial (see the following). none in patients with dementia. However, case reports have
suggested both oral and injectible forms of the medication
25.6.4 QUETIAPINE FUMARATE may be well tolerated and of some benefit in the treatment
of agitation in this population.
Two open trials and a case series with quetiapine within Results from a placebo-controlled trial of aripiprazole in
a dose range of 25–800 mg/day suggested possible behav- 208 outpatients meeting new clinical criteria for psychosis
ioural benefits for agitation (Tariot et al., 2000a; Scharre of AD (Jeste and Finkel, 2000; De Deyn et al., 2005) showed
and Chang, 2002; Davis and Baskys, 2002). A 10-week that the medication was generally well tolerated, with
multicentre placebo-controlled trial of quetiapine versus numerically more frequent sedation in the active treatment
haloperidol was conducted in elderly nursing home patients group. The primary outcome, a measure of psychosis, did
with psychosis that was operationally defined. These crite- not show a difference between drug and placebo although
ria were implemented prior to the development of clinical there was some behavioural benefit on secondary mea-
criteria for the psychosis of AD (Jeste and Finkel, 2000). sures. A multicentre, randomized, double-blind, placebo-
This trial is notable in that it may have been the first large controlled trial of three fixed doses of aripiprazole (2 mg,
placebo-controlled study of antipsychotics in patients with 5 mg and 10 mg) for the treatment of psychosis in patients
dementia selected on the basis of psychotic symptoms. with AD indicated that 10 mg daily was safe and efficacious
Flexible doses of the medication were permitted; the mean revealing significant improvement in psychosis, agitation
daily dose of haloperidol was 2 mg/day at end point, while and clinical global impression (Mintzer et al., 2007).
that of quetiapine was about 120 mg/day. None of the treat-
ment groups differed with respect to reduction in measures 25.6.6 ATYPICALS: SUMMARY
of psychosis, which was the primary outcome of the trial,
whereas there was evidence of reduced agitation on some The data indicate that atypical antipsychotics as a class are
measures with both haloperidol and quetiapine treatment likely better tolerated than typical antipsychotics and at
but not placebo. Tolerability of quetiapine was superior to least as efficacious. There appear to be modest differences
that of haloperidol; however, evidence of improvement in within the class of atypicals in terms of effectiveness, tol-
agitation was inconsistent. erability and side effect profile. An Agency for Healthcare
Data from studies evaluating the efficacy of quetiapine Research and Quality (AHRQ) review in 2011 estimated
remain mixed. Two meta-analyses (Ballard and Waite, 2006; that among the placebo-controlled trials of elderly patients
Schneider et al., 2006a, 2006b) that included three trials with dementia reporting a total/global outcome score that
(Ballard et al., 2005; Tariot et al., 2006; Zhong et al., 2007) includes symptoms such as psychosis, mood alterations and
revealed modest improvement in agitation and aggression at aggression, small but statistically significant effect sizes
the 200 mg dose, but did not significantly improve psychotic ranging from 0.12 and 0.20 were observed for aripiprazole,
symptoms in AD patients. The lack of antipsychotic effect olanzapine and risperidone (Maglione et al., 2011). To
may have been related to the relatively low dosing (<200 mg) address this type of issue, the National Institute of Mental
and may suggest that higher doses might have been neces- Health conducted the CATIE-AD, which was designed to
sary. Quetiapine had more discontinuations due to lack of compare the effectiveness of atypical antipsychotic medi-
response but fewer discontinuations due to adverse effects. cations and placebo in patients with AD and psychosis
Both studies identified quetiapine as the atypical agent with or agitated/aggressive behaviour (Schneider et al., 2001,
the lowest mortality risk and lowest risk of injury, although 2006a, 2006b). The study included 421 outpatients with
an increased risk of cognitive decline was noted in one AD and psychosis or agitated/aggressive behaviour. In the
small study (Ballard et al., 2005). A retrospective cohort first phase of the trial, patients were assigned randomly to
study analysing data from the U.S. Department of Veterans masked, flexible-dose treatment with olanzapine, quetiap-
Affairs for patients with dementia treated with antipsychot- ine, risperidone or placebo for up to 36 weeks (Schneider
ics (risperidone, quetiapine, olanzapine or haloperidol) or et al., 2006b). The primary measure of effectiveness, time-
valproic acid, indicated reduced mortality rates in patients to-discontinuation for any reason, was not significantly
receiving quetiapine compared with those receiving other different between the treatment conditions, with median
antipsychotics (Kales et al., 2012). A randomized, double- treatment durations of 8.1 weeks for olanzapine, 5.3 weeks
blind, parallel-group study comparing the tolerability of for quetiapine, 7.4 weeks for risperidone and 8.0 weeks
extended-release quetiapine with immediate-release que- for placebo. The median time-to-­d iscontinuation due to
tiapine in patients with dementia identified haematological lack of efficacy was significantly longer with olanzapine
266 Dementia

and risperidone (22.1 and 26.7 weeks, respectively) ver- antipsychotics were associated with worsening cognition
sus quetiapine and placebo (9.1 and 9.0 weeks, respec- in multiple domains at a magnitude of one year’s deterio-
tively). However, patients treated with an atypical had a ration compared with placebo (Vigen et al., 2011). A 2006
shorter time to discontinuation due to adverse events or meta-­a nalysis also demonstrated a significant increase in
drug intolerability compared with placebo. Time to dis- respiratory tract and urinary tract infections and periph-
continuation due to intolerability occurred in 24% of eral oedema in people treated with risperidone as com-
patients receiving olanzapine, 18% receiving risperidone, pared with placebo (Ballard and Waite, 2006).
16% receiving quetiapine and 5% receiving placebo. Safety In 2005, a U.S. Food and Drug Administration warn-
findings were generally consistent with reports of prior ing highlighted a significant increase in mortality risk for
studies. Although the trial has frequently been misinter- patients with AD treated with atypical antipsychotic drugs
preted as showing that these medications are ineffective, a compared with individuals receiving placebo in 17 ran-
more accurate analysis reveals that there is a high rate of domized, controlled trials (Center for Drug Evaluation and
adversity that offsets evidence of efficacy, but that a minor- Research, n.d.). It stated that elderly patients with dementia-
ity of patients can also show clinical benefit without toxic- related psychosis treated with atypical antipsychotic drugs
ity. Indeed, in secondary analyses of the CATIE-AD trial, are at an increased risk of death compared with placebo. The
patients receiving risperidone demonstrated improvement analyses of these trials revealed a risk of death in the drug-
in a variety of behavioural ratings compared to placebo at treated patients of between 1.6 and 1.7 times that seen in
the time of discontinuation in phase 1 while patients tak- of the placebo-treated group. Although the causes of death
ing olanzapine had improvements on measures of hostil- were varied, most of the deaths appeared to be either cardio-
ity and suspiciousness, but demonstrated a worsening of vascular (e.g. heart failure, sudden death) or infectious (e.g.
depressive symptoms (Sultzer et al., 2008). pneumonia) in nature.
These results are consistent with a 2006 Cochrane Subsequent studies, mainly involving large public data-
Collaboration review of placebo-controlled trials, which bases, have found similar or even higher death rates among
concluded that risperidone and olanzapine may improve elders receiving conventional antipsychotics, which has
aggression compared with placebo and that risperidone resulted in a second broader black box warning for all
may improve psychosis relative to placebo (Ballard and antipsychotics in 2008. Several studies aimed at assessing
Waite, 2006). The lower levels of agitation, hostility and mortality risk in patients receiving atypical antipsychot-
psychotic distortions in patients treated with second-gen- ics (Ballard et al., 2009; Ray et al., 2009) have confirmed
eration antipsychotics in CATIE-AD also may be responsi- an elevated risk. Subsequent studies using case controlled
ble for the significantly lower scores reported by caregivers methodology revealed that mortality ratios varied among
on both the Burden Interview and the Neuropsychiatric individual antipsychotic agents (Rossom et al., 2010; Kales
Inventory (NPI) Caregiver Distress Scale compared with et al., 2012). In a retrospective cohort study, Kales et al.
caregivers of patients receiving placebo (Mohamed et al., reported on risks of mortality in dementia patients 65 and
2012). older starting outpatient treatment with an antipsychotic.
During the past decade, atypical antipsychotics have Among patients taking haloperidol, olanzapine and risperi-
largely replaced typical agents in the treatment of psy- done, those taking haloperidol had the highest mortality
chosis, aggression, and agitation in patients with demen- risk and quetiapine the lowest. The mortality risk was high-
tia because of perceived greater tolerability and lesser est in the first 120 days for all drugs, other than haloperi-
risk for acute EPS and TD compared with typical agents dol where the risk was highest in the first 30 days. Valproic
(Jeste et al., 1999; Stanniland and Taylor, 2000; Ritsner acid and its derivatives, included as a non-antipsychotic
et al., 2004; Gill et al., 2005; Burke and Tariot 2009). On comparison, generally had morality risks higher than que-
the other hand, these drugs have other acute and sub-acute tiapine and similar to risperidone (Kales et al., 2012). A ret-
side effects in elderly persons such as sedation, postural rospective case–control study found numbers need to harm
hypotension and in some cases, falls, especially at higher (NNH) of 27, 40 and 50 for risperidone, olanzapine and qui-
doses. Metabolic abnormalities, which have been a major etiapine, respectively, and corresponding increased mortal-
concern among younger populations on atypical anti- ity risks of 3.7%, 2.5% and 2.0%. When compared directly
psychotics, were also evaluated by the CATIE-AD study with quetiapine, dose-adjusted mortality risk was increased
(Zheng et al., 2009). The authors assessed 186 male and with both risperidone (1.7%; 95% confidence interval [CI],
235 female patients with AD. Women showed significant 0.6%–2.8%; p = .003) and olanzapine (1.5%; 95% CI, 0.02%–
weight gain with olanzapine and quetiapine in particular. 3.0%; p = .047). The authors also reported a doswe–response
This effect appeared non-significant in men. Unfavourable increase in mortality risk for these atypical antipsychotics
changes in high-density lipoprotein (HDL) cholesterol (Maust, 2015).
and girth were also noted with olanzapine. Blood pres- In interpreting these data it may be useful to consider
sure, glucose and triglyceride levels appeared unaffected the perspective of another study that examined the effects
by treatment. In addition, impairment of cognitive test of conventional and atypical antipsychotic use on the time
scores, gait disturbance and anticholinergic effects have to nursing home admission and time to death in outpatients
been reported in some instances. In CATIE-AD, atypical with mild-to-moderate probable AD. This study suggested
Treatments for behavioural and psychological symptoms in Alzheimer’s disease and other dementias 267

that it was the presence of psychiatric symptoms rather than


the use of antipsychotics that elevated the risk of institu- 25.8 BUSPIRONE
tionalization and death (Lopez et al., 2013).
Mounting evidence of possible serious adverse effects There have been no randomized, placebo-­ controlled,
of these medications underscores the need for caution doubleblind studies of this agent. Reduced agitation has
when considering use of atypicals. Careful evaluation of been reported in some open trials that we have summarized
the risks and benefits of treatment is warranted, including previously (Loy et al., 1999; Rosenquist et al., 2000), using
assessment of the level of distress and risk of injury to the doses in the range of 10–60 mg/day. The medication has the
patient as a result of insufficiently treated agitated behav- advantage of generally good tolerability, with the exception
iours. Treatment of patients with dementia should empha- of headache, nervousness and dizziness in a modest per-
size non-pharmacological approaches, including carefully centage of patients. There is no evidence regarding tolerance
investigating for possible delirium and for social or envi- or paradoxical effects in this population.
ronmental factors that lead to interventions that capitalize
on the patient’s residual strengths. The placebo response
rate in clinical trials of antipsychotics in dementia is rela-
tively high (generally 30%–40%), underscoring the notion 25.9 ANTIDEPRESSANTS
that non-specific factors such as enhanced attention can be
therapeutic in this population. However, atypicals remain a We would consider using this class of agents first in patients
valid consideration when other interventions have proven with agitation accompanied by evidence of depressive
insufficient. features as described in the introduction. There is some
evidence linking impulsivity to disordered serotonergic
function (Coccaro, 1996), lending support to the use of
serotonergic agents. They have also probably been more
25.7 BENZODIAZEPINES widely assayed because of their good tolerability profile and
widespread use in other psychiatric disorders (Table 25.5).
According to the ‘metaphor’ approach adopted in this Trazodone was one of the first studied (Table 25.5).
chapter, we would, in theory, consider these agents first for There has been only one published double-blind, placebo-­
patients with prominent anxiety. Since features of anxi- controlled study, showing no relative benefit of trazo-
ety and depression overlap extensively, and particularly done over haloperidol or placebo in outpatients with
in patients with dementia, many physicians opt to use an dementia and agitation (Teri et al., 2000). The trial also
antidepressant rather than a benzodiazepine if medica- included a non-blinded behaviour management arm; this
tion is needed. Benzodiazepines have been used for agita- also showed no relative benefit. Among the non-placebo–­
tion associated with dementia in several studies that have controlled studies, doses typically reported ranged from
been reviewed previously (Patel and Tariot, 1991; Loy et al., 50 to 400 mg/day, occasionally higher. Irritability, anxi-
1999). Most of these showed, on average, a reduced level ety, restlessness and depressed affect have been reported to
of agitation with short-term therapy. There have been very improve, along with sleep disturbance. Reported side effects
few studies in the past 10 years, none of which was placebo- have included sedation, orthostatic hypotension and delir-
controlled. In the aggregate, as reviewed in the previous ium. The most rigorous among these compared trazodone
version of this chapter, these studies suggest that agitation (mean dose about 220 mg/day) with haloperidol (mean
associated with anxiety, sleep problems and motor tension dose 2.5 mg/day) (Sultzer et al., 1997). Agitation improved
might be most likely to respond. Some of the earlier stud- equally in both patient groups, although tolerability was
ies in which benzodiazepines were compared with tradi- reported to be better in the trazodone group. The Expert
tional antipsychotics suggested that antipsychotics might Consensus Guideline (Alexopoulos et al., 2005) favours use
be superior. of trazodone primarily to treat sleep disturbance, relegating
Prior reviews indicate that side effects occur com- it to second-or-third-line use for ‘mild’ agitation. A typical
monly with benzodiazepines, including sedation, ataxia, starting dose would be 25 mg/day, with maximum doses
falls, confusion, anterograde amnesia, light-headedness, usually of 100–250 mg/day.
paradoxical agitation as well as tolerance and withdrawal Data regarding selective serotonin reuptake inhibitors
syndromes (Patel and Tariot, 1991). It is because of this (SSRIs) are summarized in Table 25.5. Nyth and Gottfries
safety profile that we reserve the use of benzodiazepines (1990) performed a review of controlled studies of cita-
for situational disturbances, such as dental procedures, lopram, which suggest some beneficial behavioural effects.
for as-needed use, or situations where routine use has Results from two open trials support this conclusion
been attempted and found to be convincingly safe. Drugs (Ragneskog et al., 1996; Pollock et al., 1997).
such as lorazepam and oxazepam are preferred because A double-blind, placebo-controlled study compared
they are metabolized via hepatic conjugation, which is citalopram and perphenazine with placebo in patients with
minimally affected by age and both have relatively short various dementia diagnoses, probable AD and demen-
half-lives. tia with Lewy bodies being the more common diagnoses,
268 Dementia

Table 25.5 Antidepressants – selective serotonin reuptake inhibitors


Dose (mg/day),
Study Diagnosis N Design duration Response Adverse effects
Citalopram
Nyth and AD, vascular 98 DB, PC 10–30, 4 weeks Decreased emotional Confusion (2), dizziness
Gottfries blunting, confusion, (1), sedation (1)
(1990) irritability, anxiety,
mood, restlessness
Nyth et al. Dementia (29) 149 DB, PC 10–30, 6 weeks Decreased anxiety, Tiredness (18), problem
(1992) fear-panic, concentrating (9),
depressed mood apathy (8), dizziness
in all (7), sedation (11),
tension (11)
Ragneskog AD (55), vascular 123 Open, two 20–40, 1–12 Decreased CGI or Tiredness (15);
et al. dementia (30), centres months GBS for irritability, dizziness (14);
(1996) alcoholic depression (60%), drowsiness, sleep
dementia (5), restlessness, disturbances,
NOS (3) anxiety restlessness (4),
aggression (3),
anxiety (2)
Pollock et al. AD 15 Open 20, 2 weeks Decreased agitation, Drop-outs from nausea
(1997) hostility, delusions, (1), myoclonus (1)
disinhibition
Pollock et al. Inpatient AD, 85 Randomized 20 citalopram or Significantly Similar scores among
(2002) vascular DB, PC, 6.5 mean improved all groups
dementia, parallel perphenazine, agitation for
mixed or other group up to 17 days citalopram vs.
not specified placebo
Pollock et al. Inpatient 103 Randomized 20–40 mg Decreased agitation Side effect burden was
(2007) vascular DB, PC citalopram or and psychosis in higher in risperidone
dementia, 1–2 mg both risperidone group
DLB, AD, risperidone, and citalopram
mixed 12 weeks group
Siddique AD 421 Retrospective 36 weeks Reduction of No difference in
­et al. analysis of irritability treatment vs.
(2009) CATIE-AD placebo group
Porsteinsson AD 186 Randomized 9 weeks Reduced agitation Worsening of
et al. DB, PC, and caregiver cognition,
(2014) parallel distress prolongation of QT
group interval
Fluoxetine
Auchus and AD 15 DB, PC 20, 6 weeks Neither more Anxiety, nervousness,
Bissey- parallel effective than confusion, tremor
Black, group vs. placebo for
(1997) haloperidol agitation
SertraLine
Burke et al. AD, NOS 19 Chart review 50, variable Improved on None reported
(1997) dementia duration Clinician’s Global
Impression of
Change (10/11
AD; 4/8 NOS)
Paroxetine
Ramadan AD or VD 15 Open 10–40, 3 months Verbal agitation Weight gain (5),
et al. reduced diarrhoea (1),
(2000) worsened EPS (1)
Treatments for behavioural and psychological symptoms in Alzheimer’s disease and other dementias 269

who had at least one symptom of psychosis or behavioural Concerns about risks of citalopram use were later revised in
disturbance. Eighty-five hospitalized patients were treated 2012 by the FDA to include a recommendation that dosages
after 3 days of dose escalation with doses of 20 mg/day cita- >20 mg/day should not be used in adults over age 60. They
lopram or 0.1 mg/kg/day of perphenazine for up to 14 days. also noted that the dose-dependent QT interval prolonga-
Both ­citalopram and perphenazine demonstrated signifi- tion seen with citalopram is approximately twice that seen
cant improvement compared with placebo for agitation, with escitalopram, the s-isomer of citalopram. In contrast,
lability and psychosis. Side effects were similar in the three a recently published large cohort study found no elevated
treatment groups (Pollock et al., 2002). risks of ventricular arrhythmia or all-cause, cardiac or
Similarly, a 12-week randomized, controlled trial of non-cardiac mortality associated with citalopram dosages
non-depressed patients with dementia hospitalized due >40 mg/day (Zivin et al., 2013). Higher dosages were associ-
to behavioural disturbances evaluated the efficacy of cita- ated with fewer adverse outcomes, and similar findings were
lopram versus risperidone for the treatment of behav- observed for a comparison medication, sertraline, not sub-
ioural disturbances and psychotic symptoms associated ject to the FDA warning.
with dementia (Pollock et al., 2007). The study concluded It has been suggested that the potential beneficial effects
that agitation and psychotic symptoms improved in both of citalopram, and conceivably other serotonergic agents,
groups, but did not differ significantly between the groups. may extend beyond its effect on aberrant behaviours.
However, the lack of a placebo comparator limits interpre- Serotonin signalling suppresses generation of amyloid-β
tation of the clinical significance of these findings. The (Aβ) in vitro and in animal models of AD. A small study
side effect burden was significantly increased in the ris- of healthy adult volunteers receiving citalopram versus pla-
peridone group. cebo demonstrated that Aβ production in cerebrospinal
Data from the CATIE-AD trial addressed the effec- fluid (CSF) was slowed by 37% in the citalopram group com-
tiveness of citalopram versus atypical antipsychotics and pared to placebo. This change was associated with a 38%
revealed no differences in time to all-cause discontinuation decrease in total CSF Aβ concentrations in the drug-treated
for any antipsychotic versus citalopram (Schneider et al., group (Sheline et al., 2014).
2009). Times to discontinuation due to lack of efficacy, or No benefit of administration has been reported yet for
intolerability also did not differ by treatment group. Overall fluoxetine or fluvoxamine, and only anecdotes have been
patients showed generally mild improvement on rating published regarding other serotonergic agents (e.g. sertra-
scales during treatment but changes did not differ signifi- line), all suggesting possible benefit at least in individual
cantly between antipsychotics and citalopram. patients. Overall, evidence for efficacy of serotonergic anti-
A retrospective review of data from the 36 CATIE-AD depressants is mounting.
trial in which patients with AD were treated in a natu- Gastrointestinal distress, loss of appetite and weight,
ralistic manner with placebo, citalopram, risperidone, sedation, insomnia, sexual dysfunction and occasional
olanzapine or quetiapine, comparing scores on the irrita- paradoxical agitation can occur with serotonergic agents,
bility, apathy, delusions and hallucinations subscales of the although, as a group, they tend to be well tolerated. Because
Neuropsychiatric Inventory suggested that the use of citalo- of the suggestion of benefit and the relatively benign side
pram was associated with greatly reduced irritability with- effect profile, consensus statements endorse their use in
out sedation (Siddique et al., 2009). some cases (Alexopolous et al., 2005).
The Citalopram for Agitation in AD Study was a multi-
centre, randomized, placebo-controlled, double-blind and
parallel group trial that enrolled 186 patients with prob-
able AD and clinically significant agitation. A significant
25.10 ANTICONVULSANTS
improvement on both primary outcomes measures was
experienced by patients with AD over 9 weeks of treatment Historically, we might have considered use of anticon-
with citalopram titrated to 30 mg daily. The size of effect vulsants first in patients with features of mania, as well
was comparable to that seen with antipsychotics. There perhaps in those with prominent impulsivity, lability
were insufficient data on efficacy for agitation at lower doses or episodic severe aggression. Evidence regarding these
(Porsteinsson et al., 2014). However, participants receiving agents was summarized in earlier versions of this book:
citalopram showed mild cognitive decline and QT interval we will review it briefly here. Carbamazepine at doses of
prolongation. about 300 mg/day showed benefit in some, but not all,
This reported QT prolongation with citalopram was small controlled trials for treatment of agitation (Tariot
relevant given the 2011 U.S. FDA drug safety communi- et al., 1994; Tariot et al., 1998; Olin et al., 2001). Although
cation regarding abnormal heart rhythms associated with tolerability was acceptable in these trials, side effects seen
high dosages of citalopram (FDA, 2012). This FDA warn- in other populations, such as rashes, sedation, haema-
ing informed health care professionals that citalopram tologic abnormalities, hepatic dysfunction and altered
should no longer be prescribed at dosages above 40 mg/day electrolytes, would be more likely to be evident with wide-
because of potential negative cardiac outcomes, specifically spread use of carbamazepine in the elderly (Tariot et al.,
QT interval prolongation and risk of torsade de pointes. 1995). Further, it has considerable potential for significant
270 Dementia

drug–drug interactions. Our view is that it has a limited significant effect of drug on agitation. Sedation occurred in
role in this population, but the findings suggested that it 36% of the drug group versus 20% of the placebo group, and
might be beneficial to examine other, potentially safer, mild thrombocytopaenia occurred in 7% of the drug group
anticonvulsants (Tables 25.6 and 25.7). and none of the placebo treated patients.
Valproic acid, also available as the enteric-coated deriva- A later randomized, double-blind, placebo-controlled
tive, divalproex sodium, is approved by the U.S. FDA for 6-week trial in 153 nursing home patients with AD com-
treatment of acute mania associated with bipolar disorder. plicated by agitation was conducted by the AD Cooperative
Preliminary studies suggested that valproate might be effec- study (ADCS) (Tariot et al., 2005). This trial showed simi-
tive and safe for the treatment of agitation associated with lar safety and tolerability findings as the previous valproate
dementia. The first randomized, placebo-controlled, parallel studies, but failed to show benefit over placebo in the treat-
group study of this agent was 6 weeks in duration, and sug- ment of agitation associated with dementia. This study was
gested but did not prove benefit (Porsteinsson et al., 2001) at the largest to address agitation prospectively as the primary
a mean divalproex sodium dose of 826 mg/day (mean level outcome.
46 mcg/day. Side effects occurred more frequently in the The ADCS also conducted a placebo-controlled trial of
drug group, chiefly consisting of sedation, gastrointestinal low doses (about 10 mg/kg/day) divalproex sodium in 313
distress, ataxia and an expected decrease in average platelet people with AD not yet complicated by agitation or psycho-
count (~20,000/mm3). sis (Tariot et al., 2011). The treatment period was 2 years,
A randomized, double-blind, placebo-controlled, cross- with a 2-month placebo washout at the end of double-
over study of valproate 480 mg/day for 3 weeks was con- blind treatment. The primary outcome was survival until
ducted in patients with dementia and aggressive behaviour incident agitation or psychosis; key secondary measures
(Sival et al., 2002). No significant differences in aggres- addressed measures of cognitive, functional and global
sive behaviour in this short-term trial were seen between progression of dementia. Valproate treatment did not
placebo and active treatments according to the primary delay emergence of agitation or psychosis or slow cognitive
outcome measure; there was a trend to improvement in or functional decline in patients with moderate AD and
measures of aggression. There were significant effects on was associated with significant toxic effects. The valproate
restless, melancholic and anxious behaviours. There were group had higher rates of somnolence, gait disturbance,
no drug–placebo differences in rate or type of adverse tremor, diarrhoea and weakness. Eighty-eight participants
events. underwent magnetic resonance imaging scans at baseline
A multicentre, randomized, placebo-controlled, 6-week, and 12 months; the valproate group showed greater loss
study of divalproex sodium was conducted in 172 nursing in hippocampal and whole-brain volume, accompanied by
home residents with dementia and agitation who also met greater ventricular expansion.
criteria for secondary mania. A target dose of 20 mg/kg/day There are no controlled studies of which we are aware
was reached in 10 days (Tariot et al., 2001a). This titra- of the newer anticonvulsants, including lamotrigine and
tion rate and dose resulted in sedation in about 20% of gabapentin. There are a few case reports and case series
the drug-treated group and a relatively high dropout rate, suggest benefit with gabapentin (Regan and Gordon, 1997;
leading to premature discontinuation of the study (n = Herrmann et al., 2000; Roane et al., 2000; Megna et al.,
100 ­completers). There were no significant drug–placebo 2002; Hawkins et al., 2000). A small double-blind random-
differences in change in manic features, but there was a ized trial comparing topiramate and risperidone suggested

Table 25.6 Anticonvulsants – carbamazepine


Dose (mg/day),
Study Diagnosis N Design duration Response Adverse effects
Tariot et al. AD, vascular 25 Crossover, PC 300 modal, Decreased Tics (1), sedation
(1994) dementia (some other 5 weeks agitation
psychotropics (16 vs.
permitted) 4 placebo)
Tariot et al. Dementia 51 Randomized PC (no 300 modal, Improved on Ataxia (9 vs.
(1998) psychotropics 6 weeks CGI (20/26); 3 placebo),
except chloral also improved dis­orientation
hydrate as needed) on OAS, (4 vs. 0 placebo),
BPRS, BRSD tics (1), sedation
Olin et al. AD 21 Randomized, DB, PC 388, 6 weeks Improved BPRS Diarrhoea more
(2001) hostility item common with
active treatment
Treatments for behavioural and psychological symptoms in Alzheimer’s disease and other dementias 271

Table 25.7 Anticonvulsants – valproate


Dose (mg/day),
plasma level
(μg/mL),
Study Diagnosis N Design duration Response Adverse effects
Herrmann AD, LB, 16 Open 750–2500 Improved on Sedation, ataxia,
(1998a) vascular (mean = 1331), CMAI, diarrhoea (1)
dementia (mean level = BEHAVE-AD
459 μM/L), (11)
5-34 weeks
Kunik et al. Dementia 13 Retrospective review 500–1750 (mean Decreased None reported;
(1998) (concurrent BDZs, = 846), (mean physical not drug interaction
antipsychotics) level = 48), verbal agitation with phenytoin
duration and aggression noted (1)
variable on CMAI (10)
Porsteinsson AD, vascular 56 Randomized PC (no 375–1375 (mean Decreased Sedation and
et al. dementia, psychotropics = 826), (mean agitation by nausea, vomiting
(2001) mixed except chloral level = 45), BPRS and CGI or diarrhoea
dementia hydrate as needed) 6 weeks
Tariot et al. AD and/or 172 Randomized, DB, PC, 1000 median Significant Somnolence,
(2001a) VD with parallel-group improvement thrombocytopenia
secondary on CMAI
mania verbal agitation
subscale
Sival et al. AD, vascular 42 Randomized, DB, PC, 480 (mean level No change in Small increase in
(2002) dementia, crossover = 41), 3 weeks aggressive incidence of
other and active behavior adverse events
mixed treatment compared to compared to
dementia placebo placebo
Porsteinsson AD, vascular 46 Open extension 250–1500 (mean Decreased Fall, sedation and
et al. dementia, = 851), (mean agitation by nausea, vomiting
(2003) mixed level = 47), BPRS and CGI or diarrhoea
dementia 6 weeks

Abbreviations used in all tables


AD Alzheimer’s disease MOSES Multidimensional Observation Scale for
BEHAVE- Behavioural Pathology in Alzheimer’s Disease Elderly Subjects
AD Rating Scale NOS Dementia not otherwise specified
BPRS Brief Psychiatric Rating Scale NPI Neuropsychiatric Inventory
BRSD CERAD Behavioural Rating Scale for Dementia NPI-NH Neuropsychiatric Inventory – Nursing Home
CATIE-AD Clinical Antipsychotic Trial of Intervention Version
Effectiveness – Alzheimer’s Disease PC Placebo controlled
CGI Clinical Global Impression PD Parkinson’s disease
CMAI Cohen-Mansfield Agitation Inventory PRN pro re nata (as needed)
DB Double blind SCAG Sandoz Clinical Assessment-Geriatric
EPS Extrapyramidal symptoms Scale
GBS Gottfries–Bråne–Steen SSRIs Selective serotonin reuptake inhibitors
MC Multicentre VD Vascular dementia

comparable efficacy of topiramate (25–50 mg daily) with against using such agents. Nevertheless, some consensus
risperidone in controlling behavioural disturbances in statements suggest a possible role as a second or third line
patients with AD (Mowla and Pani, 2010). treatment for agitation and aggressive behaviours in patients
Evidence regarding the efficacy of anticonvulsants in with dementia. We would only consider this approach if
the treatment of agitation associated with dementia argues other treatments had failed.
272 Dementia

Citing preliminary studies which were conducted in


α AND β-ADRENERGIC AGENTS
25.11  patients displaying baseline low levels of psychopathol-
ogy associated with dementia, Cummings et al. (2006)
β-Blocking agents have not been subject to rigorous study. conducted an exploratory analysis of data pertaining to
Most of the evidence comes from open trials conducted the efficacy of donepezil treatment of patients with severe
over 10 years ago (Rosenquist et al., 2000). The usefulness behavioural disturbances. The results of these analyses sug-
of β-blocking agents in this very old and frail population gest that donepezil reduces behavioural symptoms, particu-
is limited by the high frequency of relative contraindica- larly mood disturbances and delusions, in patients with AD
tions to β-blocker treatment (Peskind et al., 2005). Adverse with relatively severe psychopathology.
reactions include bradycardia, hypotension, potential A 12-week placebo-controlled, randomized trial examined
worsening of congestive heart failure or asthma, reduced the effectiveness of donepezil for the treatment of clinically
adrenergic response to hypoglycaemia in patients with significant agitation in patients with AD revealed no signifi-
diabetes, sedation, increased confusion, psychosis, depres- cant treatment advantage for donepezil as compared with
sion and increased atrioventricular block. It is unclear placebo. Howard et al. hypothesized that the previously noted
what role this class of medication has in the treatment of modest but significant improvement in the overall severity of
agitation associated with dementia at this point. If it is to behavioural and psychological symptoms of dementia (BPSD)
be attempted at all, we would argue that it should occur in previous trials may be explained by improvements in symp-
only after safer and more conventional therapies have toms other than agitation (Howard et al., 2007). For example,
failed, and only with careful monitoring for side effects. a study that examined individual items on the NPI in patients
Prazosin, a non-sedating medication used for hyperten- with AD showed significant benefits for mood-related symp-
sion and benign prostatic hypertrophy, antagonizes norad- toms but not agitation (Gauthier et al., 2002).
renergic effects at brain post-synaptic α-1 adrenoreceptors. A recent 12-week, randomized, parallel-group, ­controlled,
A small, double-blind, placebo-controlled, parallel group single-centre trial of risperidone versus galantamine
study examined the efficacy and tolerability of prazosin revealed that both galantamine and risperidone were effec-
for behavioural symptoms in nursing home patients with tive in reducing agitation. Although risperidone showed a
agitation/aggression in AD (Wang et al., 2009). Subjects significant advantage over galantamine in irritation and
received doses between 1 and 6 mg daily using a flexible agitation at week 3 and at week 12, both galantamine and
dose algorithm. Those receiving prazosin displayed a sig- risperidone treatments resulted in improved BPSD symp-
nificant improvement in behavioural symptoms compared toms and were equally effective in treating several NPI
to the placebo group with no significant increase in adverse domains (Freund-Levi et al., 2014).
effects. These encouraging preliminary results require con- By way of overview, there have not been sufficient prospec-
firmation in larger controlled studies. tive controlled trials specifically addressing whether ChEI
therapy definitely mitigates symptoms already present, or
delays emergence of symptoms in those not yet afflicted. In the
aggregate, the data available suggest that these salutary effects
25.12 CHOLINERGIC THERAPIES may exist. Since best practice is to treat most people with AD
with these agents anyway, there seems to be little reason to
As summarized by Cummings (2000), there is evidence that refrain from using them as first line therapy for treatment of
cholinergic agents, especially ChEIs, may have modest, clin- non-emergent psychopathology in people with AD.
ically relevant, psychotropic effects in some patients with
dementia. For the most part, the studies were not designed to
address behavioural outcomes as the primary goal. A review
of 24 trials involving nearly 5800 participants found benefits
25.13 MEMANTINE
of treatment noted on measures of activities of daily living
and behaviour in patients receiving donepezil (Birks, 2006; Memantine is a non-competitive inhibitor of N-methyl-
Birks and Harvey, 2006). The effects of donepezil, galan- d-aspartate (NMDA) receptors that may permit normal
tamine and rivastigmine in people with mild, moderate or memory formation but blocks their excitotoxic activation
severe dementia due to AD were also supported in a review (Winblad et al., 2002). Memantine has been approved for
of 10 randomized, double-blind, placebo-controlled trials treatment of moderate to severe AD, alone or in addi-
(Birks, 2006). The reviewers found no differences in efficacy tion to ChEIs, on the basis of three placebo-controlled tri-
between study medications. A meta-analysis of 29 parallel- als (Winblad and Poritis 1999; Reisberg et al., 2003, Tariot
group or cross-over randomized, placebo-controlled trials et al., 2004). A review of double-blind, parallel group,
of outpatients diagnosed with mild-to-moderate AD meant placebo-controlled, randomized trials of memantine in
­
to quantify the efficacy of ChEIs for neuropsychiatric and people with dementia conducted by McShane et al. (2006)
functional outcomes found that ChEIs had a modest benefi- reported a slight decrease in the development of agitation in
cial impact. No difference in efficacy among various ChEIs patients taking memantine. This effect was slightly larger, but
was observed (Trinh et al., 2003). still small, in moderate-to-severe AD. There was no evidence
Treatments for behavioural and psychological symptoms in Alzheimer’s disease and other dementias 273

about whether memantine had an effect on agitation, which associated with psychotic features. However, these medi-
is already present. A significant advantage for memantine cations are not without risk. Mounting evidence as to the
over placebo was also supported by a pooled analysis of three cerebrovascular adverse effects and other potentially deadly
studies of patients with symptoms of aggression/­agitation, side effects of both conventional and atypical antipsychotics
delusions and hallucinations (Wilcock et al., 2008). has overshadowed evidence of their efficacy. Although they
The first completed trial designed primarily to explore remain an indispensable treatment for patients displaying
the efficacy of memantine on clinically significant agitation severely agitated and psychotic behaviours, this increasing
in AD found no significant advantage of memantine versus safety data emphasizes the need for vigilance when evaluat-
placebo in the primary outcome CMAI at week 6 or 12 (Fox ing appropriate treatment options.
et al., 2012). Modest improvement was noted on the second- Emerging data regarding alternative treatment options for
ary measure NPI. A pooled analysis of 633 patients also agitated behaviours such as antidepressant therapy is promis-
revealed better outcomes in the memantine group in terms ing, but still limited. Other literature regarding non-antipsy-
of behavioural and psychological symptoms, including chotic medication includes anticonvulsants, anxiolytics and a
aggressiveness (Nakamura et al., 2014). Similar improve- variety of other agents with less foundation. As data accrue
ments in NPI scores have been demonstrated with the regarding these therapeutic approaches, we will have a clearer
extended release formulation (Grossberg et al., 2013). sense of exactly how and when they should be deployed. In
Howard et al. (2012) attempted a clinical trial to address the meantime, clinicians are obliged to deal as thoughtfully as
the relative benefit of continuing donepezil, adding or sub- possible with patients on a case-by-case basis, attempting first
stituting memantine or stopping donepezil in persons with and foremost to avoid toxicity of therapies intended to help.
moderate-to-severe dementia. Limited enrolment ham-
­
pered the conclusions that could be drawn (Tariot, 2012). REFERENCES
Participants assigned to donepezil withdrawal showed
marked decline in both cognition and daily function in Alexopoulos, G.S., Jeste, D.V., Chung, H. et al. (2005).
comparison with the other three treatment groups, and The expert consensus guideline series. Treatment
memantine treatment was associated with fewer behav- of dementia and its behavioral disturbances.
ioural symptoms. Introduction: Methods, commentary, and summary.
As with the overview of the behavioural effects of ChEIs, Postgraduate Medicine, Jan;Spec No: 6–22.
the available data suggest but do not prove that memantine Allain, H., Dautzenberg, P.H., Maurer, K. et al. (2000).
therapy may reduce behavioural symptoms or delay their Double blind study of tiapride versus haloperi-
emergence. Since it is indicated for treatment of moderate- dol and placebo in agitation and aggressiveness
to-severe AD, there is little reason not to deploy it in such in elderly patients with cognitive impairment.
patients and observe whether behavioural symptoms are Psychopharmacology (Berl), 148: 361–366.
improved. American Geriatrics Society and American Association
for Geriatric Psychiatry (2003). Consensus state-
ment on improving the quality of mental health care
in U.S. nursing homes: Management of depression
25.14 SUMMARY and behavioral symptoms associated with demen-
tia. Journal of the American Geriatrics Society, 51:
Humane treatment of patients with dementia includes vigor- 1287–1298.
ously addressing the kinds of behavioural and psychologi- APA Work Group on Alzheimer’s Disease and other
cal signs and symptoms that are likely to occur. The main Dementias, Rabins, P.V., Blacker, D., Rovner, B.W.
emphasis should be on non-pharmacological approaches, et al. (2007). American Psychiatric Association:
including carefully investigating for possible delirium and Practice guideline for the treatment of patients with
searching hard for social or environmental interventions that Alzheimer’s disease and other dementias, second edi-
capitalize on the patient’s residual strengths. It is indeed this tion. The American Journal of Psychiatry, 164 (Suppl.
process of attempting to ‘find what works’ that makes treat- 12): 5–56.
ment in this population so fascinating, as well as challenging. Auchus, A.P. and Bissey-Black, C. (1997). Pilot study of
Psychotropics should be reserved for cases where other, haloperidol, fluoxetine, and placebo for agitation in
simpler interventions have been attempted and deemed Alzheimer’s disease. Journal of Neuropsychiatry and
inadequate. We advocate a target symptom-based approach, Clinical Neurosciences, 9: 591–593.
which matches the most salient target symptoms to a rel- Ballard, C., Grace, J., McKeith, I. and Holmes, C. (1998).
evant drug class. Although there is no compelling empirical Neuroleptic sensitivity in dementia with Lewy bodies
support for this approach, it has the advantage of being face- and Alzheimer’s disease. Lancet, 351: 1032–1033.
valid and logical, which are reassuring features when one is Ballard, C., Hanney, M.L., Theodoulou, M. et al. (2009).
a consultant in a confusing situation. The dementia antipsychotic withdrawal trial (DART):
Much of the available data do in fact indicate that Long-term follow-up of randomised placebo con-
antipsychotics as a group can show benefit for agitation trolled trial. The Lancet Neurology, 8 (2): 151–157.
274 Dementia

Ballard, C., Margallo-Lana, M., Juszczak, E. et al. (2005). and severe behavioral disorders. The American Journal
Quetiapine and rivastigmine and cognitive decline of Geriatric Psychiatry, 14 (7): 605–612.
in Alzheimer’s disease: Randomized double blind Cummings, J.L., Schneider, E., Tariot, P.N., Graham, S.M.
placebo controlled trial. BMJ-British Medical Journal, and Memantine MEM-MD-02 Study Group. (2006b).
330: 874–887. Behavioral effects of memantine in Alzheimer disease
Ballard, C. and Waite, J. (2006). The effectiveness of patients receiving donepezil treatment. Neurology, 67:
atypical antipsychotics for the treatment of aggression 57–63.
and psychosis in Alzheimer’s disease. The Cochrane Davis, P. and Baskys, A. (2002). Quetiapine effectively
Database of Systematic Reviews, 1: 1–108. reduces psychotic symptoms in patients with Lewy
Birks, J. (2006). Cholinesterase inhibitors for Alzheimer’s body dementia: An advantage of the unique pharma-
disease. Cochrane Database of Systematic Reviews (1): cological profile? Brain Aging, 2: 49–53.
CD005593. De Deyn, P.P., Eriksson, H., Svensson, H. and Study 115
Birks, J. and Flicker, L. (1998). The efficacy and safety investigators. (2012). Tolerability of extended-release
of selegiline for the symptomatic treatment of quetiapine fumarate compared with immediate-
Alzheimer’s disease: A systematic review of the evi- release quetiapine fumarate in older patients with
dence (Cochrane review). The Cochrane Library, Issue Alzheimer’s disease with symptoms of psychosis
2. Oxford: Update Software. and/or agitation: A randomised, double-blind,
Birks, J. and Harvey, R.J. (2006). Donepezil for dementia ­parallel-group study. International Journal of Geriatric
due to Alzheimer’s disease. Cochrane Database of Psychiatry, 27 (3): 296–304.
Systematic Reviews, 1 (1): CD001190. De Deyn, P.P., Drenth, A.F., Kremer, B.P., Oude Voshaar,
Breder, C., Kostic, D., Forbes, A. et al. (2005). Overall R.C., Van Dam, D. (2013). Aripiprazole in the treatment
safety of aripiprazole in trials of patients with psycho- of Alzheimer’s disease. Expert Opin Pharmacother, 14:
sis of Alzheimer’s dementia (poster). Presented at the 459–474.
158th American Psychiatric Association Meeting, May De Deyn, P.P., Katz, I.R., Brodaty, H., Lyons, B., Greenspan,
21–26, 2005, Atlanta, GA. A. and Burns A. (2005). Management of agitation,
Brodaty, H., Ames, D., Snowdon, J. et al. (2003). A ran- aggression, and psychosis associated with dementia: A
domized placebo-controlled trial of risperidone for the pooled analysis including three randomized, placebo-
treatment of aggression, agitation, and psychosis of controlled double-blind trials in nursing home resi-
dementia. Journal of Clinical Psychiatry, 64: 134–143. dents treated with risperidone. Clinical Neurology and
Burke, W.J., Dewan, V., Wengel, S.P. et al. (1997). The Neurosurgery, 107(6): 497–508.
use of selective serotonin reuptake inhibitors for De Deyn, P.P., Rabheru, K. and Rasmussen, A. (1999). A
depression and psychosis complicating dementia. randomized trial of risperidone, placebo, and haloperi-
International Journal of Geriatric Psychiatry, 12: 519–25. dol for behavioral symptoms of dementia. Neurology,
Burke, A.D. and Tariot, P.N. (2009). Atypical antipsychotics 53: 946–55.
in the elderly: A review of therapeutic trends and clini- Devanand, D.P. (2000). Depression in dementia. In N.
cal outcomes. Expert Opinion on Pharmacotherapy, 10 Qizilbash et al. (ed.), Evidence-Based Dementia:
(15): 2407–2414. A Practical Guide to Diagnosis and Management.
Center for Drug Evaluation and Research (n.d.). Deaths Oxford: Blackwell Science Ltd.
with Antipsychotics in Elderly Patients with Behavioral Devanand, D.P., Marder, K., Michaels, K.S. et al. (1998).
Disturbances. Available at http://www.fda.gov/cder/drug/ A randomized, placebo-controlled, dose-comparison
advisory/antipsychotics.htm. Accessed 10 June 2016. trial of haloperidol for psychosis and disruptive
Christensen, D.B. and Benfield, W.R. (1998). Alprazolam as behaviors in Alzheimer’s disease. American Journal of
an alternative to low-dose haloperidol in older, cogni- Psychiatry, 155: 1512–20.
tively impaired nursing facility patients. Journal of the Devanand, D.P., Mintzer, J., Schultz, S.K. et al. (2012).
American Geriatrics Society, 46: 620–625. Relapse risk after discontinuation of risperidone
Coccaro, E.F. (1996). Neurotransmitter correlates of impul- in Alzheimer’s Disease. New England Journal of
sive aggression in humans. Annals of the New York Medicine, 367: 1497–1507.
Academy of Sciences, 794: 82–89. Doody, R.S., Stevens, J.C., Beck, C. et al. (2001). Practice
Cohen-Mansfield, J. and Billig, N. (1986) Agitated behav- parameter: Management of dementia (an evidence-
iors in the elderly. I. A conceptual review. Journal of based review) report of the quality standards sub-
the American Geriatrics Society, 34: 711–721. committee of the American academy of neurology.
Cummings, J.L. (2000). Cholinesterase inhibitors: A new Neurology, 56: 1154–1166.
class of psychotropic compounds. American Journal of FDA (2012). FDA drug safety communication: Abnormal
Psychiatry, 157: 4–16. heart rhythms associated with high doses of Celexa
Cummings, J.L., McRae, T., Zhang, R. and Donepezil- (citalopram hydrobromide). Silver Spring, MD: US Food
Sertraline Study Group. (2006a). Effects of donepezil on and Drug Administration. http://www.fda.gov/Drugs/
neuropsychiatric symptoms in patients with dementia DrugSafety/ucm269086.htm. Accessed 10 June 2016.
Treatments for behavioural and psychological symptoms in Alzheimer’s disease and other dementias 275

Finkel, S.I., Costa e Silva, J., Cohen, G. et al. (1996). Jeste, D.V., Caligiuri, M.P., Paulsen, J.S. et al. (1995). Risk
Behavioral and psychological signs and symptoms of of tardive dyskinesia in older patients. A prospec-
dementia: A consensus statement on current knowl- tive longitudinal study of 266 outpatients. Archives of
edge and implications for research and treatment. General Psychiatry, 52: 756–765.
International Psychogeriatrics, 8 (Suppl. 3): 497–500. Jeste, D.V. and Finkel, S.I. (2000). Psychosis of Alzheimer’s
Fox, C., Crugel, M., Maidment, I. et al. (2012). Efficacy disease and related dementias. Diagnostic criteria for
of memantine for agitation in Alzheimer’s dementia: a distinct syndrome. The American Journal of Geriatric
A randomised double-blind placebo controlled trial. Psychiatry, 8: 29–34.
PLoS One, 7 (5): e35185. Jeste, D.V., Lacro, J.P., Bailey, A. et al. (1999). Lower inci-
Freund-Levi, Y., Bloniecki, V., Auestad, B. et al. (2014). dence of tardive dyskinesia with risperidone compared
Galantamine versus risperidone for agitation in with haloperidol in older patients. Journal of the
people with dementia: A randomized, twelve-week, American Geriatrics Society 47: 716–719.
single-center study. Dementia and Geriatric Cognitive Kales, H.C., Kim, H.M., Zivin, K. et al. (2012). Risk of mortal-
Disorders, 38 (3–4): 234–244. ity among individual antipsychotics in patients with
Gauthier, S., Feldman, H., Hecker, J. et al. (2002). Efficacy dementia. American Journal of Psychiatry, 169 (1): 71–79.
of donepezil on behavioural symptoms in patients with Katz, I., Jeste, D.V., Mintzer, J.E. et al. (1999). Comparison
moderate to severe Alzheimer’s disease. International of risperidone and placebo for psychosis and behav-
Psychogeriatrics, 14: 389–404. ioral disturbances associated with dementia: A
Gill, S.S., Rochon, P.A., Herrmann, N. et al. (2005). Atypical randomized, double-blind trial. Journal of Clinical
antipsychotic drugs and risk of ischaemic stroke: Psychiatry, 60: 107–115.
Population based retrospective cohort study. BMJ- Kunik, M.E., Puryear, L., Orengo, C.A. et al. (1998). The
British Medical Journal, 330 (7489): 445. efficacy and tolerability of divalproex sodium in elderly
Grossberg, G.T., Manes, F., Allegri, R.F. et al. (2013) demented patients with behavioural disturbances.
The safety, tolerability, and efficacy of once-daily International Journal of Geriatric Psychiatry, 13: 29–34.
memantine (28 mg): A multinational, randomized, Lanctôt, K.L., Best, T.S., Mittmann, N. et al. (1998).
double-blind, placebo-controlled trial in patients with Efficacy and safety of neuroleptics in behavioral dis-
moderate-to-severe Alzheimer’s disease taking cholin- orders associated with dementia. Journal of Clinical
esterase inhibitors. CNS Drugs, 27 (6): 469–478. Psychiatry, 59: 550–561.
Hawkins, J.W., Tinklenberg, J.R., Sheikh, J.I. et al. (2000). Lavretsky, H. and Sultzer, D. (1998). A structured trial of
A retrospective chart review of gabapentin for the risperidone for the treatment of agitation in dementia.
treatment of aggressive and agitated behavior in American Journal of Geriatric Psychiatry, 6: 127–135.
patients with dementias. The American Journal of Leibovici, A. and Tariot, P.N. (1988) Agitation associated
Geriatric Psychiatry, 8: 221–225. with dementia: A systematic approach to treatment.
Herrmann, N. (1998a). Valproic acid treatment of agitation Psychopharmacology Bulletin 24: 49–53.
in dementia. Canadian Journal of Psychiatry, 43: 69–72. Lonergan, E., Luxenberg, J. and Colford, J. (2002).
Herrmann, N., Lanctot, K. and Myszak, M. (2000). Haloperidol for agitation in dementia. The Cochrane
Effectiveness of gabapentin for the treatment of Database Systematic Reviews, 2 (2): CD002852.
behavioral disorders in dementia. Journal of Clinical Loy, R., Tariot, P.N., Rosenquist, K. (1999). Alzheimer’s disease:
Psychopharmacology, 20: 90–93. Behavioral management. In I.R. Katz, Oslin, D. and
Herrmann, N., Rivard, M.F., Flynn, M. et al. (1998b). Lawton, M.P. (eds.) Annual Review of Gerontology and
Risperidone for the treatment of behavioral dis- Geriatrics: Focus on Psychopharmacologic Interventions
turbances in dementia: A case series. Journal of in Late Life. New York, NY: Springer, pp. 136–194.
Neuropsychiatry and Clinical Neurosciences, 10: Lopez, O.L., Becker, J.T., Chang, Y.F. et al. (2013). The
220–223. long-term effects of conventional and atypical antipsy-
Howard, R.J., Juszczak, E., Ballard, C.G. et al.; CALM-AD chotics in patients with probable Alzheimer’s disease.
Trial Group. (2007). Donepezil for the treatment of The American Journal of Psychiatry, 170 (9): 1051–1058.
agitation in Alzheimer’s disease. New England Journal Lyketsos, C.G., Sheppard, J.M. and Steele, C.D. (2000).
of Medicine, 357 (14): 1382–1392. Randomized, placebo-controlled, double-blind clini-
Howard, R., McShane, R., Lindesay, J. et al. (2012). cal trial of sertraline in the treatment of depression
Donepezil and memantine for moderate-to-severe complicating Alzheimer’s disease: Initial results from
Alzheimer’s disease. New England Journal of the Depression in Alzheimer’s Disease study. The
Medicine, 366: 893–903. American Journal of Psychiatry, 157: 1686–1689.
Huybrechts, K.F., Gerhard, T., Crystal, S. et al. (2012). Maglione, M., Maher, A.R., Hu, J. et al. (2011). Off-label
Differential risk of death in older residents in nurs- use of atypical antipsychotics: An update [Internet].
ing homes prescribed specific antipsychotic drugs: Rockville, MD: Agency for Healthcare Research
Population based cohort study. BMJ-British Medical and Quality (US). Report No: 11-EHC087-EF. AHRQ
Journal, 344: e977. Comparative Effectiveness Reviews.
276 Dementia

Maust, D.T., Kales. H.C. and Blow, F.C. (2015). Mental Olin, J.T., Fox, L.S., Pawluczyk, S. et al. (2001). A pilot
health care delivered to younger and older adults randomized trial of carbamazepine for behavioral
by office-based physicians nationally. Journal of the symptoms in treatment-resistant outpatients with
American Geriatrics Society, 63: 1364–1372. Alzheimer’s disease. American Journal of Geriatric
McShane, R., Areosa Sastre, A. and Minakaran, N. Psychiatry, 9: 400–405.
(2006). Memantine for dementia. Cochrane Database Patel, S. and Tariot, P.N. (1991). Pharmacologic models
Systematic Reviews, 19 (2): CD003154. of Alzheimer’s disease. Psychiatric Clinics of North
Meehan, K.M., Wang, H., David, S.R. et al. (2002). America, 14: 287–308.
Comparison of rapidly acting intramuscular olanzap- Peskind, E.R., Tsuang, D.W., Bonner, L.T. et al. (2005).
ine, lorazepam, and placebo: A double-blind, random- Propranolol for disruptive behaviors in nursing home
ized study in acutely agitated patients with dementia. residents with probable or possible Alzheimer Disease:
Neuropsychopharmacology, 26: 494–504. A placebo-controlled study. Alzheimer Disease and
Megna, J.L., Devitt, P.J., Sauro, M.D. and Dewan, M.J. Associated Disorders, 19 (1): 23–28.
(2002). Gabapentin’s effect on agitation in severely Pollock, B.G., Mulsant, B.H., Rosen, J. et al. (2002).
and persistently mentally ill patients. Annals of Comparison of citalopram, perphenazine, and placebo
Pharmacotherapy, 36: 12–16. for the acute treatment of psychosis and behavioral
Merriam, A.E., Aronson, M.K., Gaston, P. et al. (1988). The disturbances in hospitalized, demented patients.
psychiatric symptoms of Alzheimer’s disease. Journal American Journal of Psychiatry, 159: 460–465.
of the American Geriatrics Society, 36: 7–12. Pollock, B.G., Mulsant, B.H., Rosen, J. et al. (2007). A
Mintzer, J.E., Tune, L.E., Breder, C.D. et al. (2007). double-blind comparison of citalopram and risperi-
Aripiprazole for the treatment of psychoses in done for the treatment of behavioral and psychotic
institutionalized patients with Alzheimer dementia: symptoms associated with dementia. American Journal
A multicenter, randomized, double-blind, placebo- of Geriatric Psychiatry, 15 (11): 942–952.
controlled assessment of three fixed doses. The Pollock, B.G., Mulsant, B.H., Sweet, R. et al. (1997). An
American Journal of Geriatric Psychiatry, 15 (11): open pilot study of citalopram for behavioral dis-
918–931. turbances of dementia. Plasma levels and real-time
Mintzer, J., Weiner, M., Greenspan, A. et al. (2004). observations. American Journal of Geriatric Psychiatry,
Efficacy and safety of a flexible dose of ris- 5: 70–78.
peridone versus placebo in the treatment of Porsteinsson, A.P., Drye, L.T., Pollock, B.G. et al.; CitAD
psychosis of Alzheimer’s disease. Poster pre- Research Group. (2014). Effect of citalopram on agita-
sented at the International College of Geriatric tion in Alzheimer disease: The CitAD randomized clini-
Psychopharmacology; Oct 14–17, 2004; Basel, cal trial. JAMA, 311 (7): 682–691.
Switzerland. Profenno, L.A., Jakimovich, L., Holt, C.J., Porsteinsson, A.
Mohamed, S., Rosenheck, R., Lyketsos, C.G. et al. (2012). and Tariot, P.N. (2005). A randomized, double-blind,
Effect of second-generation antipsychotics on care- placebo-controlled pilot trial of safety and tolerabil-
giver burden in Alzheimer’s disease. Journal of Clinical ity of two doses of divalproex sodium in outpatients
Psychiatry, 73 (1): 121–128. with probable Alzheimer’s disease. Current Alzheimer
Mowla, A. and Pani, A. (2010). Comparison of topiramate Research, 2: 553–558.
and risperidone for the treatment of behavioral distur- Profenno, L.A. and Tariot, P.N. (2004). Pharmacologic
bances of patients with Alzheimer disease: A double- management of agitation in Alzheimer’s disease.
blind, randomized clinical trial. Journal of Clinical Dementia Geriatric Cognitive Disorders, 17: 65–77.
Psychopharmacology, 30 (1): 40–43. Porsteinsson, A.P., Tariot, P.N., Erb, R. et al. (2001).
Nakamura, Y., Kitamura, S., Homma, A. et al. (2014). Placebo-controlled study of divalproex sodium for
Efficacy and safety of memantine in patients with agitation in dementia. American Journal of Geriatric
moderate-to-severe Alzheimer’s disease: Results of Psychiatry, 9: 58–66.
a pooled analysis of two randomized, double-blind, Porsteinsson, A.P., Tariot, P.N. and Jakimovich, L.J. (2003).
placebo-controlled trials in Japan. Expert Opinion on Valproate therapy for agitation in dementia: Open-
Pharmacotherapy, 15 (7): 913–925. label extension of a double-blind trial. American
Nyth, A.L. and Gottfries, C.G. (1990) The clinical efficacy Journal of Geriatric Psychiatry, 11: 434–440.
of citalopram in treatment of emotional disturbances Rabins, P.V., Blacker, D. and Rovner, B.W. (2007).
in dementia disorders. A Nordic multicentre study. Practice guideline for the treatment of patients
British Journal of Psychiatry, 157: 894–901. with Alzheimer’s disease and other dementias. The
Nyth, A.L., Gottfries, C.G., Lyby, K. et al. (1992). A con- American Journal of Psychiatry, 164 (Suppl. 12): 5–56.
trolled multicenter clinical study of citalopram and pla- Rabins, P.V., Rovner, B.W., Rummans, T. et al. (2014).
cebo in elderly depressed patients with and without Practice Guideline for the Treatment of Patients with
concomitant dementia. Acta Psychiatrica Scandinavica, Alzheimer’s Disease and Other Dementias. Arlington,
86: 138–145. VA: American Psychiatric Association. Available at
Treatments for behavioural and psychological symptoms in Alzheimer’s disease and other dementias 277

http://psychiatryonline.org/pb/assets/raw/sitewide/ Schneider, L.S., Tariot, P.N., Dagerman, K.S. et al.


practice_guidelines/guidelines/alzheimerwatch.pdf. (2006b). Effectiveness of atypical antipsychotic drugs
Accessed 10 June 2016. in patients with Alzheimer’s disease. New England
Ragneskog, H., Eriksson, S., Karlsson, I. and Gottfries, Journal of Medicine, 355: 1525–1538.
C.G. (1996). Long-term treatment of elderly individu- Schneider, L.S., Tariot, P.N., Lyketsos, C.G. et al. (2001).
als with emotional disturbances: An open study with National institute of mental health clinical antipsy-
citalopram. International Psychogeriatrics, 8: 659–668. chotic trials of intervention effectiveness (CATIE):
Ramadan, F.H., Naughton, B.J. and Bassanelli, A.G. (2000). Alzheimer disease trial methodology. American
Treatment of verbal agitation with a selective sero- Journal of Geriatric Psychiatry, 9: 346–360.
tonin reuptake inhibitor. Journal of Geriatric Psychiatry Schneider, L.S., Vigen, C., Mack, W.J. et al. (2009).
Neurology, 13: 56–59. Effectiveness of citalopram compared to atypical
Ray, W.A., Chung, C., Murray, K. et al. (2009). Atypical anti- antipsychotics in AD patients who discontinued their
psychotic drugs and the risk of sudden cardiac death. initially assigned treatment: CATIE-AD phase 2 results.
New England Journal of Medicine, 360: 225–235. American Journal of Geriatric Psychiatry, 17 (3): A77.
Regan, W.M. and Gordon, S.M. (1997). Gabapentin for Sheline, Y.I., West, T., Yarasheski, K. et al. (2014). An
behavioral agitation in Alzheimer’s disease. Journal of antidepressant decreases CSF Aβ production in
Clinical Psychopharmacology, 17: 59–60. healthy individuals and in transgenic AD mice. Science
Reisberg, B., Doody, R., Stöffler, A. et al.; for the Translational Medicine, 6 (236): 236re4.
Memantine Study Group. (2003). Memantine in Siddique, H., Hynan, L.S. and Weiner, M.F. (2009). Effect
moderate-to-severe Alzheimer’s disease. New England of a serotonin reuptake inhibitor on irritability, apathy
Journal of Medicine, 348: 1333–1341. and psychotic symptoms in patients with Alzheimer’s
Ritsner, M., Gibel, A., Perelroyzen, G. et al. (2004). Quality disease. Journal of Clinical Psychiatry, 70 (6): 915–918.
of life outcomes of risperidone, olanzapine, and typical Sival, R.C., Haffmans, P.M., Jansen, P.A. et al. (2002).
antipsychotics among schizophrenia patients treated Sodium valproate in the treatment of aggressive
in routine clinical practice: A naturalistic comparative behavior in patients with dementia – A randomized
study. Journal of Clinical Psychopharmacology, 24 (6): placebo controlled clinical trial. International Journal of
582–591. Geriatric Psychiatry, 17: 579–585.
Roane, D.M., Feinberg, T.E. and Meckler, L. (2000). Stanniland, C. and Taylor, D. (2000). Tolerability of atypical
Treatment of dementia-associated agitation with antipsychotics. Drug Safety, 22 (3): 195–214.
gabapentin. Journal of Neuropsychiatry Clinical Street, J.S., Clark, W.S., Gannon, K.S. et al.; The HGEU
Neuroscience, 12: 40–43. Study Group. (2000). Olanzapine treatment of psy-
Rosenquist, K., Tariot, P. and Loy, R. (2000). Treatments chotic and behavioral symptoms in patients with
for behavioural and psychological symptoms in AD Alzheimer disease in nursing care facilities: A double-
and other dementias. In J. O’Brien (ed.), Dementia. blind, randomized, placebo-controlled trial. Archives
London: Edward Arnold, pp. 571–602. of General Psychiatry, 57: 968–976.
Rossom, R.C., Rector, T.S., Lederle, F.A. and Dysken, M.W. Street, J.S., Clark, W.S., Kadam, D.L. et al. (2001). Long-
(2010). Are all commonly prescribed antipsychot- term efficacy of olanzapine in the control of psychotic
ics associated with greater mortality in elderly male and behavioral symptoms in nursing home patients
veterans with dementia? Journal of American Geriatric with Alzheimer’s dementia. International Journal of
Society, 58 (6): 1027–1034. Geriatric Psychiatry, 16 (Suppl. 1): S62–S70.
Satterlee, W.G., Reams, S.G., Burns, P.R. et al. (1995). Streim, Joel E. et al. A Randomized, Double-Blind,
A clinical update in olanzapine treatment in schizo- Placebo-Controlled Study of Aripiprazole for the
phrenia and in elderly Alzheimer’s disease patients. Treatment of Psychosis in Nursing Home Patients with
Psychopharmacology Bulletin, 31 (3): 534. Alzheimer Disease. The American Journal of Geriatric
Scharre, D.W. and Chang, S.I. (2002). Cognitive and Psychiatry , Volume 16 , Issue 7 , 537 – 550
behavioral effects of quetiapine in Alzheimer’s disease Sultzer, D.L., Davis, S.M., Tariot, P.N. et al.; CATIE-AD
patients. Alzheimer Disease and Associated Disorders, Study Group. (2008). Clinical symptom responses
16: 128–130. to atypical antipsychotic medications in Alzheimer’s
Schneider, L.S., Dagerman, K. and Insel, P.S. (2006a). disease: Phase 1 outcomes from the CATIE-AD effec-
Efficacy and adverse effects of atypical antipsy- tiveness trial. American Journal of Psychiatry, 165 (7):
chotics for dementia: Meta-analysis of randomized, 844–854.
placebo-controlled trials. American Journal of Geriatric Sultzer, D.L., Gray, K.F., Gunay, I. et al. (1997). A double-
Psychiatry, 14 (3): 191–210. blind comparison of trazodone and haloperidol for
Schneider, L.S., Pollock, V.E. and Lyness, S.A. (1990). A treatment of agitation in patients with dementia.
metaanalysis of controlled trials of neuroleptic treat- American Journal of Geriatric Psychiatry, 5: 60–69.
ment in dementia. Journal of the American Geriatrics Tariot, P.N. (2012). Cessation of donepezil is asso-
Society, 38: 553–563. ciated with clinical decline in patients with
278 Dementia

moderate-to-severe Alzheimer’s disease compared to Teri, L., Logsdon, R.G., Peskind, E. et al. (2000). Treatment
continuation of donepezil or addition or substitution of of agitation in AD: A randomized, placebo-controlled
memantine. Evidence Based Medicine, 18 (2): 62–63. clinical trial. Neurology, 55: 1271–1278.
Tariot, P.N. (1999). Treatment of agitation in dementia. Trinh, N.H., Hoblyn, J., Mohanty, S. and Yaffe, K. (2003).
Journal of Clinical Psychiatry, 60 (Suppl. 8): 11–20. Efficacy of cholinesterase inhibitors in the treatment of
Tariot, P.N. (1996). Treatment strategies for agitation and neuropsychiatric symptoms and functional impairment
psychosis in dementia. Journal of Clinical Psychiatry, 57 in Alzheimer disease: A meta-analysis. JAMA, 289 (2):
(Suppl. 14): 21–29. 210–216.
Tariot, P.N. and Blazina, L. (1993). The psychopathology of Vigen, C.L., Mack, W.J., Keefe, R.S. et al. (2011).Cognitive
dementia. In J. Morris (ed.), Handbook of Dementing effects of atypical antipsychotic medications in
Illnesses. New York: Marcel Dekker Inc., pp. 461–475. patients with Alzheimer’s disease: Outcomes from
Tariot, P.N., Erb, R., Leibovici, A. et al. (1994). CATIE-AD. American Journal of Psychiatry, 168 (8):
Carbamazepine treatment of agitation in nursing home 831–839.
patients with dementia: A preliminary study. Journal of Wang, L.Y., Shofer, J.B., Rohde, K. et al. (2009). Prazosin
the American Geriatrics Society, 42: 1160–1166. for the treatment of behavioral symptoms in patients
Tariot, P.N., Erb, R., Podgorski, C.A. et al. (1998). Efficacy with Alzheimer disease with agitation and aggres-
and tolerability of carbamazepine for agitation sion. American Journal of Geriatric Psychiatry, 17 (9):
and aggression in dementia. American Journal of 744–751.
Psychiatry, 155: 54–61. Wilcock, G.K., Ballard, C.G., Cooper, J.A. and Loft, H.
Tariot, P.N., Farlow, M.R., Grossberg, G.T. et al. and (2008). Memantine for agitation/aggression and
Memantine Study Group (2004). Memantine treat- psychosis in moderately severe to severe Alzheimer’s
ment in patients with moderate to severe Alzheimer’s disease: A pooled analysis of 3 studies. The Journal of
disease already receiving donepezil: A randomized Clinical Psychiatry, 69 (3): 341–348.
controlled trial. JAMA 291: 317–24. Winblad, B., Mobius, H.J. and Stoffler, A. (2002).
Tariot, P.N., Frederiksen, K., Erb, R. et al. (1995). Lack of Glutamate receptors as a target for Alzheimer’s
carbamazepine toxicity in frail nursing home patients: disease – Are clinical results supporting the hope?
A controlled study. Journal of the American Geriatrics Journal of Neural Transmission Supplementum, 62:
Society, 43: 1026–1029. 217–225.
Tariot, P.N., Raman, R., Jakimovich, L. et al.; Alzheimer’s Winblad, B. and Poritis, N. (1999). Memantine in severe
Disease Cooperative Valproate Nursing Home Study dementia: Results of the 9M-Best Study (Benefit
Group. (2005). Divalproex sodium in nursing home res- and efficacy in severely demented patients during
idents with possible or probable Alzheimer’s disease treatment with memantine). International Journal of
complicated by agitation: A randomized controlled Geriatric Psychiatry, 14: 135–146.
trial. American Journal of Geriatric Psychiatry, 13 (11): Wooltorton, E. (2002). Risperidone (Risperdal): Increased
942–949. rate of cerebrovascular events in dementia trials.
Tariot, P.N., Salzman, C., Yeung, P.P. et al. (2000a). Long- CMAJ, 167: 1269–1270.
term use of quetiapine in elderly patients with psy- Wragg, R.E. and Jeste, D.V. (1989). Overview of depres-
chotic disorders. Clinical Therapy, 22: 1068–1084. sion and psychosis in Alzheimer’s disease. American
Tariot, P.N., Schneider, L., Katz, I. et al. (2002a). Journal of Psychiatry, 146: 577–587.
Quetiapine in nursing home residents with Alzheimer’s Zheng, L., Mack, W.J., Dagerman, K.S. et al. (2009).
dementia and psychosis. American Journal of Geriatric Metabolic changes associated with second-generation
Psychiatry, 10 (2): 93. antipsychotic use in Alzheimer’s disease patients: The
Tariot, P.N., Schneider, L., Mintzer, J. et al. (2001a). Safety CATIE-AD Study. American Journal of Psychiatry, 166:
and tolerability of divalproex sodium for the treatment 583–590.
of signs and symptoms of mania in elderly patients with Zhong, K.X., Tariot, P.N., Mintzer, J. et al. (2007).
dementia: Results of a double-blind, placebo-­controlled Quetiapine to treat agitation in dementia: A random-
trial. Current Therapeutic Research, 62: 51–67. ized, double-blind, placebo-controlled study. Current
Tariot, P.N., Schneider, L.S., Cummings, J. et al.; Alzheimer Research, 4 (1): 81–93.
Alzheimer’s Disease Cooperative Study Group. (2011). Zivin, K., Pfeiffer, P.N., Bohnert, A.S. et al. (2013).
Chronic divalproex sodium to attenuate agitation and Evaluation of the FDA warning against prescribing
clinical progression of Alzheimer disease. Archives of citalopram at doses exceeding 40 mg. American
General Psychiatry, 68 (8): 853–861. Journal of Psychiatry, 170 (6): 642–650.
26
Psychological approaches for the practical
management of cognitive impairment in
dementia

ANNE UNKENSTEIN

not imply global impairment of brain function. There will


26.1 INTRODUCTION nearly always be some retained abilities. It is important to
identify these cognitive strengths and use them positively.
A psychological approach to the management of cognitive Interventions can draw on retained skills in order to bypass
impairment in dementia aims to promote the best possible weaknesses.
quality of life for both people with dementia and their car- Cognitive impairment is just one aspect of dementia.
ers, by offering positive, individualized and effective man- People with dementia also experience changes in behaviour,
agement. With the growing emphasis on early diagnosis of emotional control, personality and self-care. These changes
dementia, there are increasing opportunities to provide psy- need to be taken into account when working on strategies
chological interventions from the earliest stages of demen- for cognitive impairment. A team approach with agreement
tia. This chapter focuses on practical and personalized about realistic goals and consistent strategies is recom-
strategies. Assessment and feedback processes are outlined mended. Carers need to be included in management plan-
and specific techniques for managing cognitive impairment ning. Wherever the person with dementia lives, carers will
in early, moderate and later stage dementia are described. be involved. They may be relatives, friends, neighbours or
health professionals.
Not all of the person’s symptoms are directly due to brain
disease. People with dementia are very sensitive to their
physical and psychological environment. Dementia is some-
26.2 FACTORS INFLUENCING
times first suspected when a person experiences a change
MANAGEMENT OF COGNITIVE to his or her usual environment such as moving to a new
IMPAIRMENT IN DEMENTIA house or the death of a spouse. It is important to be aware
of environmental factors that may adversely affect cognitive
Each person with dementia is unique, with a complex inter- function and make appropriate changes to the environment
action of brain disease, personality, biography, health and in order to enhance cognitive abilities.
social psychology (Kitwood, 1993). Each person needs to Health factors can exacerbate any cognitive impair-
be treated as an individual. An approach that might prove ment. Sensory impairment, especially of vision and
successful for one person may prove ineffective for another hearing, can adversely affect cognitive function. People
(Woods and Bird, 1999). There is no single correct approach with dementia commonly experience anxiety, distress
to the management of cognitive impairment in dementia. and depression. Confusion can also be increased during
The help that is relevant can vary considerably. acute or chronic illness and with any physical discomfort
There is a great deal of variation in cognitive function or pain. Alcohol, some medications and fatigue may
in dementia according to the stage of dementia and differ- cause transient exaggeration of cognitive impairment.
ent causes and patterns of brain pathology. Dementia does It is necessary to be alert to any superimposed health

279
280 Dementia

problems and to control these to help people maintain they have had with strategies that they have already tried. It
maximum function. can also provide emotional support and practical help and
Progressive decline in cognitive function is characteris- assistance. Carers have access to a lot of general information
tic of dementia. Management strategies need to be flexible about dementia (e.g. Draper, 2011). However, specific infor-
and adapted over time. Assessment at regular intervals will mation that is more relevant to their particular situation can
allow for the development of realistic expectations. The aim be provided in a feedback session.
of any strategies employed is to maintain as high a level of Feedback usually includes the person with dementia,
function as possible by helping the individual to better uti- but can be given to carers alone where appropriate. The
lize the remaining function. psychologist can provide a description of the person’s
cognitive strengths and weaknesses. This information
is then used to help carers make sense of behaviour
that otherwise would seem to suggest that the person is
26.3 ASSESSMENT purposefully being annoying. For example, by outlining
the different types of memory and how each is affected,
A thorough assessment is required in order to design an it becomes easier to understand why a person can recall
individualized approach to management of cognitive information from their remote past but not what was
impairment. This should include assessment of the person’s said 2 minutes ago. Furthermore, if carers know that the
medical and psychological status and assessment of cog- person has memory impairment they are more likely to
nition. Assessment of the person’s health is important, as understand why the person persistently repeats the same
there may be additional treatable medical conditions. question or forgets to pass on a phone message. It can be
It is beneficial to interview the carers to gain informa- reinforced that this is not done deliberately. Behaviour
tion about the person’s personality, interests, home environ- is thus reinterpreted as related to brain disease and not
ment and the history of events leading up to the assessment. purposeful or intended. This information is also useful
Information about strategies that the carers have already when it comes to management of cognitive impairment.
tried and their willingness to provide support can also be For example, if carers know that forgetfulness is due to
gathered. memory loss from brain disease they are more likely to
A neuropsychological assessment provides a basis for understand that the person is unlikely to learn by repeti-
individual intervention. It allows for a focus on strengths tion. They are also more likely to use strategies that avoid
by outlining abilities that remain unaffected and also reliance on intact memory ability.
outlines areas of cognitive impairment to be managed. A feedback session offers an opportunity to ask the per-
Serial neuropsychological assessment can be used to son with dementia and the carers how the cognitive prob-
monitor progression of symptoms over time. It can pro- lems manifest themselves in everyday life. This information
vide an objective viewpoint to the carers and to the person can be used to make strategies realistic and relevant to the
with dementia with regard to the progression of symp- life of the person with dementia (Wilson, 2004).
toms. Part of the assessment is to check the attitude of the A written reminder of what was discussed during the
person to their symptoms, as this will influence choice of feedback session can be useful. Both the person with demen-
strategies. tia and the carers may forget what was discussed, especially
The assessment phase will identify the problem but it is if they are just coming to terms with the diagnosis.
important to go beyond this phase for a better management An initial discussion of strategies to help manage cog-
of cognitive impairment. It is beneficial to share the infor- nitive impairment can be included in a feedback session.
mation gathered with the person with dementia and their The types of strategies employed will vary according to
carers. the person’s individual situation and the cause and stage of
dementia.

26.4 FEEDBACK AFTER
PSYCHOLOGICAL ASSESSMENT 26.5 EARLY STAGE DEMENTIA –
MEMORY STRATEGIES
A feedback session following psychological assessment can
serve many purposes. It provides information about how the In early dementia, the most significant cognitive impair-
disease is affecting aspects of cognitive function in order to ment is typically in memory function. Impairment of new
improve understanding. While it is important to acknowl- learning is most prominent, including acquisition and
edge areas of loss, a positive approach can be maintained by retention of new information. People with early demen-
highlighting what the person can still do. tia may forget what you have just told them, forget where
The carers are often the people who provide ongoing they have put something, repeat the same thing as if they
management of the person with cognitive impairment. have not said it before or have difficulty learning how to use
Feedback can empower carers by acknowledging the success something new.
Psychological approaches for the practical management of cognitive impairment in dementia 281

At this stage, the person often remains aware of cogni- When people with dementia are supported with repeated
tive losses and may be motivated to seek methods to com- training sessions, they can learn and retain new informa-
pensate for them. The person is usually living in the family tion using specific learning methods. One technique that
home. Initial strategies therefore focus on the individual has been found to assist learning is spaced retrieval, which
with increasing involvement of family carers over time. The requires people to recall information over increasingly lon-
aim is to maintain as much independence as possible and to ger periods of time. This is sometimes combined with other
promote confidence in remembering. Family members and learning techniques, such as the errorless learning approach,
friends need to avoid taking over responsibilities too soon. where individuals are not allowed to commit errors when
Many of the strategies that are used by people experi- they are receiving memory training (see Clare, 2008).
encing normal memory change with ageing and are use- Some well-known internal strategies or mnemonic tech-
ful at this stage (see Sargeant and Unkenstein, 2001, for a niques, such as the method of loci and peg word systems,
detailed description of memory strategies). It is important are quite complex. These techniques require considerable
to ask people whether they are already using any memory remaining cognitive capacity including new learning and
aids before explaining the use of memory strategies. People motivation for success. People with early stage demen-
should be encouraged to draw on strategies that they have tia usually do not have the motivation or learning ability
used in the past. They will be more likely to use strategies required to use these techniques (Grandmaison and Simard,
if they are already familiar with them (Kotler-Cope and 2003).
Camp, 1990). Everyone has different preferences. Some External strategies aim to compensate for memory
people like to use diaries while others prefer a noticeboard. impairment by reducing the demand on a person’s memory.
People typically need to use a range of strategies, rather They are generally easier to use and more suitable for every-
than just one technique. It is important to find out the typi- day remembering than internal strategies. They may involve
cal things a person tends to forget and work out strategies written reminders, storage of objects in specially designated
that will suit these specific situations and be usable for that places, technical memory devices and reminders from other
person. Strategy use will vary according to differing inter- people.
ests, lifestyles and home environments. A home visit can be Written reminders assist with remembering locations of
useful to help tailor strategies to particular situations. objects, a task that has to be done, appointments, shopping
The emphasis is on practical compensatory techniques, items, messages, names and instructions. Calendars, dia-
which avoid challenging the person’s lost memory function. ries, notebooks, notepads, noticeboards and sticky notes are
For example, rather than asking a question that relies on all useful. Having a pen or pencil attached reduces the load
intact memory function such as ‘What show did you watch on memory even further. Such written aids should not only
on television last night?’, rephrase the question to ‘Did be readily visible and accessible but should also be in close
you enjoy the show that you watched last night?’ If people proximity to the to-be-remembered activity. For example, a
become repetitive it may be best to try distracting them shopping list pad on the refrigerator door or instructions for
with another topic of conversation. Enhancing familiarity how to operate a piece of equipment permanently displayed
is also beneficial. Developing a regular routine and doing on that equipment.
things at a set time of the day or week will reduce the load External memory strategies can assist with orientation
on memory. in early stage dementia. It is helpful to mark off the days as
Memory strategies can be thought of as internal or exter- they pass on a calendar, or use a flip-top desk calendar and
nal. Internal strategies involve some form of internal mental turn each day over as it passes. Newspapers can also serve
manipulation, for example, mentally retracing one’s steps as prompts. Clocks should be positioned in places where
to remember where an object was left. External strategies they are readily seen and ideally should have day and date
involve using some sort of external aid, for example a note built in.
pad for writing a list or a diary. They can also involve mak- Storing objects in specially designated places can help
ing changes to the environment, for instance, having desig- with remembering the location of objects, a task that has
nated locations in which to store particular items. to be done, or something to be taken out. For example, keys
Internal strategies focus on enhancing encoding and can be stored near the front door of the house. This makes
retrieval processes. The emphasis is on making more it easy to pick up the keys when leaving the house and to get
efficient use of current cognitive capacity. A motivated into the habit of putting them back when returning.
individual with early stage dementia may employ simple Leaving objects in visible locations can act as a reminder
internal strategies. These strategies typically involve one of to do something. Medications can be left somewhere obvi-
three main features: focusing attention, adding meaning ous in the kitchen if they are to be taken with meals. Objects
to the information to be remembered or reducing the that a person needs to take out with them can be located in a
amount of information to be remembered. Simple internal special place near the front door, such as a hall table.
strategies such as ensuring understanding, associating A useful storage device for managing medications is a
new information with information that is known well and dosette box, divided into compartments that relate to the
visualization may be useful for motivated individuals with day of the week or time of the day into which a set number
early dementia. of tablets can be placed. The box has to be filled at weekly
282 Dementia

or other intervals, but is simple to use and let people know for other areas of cognitive impairment are also required.
whether they have taken the necessary tablets. Providing support and information to the carers can help to
Basic alarms are useful to help people remember some- further tailor strategies to make them suit the individual’s
thing they have to do. A common example is an oven timer current cognitive strengths and weaknesses.
used as a reminder to turn the oven off. Many household Carers will need to become increasingly involved in the
objects have built in memory aids. Most telephones have use of external memory strategies and can assist with set-
a system that enables storage of frequently used numbers ting up a designated ‘memory centre’ in a visible location
that can then be dialled by pressing only one appropriately in the home environment (Sargeant and Unkenstein, 2001).
labelled button. Automatic shut-off devices on electrical Here the carer could hang a whiteboard in which the day,
and gas appliances, such as stovetops, kettles or lights can date and daily schedule are written or a clock with the day
be installed. These devices turn the power off after a period and date on it. A pin board next to the whiteboard can be
of time if the appliances are left on. More complex electronic used to display other important information. The carer may
memory aids, including smart phones and computers, can need to keep reminding the person to check the memory
be useful for motivated and insightful individuals but may centre. Carers can also develop ‘communication books’. For
require training to know how to operate them. They usually this a notebook is left in a special location where visitors can
rely on other cognitive processes remaining intact and may write messages. The person can be encouraged to check this
not suit the person with early stage dementia. book regularly as a reminder of who has visited recently.
Reminders from other people can provide an excellent These strategies are a form of reality orientation, an
memory back-up system. Family members can provide tele- approach that emphasizes the use of external cues and
phone reminders or leave notes in a regular and prominent structures to assist the person in maintaining contact with
location. Reminders need to be specific. For example, a note the environment (see Woods, 2002 for a review of reality
saying “back in 5 minutes” does not provide enough infor- orientation). Carers can help to provide personalized real-
mation. The person with dementia needs to know when the ity orientation, by identifying to which realities the person
person will return. needs or desires to be oriented. A structured programme
People with mild to moderate dementia can be taught of reality orientation and cognitive stimulation for people
to use memory strategies using a range of approaches. with dementia can improve memory and functional abili-
Cognitive training programmes typically involve repeated ties (Spector et al., 2010). Typically, a structured stimulation
practice of cognitive tasks or specific memory strategies, programme would consist of several weeks of group inter-
whereas cognitive rehabilitation aims to improve an indi- vention sessions (see Droes et al., 2011 for a review of cogni-
vidual’s everyday functioning. Bahar-Fuchs et al. (2013) and tive stimulation therapy).
Choi and Twamley (2013) review the efficacy of cognitive Carers can continue to encourage a regular routine of
training and rehabilitation for people with dementia. familiar activities. Sudden unexplained changes or unfa-
miliar activities could cause the person distress. Carers can
help the person focus on activities that are familiar and
based on previous interests, such as gardening, walking the
26.6 MODERATE DEMENTIA –
dog or other household tasks. Drawing on the continued
WORKING WITH CARERS strength of memories of procedures and lifetime memories
can provide meaningful activity. Carers at accommodation
As dementia progresses people can experience increasing facilities can find out about the person’s past likes, dislikes,
memory loss, language difficulties and specific disorders lifestyle and life interests from family members. This infor-
such as apraxia. They may be unable to function indepen- mation and any old photos available can be recorded in a
dently outside the home and require increasing assistance book that can be used as a cue for discussion and reminis-
with tasks of personal care. They could be living in their cence (see Subramaniam and Woods, 2012 for a review of
own home or supported accommodation. By this stage reminiscence therapy).
the person’s insight into their cognitive losses usually has A familiar environment will reduce the demand on the
diminished. person’s memory. Unnecessary changes such as rearranging
With little awareness of their difficulties and increased furniture should be avoided. If changes have to be made,
cognitive impairment, it becomes difficult to work on strat- they should be introduced slowly, with explanation.
egies with the person with dementia alone. The manage- Carers may need assistance with strategies for other
ment of cognitive impairment, therefore, becomes focused areas of cognitive impairment. For example, the person at
on working in collaboration with family members or other this stage may experience apraxia, an inability to carry out
carers. Carers know the person best and often come up with routine patterns of voluntary movement. A common prob-
strategies that work well for that person. Carers can pro- lem caused by apraxia is difficulty with dressing. Helping
vide reminders for the person, encourage the person to use the carer to understand that a person can sometimes dress
external memory aids and make appropriate changes to the automatically, but has difficulty when the movement comes
environment. Without this assistance the person is unlikely under conscious control is useful. To remove the degree of
to make use of memory strategies. By this stage strategies voluntary focus from the action, avoid giving the person
Psychological approaches for the practical management of cognitive impairment in dementia 283

direct instructions about performing the task. Specific tech- To maximize cognitive function it helps to simplify the
niques include distracting the person’s attention from the environment to avoid the person becoming overwhelmed.
task, imitation, laying the clothes out in the order in which If there is excessive stimulation then confusion may be
the person likes to put them on and giving reassurance and exacerbated. It helps to remove distractions, keep noise lev-
praise for each successful step. See Holden (1995) for strate- els down and not have too many people around (Robinson
gies for other specific cognitive disorders. et al., 2001).
It is often difficult to communicate with the individual
with this stage of dementia. Using an empathic, validating
approach that displays respect and sensitivity to feelings can
26.7 LATER STAGE DEMENTIA – help to ensure understanding.
ENVIRONMENTAL ADAPTATION

In the later stages of dementia people experience marked 26.8 CONCLUSION


confusion and widespread cognitive disturbance. Most
people with advanced dementia are living in supported
accommodation with professional carers. Strategies for Improving the practical management of cognitive impair-
management of cognitive impairment need to be focused ment in dementia using a psychological approach is just
more on the environment in which the person is living. one part of the challenge to maintain maximum function
Adaptations are made to the environment rather than throughout the course of the illness (Kitwood, 1993). For
expecting the individual to change. Visiting the person’s any symptom of dementia, the first step involves investi-
residence allows one to observe environmental cues, how gating what may be causing the person to present in that
they might best be used and/or changed and how new ones particular way. Multidisciplinary assessment will establish
might be introduced (Kapur et al., 2004). whether there are any environmental factors that can be
At this stage, people become dependent on their envi- altered or health factors that can be treated to enhance the
ronment to support them. Making adaptations to the envi- individual’s function. Effective management strategies that
ronment can enhance feelings of security and help people are relevant to the stage of dementia and other individual
cope with their confusion and disorientation. A ‘demen- variables can be developed using a collaborative approach
tia friendly environment’ provides orientation strategies involving the person with dementia, carers and health
that help to reduce confusion and promote independence professionals.
(Davis et al., 2009). When one first moves into a new resi-
dence there is often a settling in period. An unfamiliar and REFERENCES
a new environment can increase confusion. It helps to make
the environment as familiar as possible. Possessions, such Bahar-Fuchs, A., Clare, L. and Woods, B. (2013). Cognitive
as photos, objects and furniture can be put in the person’s training and cognitive rehabilitation for persons with
room. Assistive technology, involving sensor systems and mild to moderate dementia of the Alzheimer’s or
computers, can provide further support (Charness et al., vascular type: A review. Alzheimer’s Research and
2012; Kurz et al., 2013). Therapy, 5: 1–14.
A routine and daily timetable should be emphasized. Charness, N., Best, R. and Souders, D. (2012). Memory
Orientation information or cues can lessen confusion. For function and supportive technology. Gerontechnology,
example, having access to a window helps a person to tell 11: 22–34.
the time of the day and the weather. A good sensory envi- Choi, J. and Twamley, E. (2013). Cognitive rehabilitation
ronment with adequate light is important. Making each therapies for Alzheimer’s disease: A review of methods
part of a large place unique in some way is useful for orien- to improve treatment engagement and self-efficacy.
tation to the environment. Colour and furniture can high- Neuropsychology Reviews, 23: 48–62.
light the difference. Smaller spaces are less confusing than Clare, L. (2008). Neuropsychological Rehabilitation and
large spaces. People with Dementia. Hove: Psychology Press.
Clear labelling throughout the accommodation facil- Davis, S., Byers, S., Nay, R. and Koch, S. (2009). Guiding
ity can further lessen confusion. Way finding cues can be design of dementia friendly environments in residen-
marked on the floor or walls with coloured lines or arrows. tial care settings. Considering the living experiences.
Individual rooms can be clearly labelled. Large uppercase Dementia, 8: 185–203.
print with black against white is often best for older people Draper, B. (2011). Understanding Alzheimer’s and Other
(Kapur et al., 2004). Pictures may be more easily under- Dementias. Woollahra, NSW: Longueville Books.
stood than written labels (e.g. a picture of a toilet on the Droes, R., van der Roest, H., van Mierlo, L. and Meiland, F.
toilet doors). Carers can wear name badges, with their first (2011). Memory problems in dementia: Adaptation and
name only, that are large enough to read. People will require coping strategies and psychosocial treatments. Expert
continued instruction on how to use these orientation aids. Review of Neurotherapeutics, 11: 1769–1782.
284 Dementia

Grandmaison, E. and Simard, M. (2003). A critical review Related Illnesses. Ann Arbor, MI: The Alzheimer’s
of memory stimulation programs in Alzheimer’s Program, Eastern Michigan University.
disease. Journal of Neuropsychiatry and Clinical Sargeant, D. and Unkenstein, A. (2001). Remembering
Neuroscience, 15: 130–144. Well: How Memory Works and What to do When it
Holden, U. (1995). Ageing, Neuropsychology and the Doesn’t. St Leonards, NSW: Allen and Unwin.
‘New’ Dementias. London: Chapman and Hall. Spector, A., Orrell, M. and Woods, B. (2010). Cognitive stim-
Kapur, N., Glisky, E. and Wilson, B. (2004). External ulation therapy (CST): Effects on different areas of cog-
memory aids and computers in memory rehabilitation. nitive function for people with dementia. International
In A.D. Baddeley, M. Kopelman and B.A. Wilson (eds.), Journal of Geriatric Psychiatry, 25: 1253–1258.
The Essential Handbook of Memory Disorders for Subramaniam, P. and Woods, B. (2012). The impact of
Clinicians. Chichester: John Wiley, pp. 301–328. individual reminiscence therapy for people with
Kitwood, T. (1993). Person and process in dementia. dementia: Systematic review. Expert Review of
International Journal of Geriatric Psychiatry, 8: 541–545. Neurotherapeutics, 12: 545–555.
Kotler-Cope, S. and Camp, C.J. (1990). Memory interven- Wilson, B.A. (2004). Management and remediation of
tions in aging populations. In E. Lovelace (ed.), Aging memory problems in brain-injured adults. In A.D.
and Cognition: Mental Processes, Self-awareness Baddeley, M. Kopelman and B.A. Wilson (eds.),
and Interventions. North Holland: Elsevier Science The Essential Handbook of Memory Disorders for
Publications, pp. 231–261. Clinicians. Chichester: John Wiley, pp. 199–226.
Kurz, A., Clare, L. and Lautenschlager, N. (2013). Add life Woods, B. (2002). Reality orientation: A welcome return?
to years: Psychosocial interventions for people with Age and Ageing, 31: 155–156.
cognitive disorders. International Psychogeriatrics, 25: Woods, B. and Bird, M. (1999). Non-pharmacological
1389–1391. approaches to treatment. In G. Wilcock, R. Bucks and
Robinson, A., Spencer, B. and White, L. (2001). K. Rockwood (eds.), Diagnosis and Management of
Understanding Difficult Behaviours. Some Practical Dementia: A Manual for Memory Disorders Teams.
Suggestions for Coping with Alzheimer’s Disease and Oxford: Oxford University Press, pp. 311–331.
27
Sexuality and dementia

JOE STRATFORD

more sexually active than in previous years. In a Swedish


27.1 INTRODUCTION study stretching over a 30-year period looking at trends,
Beckman et al. (2008) studied four cohorts of 70-year-old
Dementia and its relationship with sexuality raises many community-dwelling older people. Over 1500 subjects were
significant medical, ethical, cultural and relationship issues. assessed at four separate intervals (1970–1971, 1976, 1992
Our sexuality is one of the most basic aspects of our human- and 2000), each assessment being identical although each
ity and encompasses a wide range of elements including assessed a different cohort of 70 year olds. Sexual activity
gender identities, sexual orientation, intimacy, beliefs, behav- (defined as intercourse within the last 12 months) in males
iours and roles. Moreover, society is becoming increasingly increased from 47% in 1970–1971 to 66% in 2000. In females,
open about sexuality and sexual behaviour in general. As the increase was from 12% to 34% over the same time.
the world’s population ages and we see associated increases Overall, older women are less likely to be sexually active
in numbers of individuals affected by dementia, the issue of than older men. This may be in part due to the decreased
sexuality and dementia will only increase in its relevance. likelihood of women being in a spousal or intimate relation-
This chapter addresses some of these complex issues and ship, often as a result of separation or bereavement. For both
highlights some approaches towards managing various dif- sexes, sexual activity is also related to physical health. Those
ficulties raised. who rate their health as poor are less likely to be sexually
active than those who rate their health as very good or even
excellent (Lindau et al., 2007).
27.1.1 SEXUALITY AND AGEING Sexual activity of course does not always have to mean
sexual intercourse. As people age, intimacy with a partner
Many myths about sexuality and ageing exist within popu- appears to be expressed in different ways and there may be
lar culture in a way which would not be tolerated on grounds physiological reasons behind this. Touching, holding hands,
of race or religion. Sexuality in older adults is often regarded hugging and kissing can assume greater importance when
as being amusing or revolting (Kessel, 2001). Younger pop- compared to activities such as intercourse or masturbation
ulations also overestimate the degree of sexual problems, (Ginsberg et al., 2005).
which older people encounter (Allen et al., 2009).
Although sexual activity appears to decline with advanc-
ing age (Schiavi et al., 1990; Kaiser, 1996), it does not appear 27.1.2 SEXUALITY AND RESIDENTIAL
to cease completely. In a large U.S. study, Lindau et al. (2007) CARE
studied over 3000 older adults (age range 57–85 years) and
found that while 84% of the 57–64 year olds reported sexual As the numbers of ageing individuals increase, so does the
activity in the last 12 months, 39% of the 75–85 year olds population of those living in care, be it residential or nursing
were still active. And in a study of almost 3000 community- home establishments. The expression of these residents’ sexu-
dwelling elderly Australian males, 11% of the 90–95 year ality is particularly challenging in such institutions for a vari-
olds had been sexually active in the preceding 12 months ety of reasons including ill health or a lack of partners (Bauer
(Hyde et al., 2010). et al., 2009). The move into institutional residential care inev-
Although overall levels of sexual activity may decrease itably leads to a loss of privacy especially where personal care
with age, there is evidence that the elderly are becoming and supervision are required (Bouman et al., 2006).

285
286 Dementia

Overall, much of the evidence related to sexuality and its 27.2.2 CAPACITY AND ETHICAL ISSUES
expression in residential facilities is mixed. There seems to
be a huge variation in attitudes to this issue dependent on People with dementia should have a right to express their
what kind of facility, its ‘culture’ and which staff group is sexual feelings engage in sexual behaviour in a way that is
being assessed (Doll, 2013). no different to the rest of society. The difficulty which arises
within the context of dementia relates to firstly whether the
expression of such feelings conflicts with the basic rights of
27.2 EFFECTS OF DEMENTIA ON those affected by such activity but also importantly, whether
the issue of capacity has been considered. Failure to do so
SEXUALITY AND SEXUAL
runs the risk of committing a sexual offence (e.g. sexual
BEHAVIOUR activity without consent).
Capacity is not an all-or-nothing concept. It can vary
The onset of dementia may have a significant effect on over time and is decision specific. The same would apply
the sex life of the individual and their partner, although to the capacity for an individual to engage in a sexual rela-
the evidence base for such changes in sexual behaviour in tionship. In the United Kingdom, the Mental Capacity Act
dementia remains small. Wright (1991) undertook a con- (2005) specifically excludes decisions about sexual rela-
trolled study of the impact of Alzheimer’s disease (AD) on tions as it is felt that this is not something that one indi-
marital relationships. Eight of the 30 (27%) couples of the vidual can decide for any other. This obviously leads to
dementia-affected group were sexually active, compared complexities when addressing issues of sexual behaviour
with 14 of the 17 (82%) controls. In those individuals still where one (or even both) party felt a lack of capacity in
sexually active, compared with controls, sex took place this regard.
more often in couples where one partner had dementia. Due to its sensitive and complex nature, the capacity of
Derousesné et al. (1996) in a survey of 135 married cou- a demented individual to enter into a sexual ­relationship
ples, one of whom had possible or probable AD, found that could be difficult to establish. In an attempt to address this,
80% of spouses reported a change in patient’s sexual activity. Lichtenberg and Strzepek (1990) ­developed a structured
This was not linked to the degree of cognitive impairment assessment technique to aid decisions about patients’ abil-
or the gender of the patient. Eloniemi-Sulkava et al. (2002) ity to consent to sexual relationships. It includes s­ections
questioned the spousal caregivers of 42 demented patients addressing the patient’s awareness of the relationship, their
over a 7-year period. At the onset of dementia, 32 (76%) ability to avoid exploitation and an assessment of their
couples engaged in regular sexual intercourse. By the sev- awareness of any potential risks that could arise from it.
enth year, seven (28%) couples continued to engage in such The right of an individual with dementia and who lacks
activity. While 25 caregivers (60%) reported some negative capacity to make decisions about his or her sexual activity
behavioural changes in the sexual side of the marriage, four raises some profound ethical issues. In an attempt to explore
(10%) reported positive changes. In most studies of this type, the area, Mahieu and Gastmans (2011) helpfully suggest
sexual demands and activity seem to decrease following the considering familiar medical ethical issues of autonomy,
onset of dementia but only in small proportions do couples beneficence, non-maleficence and justice when dealing with
report significant dissatisfaction (Dourado et al., 2010). these issues. They also add that principles of dignity and
vulnerability need also to be incorporated.
A lack of capacity can expose an individual to the risk
27.2.1 RELATIONSHIP EFFECTS
of abuse in its many forms. Sexual abuse of demented indi-
Dementia leads to an inexorable change in the nature of most viduals can occur and their vulnerability arguably makes
relationships, but none is more poignant than that between them even more at risk (Burgess and Phillips, 2006).
partners. Partners of individuals with dementia become
carers as the disease process robs an individual of their
autonomy. This can lead to such carers being vulnerable to
depression and anxiety as carer burden increases (see other
27.3 INAPPROPRIATE SEXUAL
chapters). Although not all dementia-affected couples experi-
ACTIVITY
ence a change in sexual activity, an altered sexual relationship
is a difficulty many couples face after the onset of dementia. There is no clear consensus on what constitutes inappro-
The diagnosis should not be assumed to herald cessation of priate sexual activity in the context of dementia or even
an active sex life, and couples should be encouraged to think its prevalence. This is probably in part due to the relatively
and act flexibly in order to adjust to the impact of the disor- small and heterogeneous populations studied, as well as
der (Davies et al., 1998). If sexual activity is experienced to inconsistent definitions of such sexual behaviour (Ward and
be problematic, it is important to stress to patients and carers Manchip, 2013). Despite this lack of consensus, a reason-
that physical intimacy, such as cuddling or simply holding ably large body of evidence has developed in recent years
hands, can be just as beneficial and rewarding to both. examining the nature and range of sexually inappropriate
Sexuality and dementia 287

behaviour in dementia. The terms ‘hypersexuality’, ‘sexual however, in their study of 133 patients with dementia found
disinhibition’ and ‘inappropriate sexual behaviours’ (ISB) no such differences between dementias of various types.
are also often used interchangeably.
27.3.3 CAUSES OF HYPERSEXUALITY
27.3.1 TYPES OF BEHAVIOUR
Hypersexuality can arise from a variety of causes, even in
In addition to considering whether any particular behav- the absence of dementia. Structural factors such as frontal
iour is abnormal, we need to also try and define specific lobe damage and temporal lobe dysfunction, leading to the
behaviours – as the range of unwanted acts/symptoms can Klüver–Bucy syndrome, have been associated with sexually
be varied. As Series and Dégano (2005) note, the decision disinhibited behaviours. Hypothalamic dysfunction as seen
of when a particular behaviour becomes inappropriate or in the Kleine–Levin syndrome can result in hypersomnia,
aberrant needs to be based on a judgement of what is normal hyperphagia and hypersexuality, especially in males. It is
or appropriate for a specific individual in a particular situa- important to draw a distinction as to whether this behav-
tion. This may differ according to the environment or on the iour is simply part of a generalized disinhibition syndrome
level of risk or discomfort to others. as we often see in dementia or a true ‘hypersexual’ disorder
Szasz (1983) offered a framework for objectively describ- manifesting in markedly increased sexual drive or activity.
ing ISB. This categorization splits these behaviours into the Pharmacological agents such as alcohol and other
following: sedatives can produce behavioural and sexual disinhibi-
tion in both healthy individuals and those with dementia.
●● Sexual talk – inappropriate comments and propositions Dopamine agonists can also cause hypersexuality in patients
●● Sexual acts – inappropriate touching of oneself or with Parkinson’s disease as well as a range of other impulse
others, exposure of genitals, masturbation or even control disorders such as gambling (Pontone et al., 2006).
intercourse In dementia, it is not difficult to see how ISB could arise.
●● Implied sexual acts – the reading/watching of porno- Social factors such as the misinterpretation of cues (e.g. dur-
graphic material ing the delivery of personal care such as washing or being
aided to dress/undress) and premorbid patterns of sexual
Despite the relative lack of consensus in categorizing ISB, activity may also all play a part in the development of sexu-
the literature often includes making explicit sexual com- ally inappropriate behaviour (Series and Dégano, 2005).
ments, inappropriate touching of one’s own genitals or the
genitals/breasts of others. Inappropriate disrobing or mas-
turbation in public areas right through to attempted inter-
course with a non-consenting person all have been described. 27.4 MANAGEMENT OF ISB
The context of such behaviour is critical as what might be
considered acceptable in private homes or bedrooms could Any strategy to manage sexually inappropriate behaviour
well result in distress, fear or even outrage in public spaces needs to begin with a comprehensive assessment of the
or communal areas of residential facilities. behaviour. This will involve a description of the behav-
iour but also a consideration of the frequency and context.
27.3.2 PREVALENCE Careful and sensitive exploration, where possible, of an
individual’s sexual history, behaviour and attitudes may
Prevalence rates of such behaviour vary significantly well shed light and provide an insight into current problem-
between 2% (Devanand et al., 1992) and 25% (Szasz, 1983) atic symptoms Another vital consideration is how much of
depending on the study, the setting and the relatively het- a problem this behaviour presents and to whom, as well as a
erogeneous nature of the criteria used. Burns et al. (1990) comprehensive risk assessment (Series and Dégano, 2005).
reported disinhibition to be present in 7% of a sample of A critical element of the assessment is to try and deter-
178 patients with AD. In this study there was no difference mine whether these unwanted behaviours occur within the
between males and females. However, Black et al. (2005) in context of other challenging behaviours of dementia such as
their review, state that these behaviours are more common in agitation or aggression or if the hypersexuality per se is the
males. There is also evidence to show that these behaviours primary issue. This may provide clues not only to the aetiol-
can present in mild cognitive impairment right through to ogy but also could dictate the treatment strategy.
advanced dementia (Alagiakrishnan et al., 2005), although Knight et al. (2008) have developed a validated rating
Bardell et al. (2011) suggest that they are more common scale to evaluate ISB in people with cognitive impairment
with advancing cognitive impairment. Patients with AD and brain injury, but other tools exist such as the Manchip
have been reported as being less likely to present with such Rating Scale (Ward and Manchip, 2013) to specifically
behaviour when compared with other types of dementia examine these difficulties in the context of dementia. Such
(Zeiss et al., 1996; Alagiakrishnan et al., 2005). Most stud- scales are useful in identifying target symptoms, which in
ies suggest that sexual disinhibition is found in dementias turn if used at intervals, can be used to measure the efficacy
with a significant vascular component. Tsai et al. (1999), of any intervention.
288 Dementia

27.4.1 NON-PHARMACOLOGICAL (Series and Dégano, 2005). The sexual dysfunction, which is


APPROACHES sometimes seen with the use of such medications (Stimmel
and Gutierrez, 2006) in everyday clinical practice, could well
Ward and Manchip (2013) note that non-drug approaches be the mechanism behind the beneficial effect seen in cases
need to take into consideration the environment, culture and of ISB. Case reports suggest that these agents can be effec-
attitudes of carers. If the setting is a care home, then provid- tive even after a variety of other psychotropic medications
ing adequate space for privacy may well lessen the need or have failed (Tucker, 2010). Leo and Kim (1995) also found
impulse for individuals to engage in ISB in more public areas, that clomipramine – a tricyclic antidepressant but with sig-
etc. Staff need to remain sensitive to the needs of their resi- nificant serotonergic properties – was effective in treating
dents in expressing their sexuality in appropriate ways, and two dementia patients exhibiting sexual disinhibition.
not discouraging discussion or attempts to address the issue.
Specific interventions to reduce such unwanted behav- 27.4.2.2 Antipsychotics
iours have been suggested (Jensen, 1989; Howell and Watts,
1990). Most rely on behavioural modification, including Antipsychotics have been used in attempts to manage a vari-
removing reinforcement during the undesired behaviour ety of challenging behaviours within the context of demen-
and increasing reinforcement of appropriate alternative tia, not just sexually disinhibited behaviour. These drugs
behaviours. Bardell et al. (2011) found that consistently exert their anti-libidinal effect through dopamine receptor
applied distraction techniques were effective in reducing blockade (Black et al., 2005) but have been the subject of sig-
such behaviour. Other more practical strategies such as nificant debate given their potential and serious side effects
the use of same-sex caregivers if appropriate, or the use of including cerebrovascular risk (O’Brien, 2008). Macknight
clothing specially designed to prevent inappropriate disrob- and Rojas-Fernandez (2000) report the beneficial use of
ing (e.g. with buttons at the back) may have a value and offer quetiapine in an 85-year-old man with dementia and par-
simple yet effective means to address such difficulties. The kinsonism who presented with excessive masturbation ‘to
utility of these approaches such as these has not been fully the point of self-injury’. The patient had previously been
established and most evidence exists in the form of indi- tried on cyproterone acetate (CPA) and paroxetine with no
vidual or multiple case reports. success. Within 2 days of the quetiapine being commenced,
Finally, before any consideration is given to additional the sexual behaviours had improved with no worsening of
medication that may be directed towards target symptoms, his parkinsonism. Case reports with positive outcomes have
common sense suggests it would be advisable to consider also been seen with haloperidol, zuclopenthixol, olanzapine
the removal of any medications, which could be causing or and risperidone (Tucker, 2010; Bardell et al., 2011). Reeves
at least exacerbating the problem. Drugs such as benzodiaz- and Perry (2013) describe how a 61-year-old male with fron-
epines and dopamine agonists would fall into this category totemporal dementia and excessive sexual vocalizations was
(Joller et al., 2013). successfully treated with aripiprazole after a variety of psy-
chotropics either failed or were not tolerated.
27.4.2 PHARMACOLOGICAL APPROACHES
27.4.2.3 Anti-androgens
Despite the most comprehensive assessment and detailed
behavioural management strategies, some sexually disin- These agents can be further subdivided into hormonal and
hibited behaviour can continue leading to the prescription non-hormonal groups. They both act to decrease serum
of medication. Currently, no medication has been licensed testosterone levels with the aim of reducing sexual drive,
either in the United Kingdom (Series and Dégano, 2005) or although the extent to which serum testosterone corre-
in the United States (Kettl, 2008) for this specific purpose. lates with hypersexual behaviour is not clear (Levitsky and
While there have been no double-blind, placebo-controlled Owens, 1999). The issue of using medication to suppress the
trials for any of the drugs used to combat such symptoms, libido in a dementia patient is a complex and an ethical issue.
their use has been reported in the literature and again exists CPA is a progestogen and exerts its effect by both inhib-
mainly in the form of case reports or series and retrospec- iting testosterone release and blocking androgen receptors.
tive case control series. A summary is outlined below. Haussermann et al. (2003) describe the successful reduction
in ISB following treatment with CPA. Huertas et al. (2007)
27.4.2.1 Antidepressants conducted a study comparing CPA against haloperidol
admittedly looking at non-sexual aggressive behaviours.
Antidepressants are among the commonly prescribed Twenty-four patients completed the study that revealed a
psychotropic medications. Their relatively favourable significant reduction in such behaviours favouring the CPA.
side effect profile makes them ideal first-line medications Medroxypreogesterone acetate (MPA) acts in a simi-
for addressing ISB (Cross et al., 2013). Selective serotonin lar manner to CPA, although it is less potent in its mode
reuptake inhibitors (SSRIs) such as citalopram or parox- of action (Guay, 2008). Cross et al. (2013) reported on
etine are thought to act by not only their negative effect a series of 10 individual cases of elderly males with
on the libido but also via their anti-obsessional properties dementia and exhibiting ‘inappropriate hypersexuality’.
Sexuality and dementia 289

Positive outcomes in terms of a reduction in unwanted is being used by old age psychiatrists for this purpose (Ward
behaviour were found in 7 out of the 10. This was after and Manchip, 2013).
antidepressant and antipsychotic trials failed to achieve
improvements in 7 and 6 of the group, respectively. 27.4.2.5 Other medications
Luteinizing hormone releasing hormone (LHRH) ana-
logues such as leuprorelin, triptorelin and goserelin inhibit Finally, in addition to those described above, there are case
gonadotrophin release suppressing ovarian and testicular reports indicating beneficial effects of other medications
hormone production and resulting in a “chemical castration” including carbamazepine (Freymann et al., 2005), gabapen-
(Guay, 2008). Amadeo (1996) found beneficial effects of leu- tin (Alkhalil et al., 2003), buspirone (Tiller et al., 1998) and
prorelin in three subjects with dementia exhibiting sexual pindolol (Jensen, 1989). The sparse nature of the published
disinhibition. Ott (1995) also reports the successful treatment evidence reflects the uncertainty that remains on which
of a demented patient with Klüver–Bucy syndrome. pharmacological strategies are the most effective. For a com-
Oestrogens such as diethylstilbestrol (DES) have also prehensive review of the medications used in the treatment of
been used in this context. They act by reducing lutein- ISB, see Tucker (2010) and Ward and Manchip (2013).
izing hormone (LH) and follicle stimulating hormone
(FSH), which, in turn, leads to a reduction in testoster- 27.4.2.6 Ethical issues of drug management
one production (Black et al., 2005). Lothstein et al. (1997)
reported marked improvements in behaviour in 38 out of 39 There are complex ethical considerations when drugs are
demented subjects who were exhibiting sexually inappro- used to combat sexually inappropriate behaviour in patients
priate behaviour, when treated with DES. who are unable to provide informed consent. Central to this
Care must be taken to consider the side effect profile of issue is the concept of human rights both those belonging to
these medications, which can include elevated thromboem- the perpetrator and those on the receiving end of such behav-
bolic risk, fluid retention, osteopaenia and bone pain. Cross iour. Any medication decision must be made in the light of an
et al. (2013) suggest that MPA should be considered before assessment of risk to all parties and involving all relevant indi-
oestrogen and LHRH analogues due to its more favourable viduals. This decision should form part of a multidisciplinary
level of adverse effects and ease of administration/dosages, etc. care plan that should be reviewed at regular individuals.
Other drugs with anti-androgenic properties such as
cimetidine (a histamine H2 blocker), ketoconazole (an anti-
fungal) and spironolactone (a potassium-sparing diuretic)
have also been studied. Wiseman et al. (2000) reported on 27.5 CONCLUSIONS
20 demented patients with sexually inappropriate behav-
iour. Fourteen of the 20 patients responded favourably Clearly, the impact of dementia upon the sexuality of an
to the cimetidine while the remaining six responded to individual has profound implications for the patient, their
combinations of cimetidine and ketoconazole, spironolac- partner and all those involved in caring for them. When
tone or both. Na et al. (2009) described how finasteride, a difficulties do occur, they can be significant and a compre-
5α-reductase inhibitor commonly used for treating benign hensive assessment of all the relevant factors needs to be con-
prostatic hyperplasia, successfully extinguished ISB in 6 out sidered. If specific management is required, then this needs
of 11 men with vascular dementia. to take account of sometimes complex ethical issues such as
capacity, consent and basic human rights. Professionals of
27.4.2.4 Cognitive enhancers all disciplines need to be alert to these problems and offer
support and help when necessary.
If the dementia pathology lies behind the aetiology of ISB, it
might be reasonable to predict that drugs shown to be effec- REFERENCES
tive in treating cognitive impairment such as cholinesterase
inhibitors or memantine may also have an important role in Alagiakrishnan, K., Lim, D., Brahim, A. et al. (2005).
managing these behaviours. At present, however, the evidence Sexually inappropriate behaviour in demented elderly
remains conflicting. While Canevelli et al. (2013) report the people. Postgraduate Medical Journal, 81: 463–466.
improvement of sexually inappropriate behaviour in an elderly Alkhalil, C., Hahar, N., Alkhalil, B. et al. (2003). Can
dementia patient given rivastigmine, Freymann et al. (2005) gabapentin be a safe alternative to hormonal therapy
found that such behaviour was made worse by galantamine. in the treatment of inappropriate sexual behavior
Case reports have also been published showing how donepezil in demented patients? International Urology and
led to ISB (Chemali, 2003; Lo Coco and Cannizzaro, 2010). Nephrology, 35: 299–302.
While memantine has been shown to be an effective Allen, R.S., Petro, K.N. and Phillips, L.L. (2009). Factors
treatment for agitation in the context of dementia, so far influencing young adults’ attitudes and knowledge of
no evidence has been published showing its efficacy specifi- late-life sexuality among older women. Ageing and
cally in ISB. Despite this, there is evidence that memantine Mental Health, 13: 238–245.
290 Dementia

Amadeo, M. (1996). Antiandrogen treatment of aggressiv- patients and spouses. Sexuality and Disability,
ity in men suffering from dementia. Journal of Geriatric 28: 195–203.
Psychiatry and Neurology, 9: 142–145. Eloniemi-Sulkava, U., Notkola, I.L., Hämäläinen, K. et al.
Bardell, A., Lau, T. and Federoff, J.P. (2011). Inappropriate (2002). Spouse caregivers’ perceptions of influence of
sexual behaviour in a geriatric population. International dementia on marriage. International Psychogeriatrics,
Psychogeriatrics, 23: 1182–1188. 14: 47–58.
Bauer, M., Nay, R. and McAuliffe, L. (2009). Catering to Freymann, N., Michael, R., Dodel, R. and Jessen, F. (2005).
love, sex and intimacy in residential aged care: What Successful treatment of sexual disinhibition in
information is provided to consumers? Sexuality and dementia with carbamazepine – A case report.
Disability, 27: 3–9. Pharmacopsychiatry, 38: 144–145.
Beckman, N., Waern, M., Gustafson, D. and Skoog, I. (2008). Ginsberg, T.B., Pomerantz, S.C. and Kramer-Feeley,
Secular trends in self reported sexual activity and V. (2005). Sexuality in older adults: Behaviours and
satisfaction in Swedish 70 year olds: Cross sectional ­preferences. Age and Ageing, 34: 475–480.
survey of four populations, 1971–2001. British Medical Guay, D.R.P. (2008). Inappropriate sexual behaviours
Journal, 337: 151–154. in cognitively impaired older individuals. American
Black, B., Muralee, S. and Tampi, R.R. (2005). Journal of Geriatric Pharmacology, 6: 269–288.
Inappropriate sexual behaviours in dementia. Journal Haussermann, P., Goecker, D., Beier, K. et al. (2003).
of Geriatric Psychiatry and Neurology, 18: 155–162. Low-dose cyproterone acetate treatment of sexual
Bouman, W.P., Arcelus, J. and Benbow, S.M. (2006). acting out in men with dementia. International
Nottingham study of sexuality and ageing (NoSSA Psychogeriatrics, 15: 181–186.
I). Attitudes regarding sexuality and older people: Howell, T. and Watts, D.T. (1990). Behavioral complications
A review of the literature. Sexual and Relationship of dementia: A clinical approach for the general inter-
Therapy, 21: 149–161. nist. Journal of General Internal Medicine, 5: 431–437.
Burgess, A.W. and Phillips, S.L. (2006). Sexual abuse, Huertas, D., López-Ibor Aliño, J.J., Molina, J.D. et al.
trauma and dementia in the elderly: A retrospec- (2007). Anitiaggressive effect of cyproterone versus
tive study of 284 cases. Victims and Offenders, haloperidol in Alzheimer’s disease: A randomized
1 (2): 193–204. double-blind pilot study. Journal of Clinical Psychiatry,
Burns, A., Jacoby, R. and Levy, R. (1990). Psychiatric 68: 439–444.
phenomena in Alzheimer’s disease IV: Disorders of Hyde, Z., Flicker, L., Hankey, G.J. et al. (2010). Prevalence
behaviour. British Journal of Psychiatry, 157: 86–94. of sexual activity and associated factors in men
Canevelli, M., Talarico, G., Tosto, G. et al. (2013). aged 75 to 95 years. Annals of Internal Medicine,
Rivastigmine in the treatment of hypersexuality in 153: 693–702.
Alzheimer disease. Alzheimer Disease and Associated Jensen, C.F. (1989). Hypersexual agitation in Alzheimer’s
Disorders, 27: 287–288. disease. Journal of the American Geriatrics Society,
Chemali, Z. (2003). Donepezil and hypersexuality: A report 37: 917.
of two cases. Primary Psychiatry, 10 (7): 78–79. Joller, P., Gupta, N., Seitz, D.P. et al. (2013). Approach to
Cross, B.S., De Young, G.R. and Furmaga, K.M. (2013). inappropriate sexual behaviour in people with demen-
High-dose medroxyprogesterone for inappropriate tia. Canadian Family Physician, 59: 255–260.
hypersexuality in elderly men with dementia: A case Kaiser, F.E. (1996). Sexuality in the elderly. Urologic Clinics
series. Annals of Pharmacotherapy, 47 (1): e1. of North America, 23: 99–109.
Davies, H.D., Zeiss, A.M., Shea, E.A. and Tinklenberg, Kessel, B. (2001). Sexuality and the older person. Age and
J.R. (1998). Sexuality and intimacy and Alzheimer’s Ageing, 30: 121–124.
patients and their partners. Sexuality and Disability, Kettl, P. (2008). Inappropriate behavior in long-term care.
16: 193–203. Annals of Long Term Care, 16 (12): 29–35.
Derousesné, C., Guigot, J., Chermat, V. et al. (1996). Knight, C., Alderman, N., Johnson, C. et al. (2008). The
Sexual behavioural changes in Alzheimer disease. St Andrews sexual behaviour assessment (SASBA):
Alzheimer Disease and Associated Disorders, Development of a standardised recording instrument
10: 86–92. for the measurement and assessment of challeng-
Devanand, D.P., Brockington, C.D., Moody, B.J. et al. ing sexual behaviour in people with progressive and
(1992). Behavioural syndromes in Alzheimer’s disease. acquired neurological impairment. Neuropsychological
International Psychogeriatrics, 4 (Suppl. 2): 161–184. Rehabilitation, 18: 129–159.
Doll, G.M. (2013). Sexuality in nursing homes: Practice Leo, R.J. and Kim, K.Y. (1995). Clomipramine treatment of
and policy. Journal of Gerontological Nursing, paraphilias in elderly demented patients. Journal of
39 (7): 30–37. Geriatric Psychiatry and Neurology, 8: 123–124.
Dourado, M., Finnamore, C., Barrosa, M.F. et al. (2010). Levitsky, A.M. and Owens, N.J. (1999). Pharmacologic
Sexual satisfaction in dementia: Perspectives of treatment of hypersexuality and paraphilias in nursing
Sexuality and dementia 291

home residents. Journal of the American Geriatrics Reeves, R.R. and Perry, C.L. (2013). Aripiprazole for sexu-
Society, 47: 231–234. ally inappropriate vocalizations in frontotemporal
Lichtenberg, P.A. and Strzepek, D.M. (1990). Assessments dementia. Journal of Clinical Psychopharmacology, 33:
of institutionalized dementia patients’ competen- 145–146.
cies to participate in intimate relationships. The Schiavi, R.C., Schreiner-Engel, P., Mandeli, J. et al. (1990).
Gerontologist, 30: 117–120. Healthy aging and male sexual function. American
Lindau, S.T., Schumm, L.P., Laumann, E.O. et al. (2007). Journal of Psychiatry, 147: 766–771.
A study of sexuality and health among older adults in Series, H. and Dégano, P. (2005). Hypersexuality in
the United States. New England Journal of Medicine, dementia. Advances in Psychiatric Treatment, 11:
357: 762–774. 424–431.
Lo Coco, D. and Cannizzaro, E. (2010). Inapprorpiate Stimmel, G.L. and Gutierrez, M.A. (2006). Sexual dysfunc-
sexual behaviors associated with donepezil treatment: tion and psychotropic medications. CNS Spectrums,
A case report. Journal of Clinical Psychopharmacology, 11: 24–30.
30: 221–222. Szasz, G. (1983). Sexual incidents in an extended care
Lothstein, L.M., Fogg-Waberski, J. and Reynolds, P. (1997). unit for aged men. Journal of the American Geriatrics
Risk management and treatment of sexual disinhi- Society, 31: 407–411.
bition in geriatric patients. Connecticut Medicine, Tiller, J.W.G., Dakis, J.A. and Shaw, J.M. (1998). Short-term
61: 609–618. buspirone treatment in disinhibition with dementia.
Macknight, C. and Rojas-Fernandez, C. (2000). Quetiapine Lancet, 2: 510.
for sexually inappropriate behavior in dementia. Tsai, S.-J., Hwang, J.-P., Yang, C.-H. et al. (1999).
Journal of the American Geriatrics Society, 48: 707. Inappropriate sexual behaviours in dementia: A pre-
Mahieu, L. and Gastmans, C. (2011). Sexuality in insti- liminary report. Alzheimer Disease and Associated
tutionalized elderly persons: A systematic review Disorders, 13: 60–62.
of argument-based ethics literature. International Tucker, I. (2010). Management of inappropriate sex-
Psychogeriatrics, 24: 346–357. ual behaviors in dementia: A literature review.
Na, H.R., Lee, J.W., Park, S.M. et al. (2009). Inappropriate International Psychogeriatrics, 22: 683–692.
sexual behaviours in patients with vascular demen- Ward, R.F. and Manchip, S. (2013). ‘Inappropriate’
tia: Possible response to finasteride. Journal of the sexual behaviours in dementia. Reviews in Clinical
American Geriatrics Society, 57: 2161–2162. Gerontology, 23: 75–87.
O’Brien, J. (2008). Antipsychotics for people with demen- Wiseman, S.V., McAuley, J.W., Freidenberg, G.R. et al.
tia. British Medical Journal, 337: 64–65. (2000). Hypersexuality in patients with dementia:
Ott, B.R. (1995). Leuprolide treatment of sexual Possible response to cimetidine. Neurology, 54: 2024.
­aggression in a patient with dementia and the Wright, L.K. (1991). The impact of Alzheimer’s disease
­Kluver-Bucy syndrome. Clinical Neuropharmacology, on the marital relationship. The Gerontologist, 31:
18: 443–447. 224–237.
Pontone, G., Williams, J.R., Spear Bassett, S. and Marsh, Zeiss, A.M., Davies, H.D. and Tinklenberg, J.R. (1996).
L. (2006). Clinical features associated with impulse An observational study of sexual behaviour in
control disorders in Parkinson disease. Neurology, demented male patients. Journals of Gerontology, 51:
67: 1258–1261. M325–M329.
28
Residential care for people with dementia

DAVID CONN, JOHN SNOWDON AND NITIN PURANDARE

in line with observed differences in utilization. This varia-


28.1 A GLOBAL PERSPECTIVE ON tion reflects differences in care needs, in the structure and
RESIDENTIAL CARE PROVISION comprehensiveness of formal LTC systems, as well as in
family roles and caring cultures. Most of the cost of LTC
Availability of residential long-term care (LTC) facilities for resides within the institutional sector with the exception
people with dementia and physical disabilities varies from of four OECD countries (Denmark, Austria, New Zealand
country to country and within countries. In many areas of and Poland), where expenditure of home care exceeds that
the world, relatively little funding is allocated to the LTC of institutional care.
sector. Even between developed nations there is considerable Worldwide, the population of older people, especially the
variation in the types, organization, funding and policies of very old, is expected to rise considerably. There is a steep
residential facilities. Hence, caution needs to be exercised rise in the prevalence of dementia in late old age. In Japan,
when considering generalizations about how residential a doubling of the proportion of people aged over 80 years,
care is or should be provided for people with dementia. from 2.8% to 5.7%, was expected to occur between 1993 and
Ribbe et al. (1997) noted that there are no universally 2010, with a further rise to 9.7% by 2025. Substantial rises
accepted definitions for the different LTC services. They in the proportion aged over 80 years are occurring in many
compared provision of residential care in 10 developed countries (Ribbe et al., 1997). The fact that the worldwide
countries based on an understanding that nursing homes number of persons with dementia is anticipated to increase
provide nursing care 24 hours per day, while residential from 24.3 million in 2001 to over 81.1 million by 2040 (Ferri
homes provide personal care and social involvement for et al., 2005) has startling implications regarding funding,
people unable to manage adequately at home, but who need policies and planning of LTC for people with dementia. In
no more nursing than visiting nurses can offer. They found the United Kingdom, the demand for LTC beds is expected
that, in spite of having similar age distributions, the propor- to rise from 450,000 in 2000 to 1.1 million in 2051, with
tion of those aged 65 years or more in the Netherlands who quadrupling of the annual LTC cost from £12.9 billion to
were in nursing homes was half that found in the United £53.9 billion (Wittenberg et al., 2004).
States, but the proportion in residential homes was four
times higher. In France, nobody was in a nursing home but
4% of the elderly were in residential homes. In 2004, of those
28.2 PREVALENCE OF DEMENTIA IN
over 65 years in the United States, 3.63% were in nursing
homes, in a total nursing home resident population of 1.49
LTC FACILITIES
million (National Nursing Home Survey, 2004).
The most reliable comparative international data on LTC Researchers in diverse countries have reported varying rates
come from the Organization for Economic Co-operation of dementia in LTC facilities. A recent review of 30 studies,
and Development (OECD, 2011). On average, LTC expen- dating back to 1986, noted a median prevalence of 58.6%
diture (including homecare) accounts for 1.5% of gross (Seitz et al., 2010). A large UK study found that 62% of LTC
domestic product (GDP) across the OECD. The LTC work- residents had dementia (Matthews and Dening, 2002). Recent
force (mostly women, often working part-time) is about nursing home studies have shown rates of 69.5% in Helsinki
1.3% of the total OECD workforce. There is significant (Hosia-Randell and Pitkälä, 2005), 78% in Sydney (Brodaty
cross-country variation in the resources allocated to LTC et al., 2001) and 80.5% in Norway (Selbaek et al., 2007).

292
Residential care for people with dementia 293

The proportions with Alzheimer’s disease (AD), vascular dementia care in LTC facilities – though it can help. Factors
dementia (VaD), mixed (AD + VaD), dementia with Lewy that can contribute to good care will be discussed later in
bodies (DLB), frontotemporal dementia (FTD), alcoholic, this chapter.
traumatic or other types of dementia have not usually been
reported. Cases of young-onset dementia (including FTD
and human immunodeficiency virus [HIV]) are relatively 28.3 BEHAVIOURAL AND
uncommon in nursing homes but may well require special
PSYCHOLOGICAL SYMPTOMS OF
attention because of a greater incidence of behavioural prob-
lems or the difficulty of providing age-appropriate activities
DEMENTIA IN LTC FACILITIES
and involvement.
The prevalence of dementia in residential homes has gen- The prevalence of behavioural and psychological symptoms
erally been reported as lower than that in nursing homes, of dementia (BPSD) in nursing homes varies between 24%
though in Maryland (United States) the difference was and 93% in the United States (Beck and Shue, 1994; Gruber-
small. There, 67.7% of assisted living residents (74% response Baldini et al., 2004). Selbaek et al. (2007) reported that 65.2%
rate) fulfilled criteria for dementia and an additional 6.6% of residents in Norwegian nursing homes exhibited one or
had clinically significant cognitive dysfunction (Rosenblatt more clinically significant behavioural symptoms and that
et al., 2004). Small facilities had higher percentages with frequency of delusions, hallucinations, aggression/­agitation,
dementia (81% versus 63%). Some 83% of those with demen- apathy, disinhibition and aberrant motor behaviour
tia had exhibited neuropsychiatric symptoms in the pre- increased with progression of dementia. An English study
vious month. Residents with dementia required twice the reported that 79% of residents with dementia in both nurs-
amount of staff time given to those without dementia. ing and residential homes had clinically significant BPSD
A report commissioned by Alzheimer’s Australia illus- (Margallo-Lana et al., 2001).
trates the huge economic impact of dementia and its poten- Brodaty et al. (2001) reported that 80% of nursing home
tial workforce demands (Access Economics, 2009). The cost residents showed behavioural disturbances with persons
implications of the projected increase in need for LTC of who did not have dementia being rated nearly as highly on
people with dementia are considerable. Of 230,000 people behavioural disturbance scales as those with dementia. This
with dementia in a population of 21 million in Australia in raises questions about the relative importance of organic
2008, 90,200 were living in residential care facilities, cared and environmental factors in development of BPSD. Brodaty
for by 78,000 full-time equivalent (FTE) staff. There were et al. (2001) found a three-fold difference in the prevalence
0.9 and 0.8 FTEs per high care (nursing home) and low care of different BPSD across nursing homes, but size accounted
(residential home) resident, respectively. Modelling in the for only 3% of the variance. The extent of the difference is
report suggested that, in the absence of changes of policy or difficult to explain. Some nursing homes may be reluctant to
other arrangements, there will be a shortage of 71,000 FTEs accept ‘difficult’ referrals. The study did find greater func-
within two decades. There is already difficulty in recruiting tional incapacity to be associated with increased rates of
staff skilled in dementia care, partly attributable to unat- behavioural disturbance, including aggressiveness.
tractive rates of pay, and this has obvious impact on the In Assisted Living facilities, 49% of residents with demen-
quality of care provided in LTC facilities. tia were observed to exhibit one or more behavioural symp-
The quality of care in U.S. nursing homes in the last three toms at least once weekly, 19% showing aggressive and 34%
decades has been described as poor, largely attributable to physically non-aggressive behaviour (Gruber-Baldini et al.,
inadequate staffing and a poor mix of skills (Harrington, 2004). Rates were higher in facilities with fewer residents.
2001). This is in spite of legislation (the Omnibus Budget Reliable and valid measurement of BPSD is of great
Reconciliation Act, 1987; OBRA-87) brought in to ensure importance in both clinical practice and research. A recent
higher standards of care and the development of best prac- review described 83 different instruments that have been
tice guidelines. The government was said to be reluctant to utilized to assess and track BPSD (van der Linde et al., 2014).
impose higher standards for staffing because of concerns Examples include the Neuropsychiatric Inventory (NPI),
over cost. Furthermore, Harrington (2001) commented the Cohen-Mansfield Agitation Inventory (CMAI) and the
that fraud and financial mismanagement were widespread Behavioral Pathology in Alzheimer’s Disease Rating Scale
throughout the nursing home industry. (BEHAVE-AD) (Reisberg et al., 1987; Cohen-Mansfield
To provide good care in such facilities, ‘one must battle et al., 1989; Cummings et al., 1994). Tracking of behaviours
against nihilism, cynicism and resistance to change which utilizing basic charting approaches, such as documenta-
is often present in geriatric institutions’ (Conn, 2007). tion of antecedent–behaviour–consequences (ABC) can
OBRA-87 mandated formal psychiatric assessments of nur­ also be very useful for identifying triggers and designing
sing home residents suspected of having mental disorders interventions.
but it was observed that this further entrenched a pattern Brodaty et al. (2001) noted positive associations between
of relative neglect of patients with dementia in formal men- ratings of behavioural disturbance and psychosis and
tal health services planning (Borson et al., 1997). It is evi- depression. Wancata et al. (2003) reported that 38% of newly
dent that money alone cannot ensure a high standard of admitted residents with dementia had marked or severe
294 Dementia

non-cognitive symptoms of dementia, including 30% with with those of dementia. Nursing home resident scores on
depressive symptoms and 12.7% with ‘aggressive-psychotic the Cornell Scale and on the Neuropsychiatric Inventory
symptoms’; one-third remitted from these symptoms within (Cummings et al., 1994) correlated highly (r = 0.7) (Barca
6 months. Menon et al. (2001) analysed data from residents et al., 2009). Apathy (which needs to be distinguished from
with dementia newly admitted to Maryland nursing homes. depression) is common early in AD and becomes more
Nursing staff observed physical aggressive behaviours in severe as cognitive impairment worsens.
10.5% and verbal aggressive behaviours in 10.4%. The rates Examination of the factors associated with depression,
varied with degree of cognitive impairment, with physical agitation and other behavioural and mental disorders in
aggression displayed by 15% of those with severe demen- LTC facilities is relevant to discussions concerning optimal
tia and 2.4% with mild dementia, and verbal aggression by management, interventions and therapy for these disorders.
13% and 5.4%, respectively. They found that those manifest- For example, depression among residents with dementia has
ing physical or verbal aggression (mainly those with more been reported as more common in for-profit than not-for-
severe cognitive impairment) were more likely to be rated profit nursing homes (Gruber-Baldini et al., 2005). To what
as depressed. extent are depressive symptoms a result of demoralizing
Studies have found the prevalence of depression in LTC situations and circumstances? To what extent is disruptive
facilities to be much higher than that reported among behaviour attributable to neurobiochemical or structural
older people living at home. Depression levels were some- brain changes, maybe interacting with environmental fac-
what higher in nursing homes than in residential homes tors? Outcome studies exploring whether medications, psy-
(Snowdon and Fleming, 2008). Of residents newly accom- chological therapies or environmental adjustments appear
modated in Maryland nursing homes (assessed 21–65 days to be related to reduction in symptoms will provide clues.
after admission, thus minimizing the effect of relocation The finding that, in a 1-year study of residents with demen-
stress on results), 23.6% of demented and 22.3% of non- tia in LTC facilities, 45% of agitation cases, nearly 60%
demented subjects were recorded as depressed (Kaup et al., with delusions and over 60% of depression cases resolved
2007). Among those with dementia, depression in residen- independently of pharmacological treatment (Ballard et al.,
tial homes was not significantly less prevalent than in nurs- 2001) should provoke consideration of what factors specific
ing homes (Gruber-Baldini et al., 2005). Most residents with to LTC situations contribute to development of these condi-
anxiety symptoms are also depressed (Parmelee et al., 1993). tions. This also highlights the self-limiting course of certain
Evers et al. (2002) drew attention to the high prevalence of BPSD, and therefore the propriety of regular reviews of med-
major depression among residents in the last 6 months of ication, with attempts to cease unnecessary prescriptions.
life, both those with dementia and those without. The fact that, in an LTC facility in California for people
Among nursing home residents, the evidence regard- with cognitive impairment, 40 residents scored >12 on the
ing an association of depression with cognitive impair- Cornell Scale soon after admission, but that only 6 of them
ment is unclear. Some have observed no association (Katz were still depressed after 6 months (in contrast to worse out-
and Parmelee, 1994). Others have reported depression to be comes elsewhere), raises questions about factors that main-
more prevalent among those with mild-to-moderate demen- tain depressive symptoms (Payne et al., 2002). Commenting
tia (Evers et al., 2002; Jones et al., 2003). In contrast, Teresi on a decrease in depressive symptoms from 41.3% to 28.9%
et al. (2001) diagnosed major depression in 5.8% of nursing during the first 6 months of admission to Dutch nursing
home residents with little or no cognitive impairment, but in homes, Smalbrugge et al. (2006) suggested that adaptation
22.8% of those with moderate-to-severe dementia. Gruber- to pre-admission factors, facilitated by the nursing home
Baldini et al. (2005), examining scores on the Cornell environment, may explain the fall. When the prevalence
Scale for Depression in Dementia (Alexopoulos et al., 1988) does not fall (as in the study by Scocco et al., 2006), reasons
among residents of nursing homes and residential homes, should be sought.
found that participants with depression were more likely to
be severely or very severely cognitively impaired. The asso-
ciation of behavioural symptoms with Cornell Scale scores 28.4 RECOGNITION OF DEPRESSION
was significant, though it was much stronger among those
IN CASES OF DEMENTIA IN LTC
with dementia in residential homes. Barca et al. (2008), who
conducted a factor analysis of ratings on the Cornell, noted
FACILITIES
that scores on the mood sub-scale did not differ across var-
ied levels of dementia severity, but on the other four sub- Cohen et al. (2003) have commented on the evidence (e.g.
scales increases were associated with severity of dementia. Bagley et al., 2000) that depression in nursing homes is
In an earlier factor analysis (Kurlowicz et al., 2002), a clear frequently overlooked and undertreated. They found that
separation had been noted between somatic/vegetative despite using the Minimum Data Set (MDS) in U.S. nurs-
features and a ‘core depression factor’, raising questions ing homes, 75% of major depressive disorders and 87% of
about the scale’s validity in a population with high levels of all types of depression escaped recognition; further, only
dementia, medical illness and/or functional disability. Some 33% of those identified as depressed were treated with anti-
symptoms of depression as rated by the Cornell overlap depressants. These authors showed that use of the Cornell
Residential care for people with dementia 295

Scale resulted in a significant increase in the percentage of the likelihood of staff initiating or calling for interventions
residents given antidepressants. Before screening, only 16% when residents were rated as depressed. File notes showed
of patients who scored 5 or more on the Cornell were pre- that less than one-third of the residents with high scores
scribed antidepressants, whereas after routine screening were monitored for evidence of depression by staff following
was introduced 36% were given antidepressants. They rec- the assessment. Only 18% of residents with high scores were
ommended mandatory screening and suggested that refer- referred for further assessment of depression, while 10%
rals to staff psychiatrists of all those scoring 5 or more on received a treatment change (Davison et al., 2012).
the Cornell would not impose an undue burden on them.
The American Geriatrics Society and American Asso­
ciation of Geriatric Psychiatry (2003a), in a consensus
statement, recommended that residents should be screened 28.5 MANAGEMENT OF DEMENTIA IN
for depressive symptoms within the first 2–4 weeks of LTC FACILITIES
admission to an LTC facility, and subsequently at least
every 6 months. Use of the Cornell Scale was supported
Prevention and management of dementia, BPSD and dep­
for residents with moderate or severe dementia, and of
ression in cases of dementia are discussed in chapters 7, 8,
the Geriatric Depression Scale (Yesavage et al., 1983) for
23, 24, 25 and 53. Aspects specific to care in LTC facilities
residents with no more than mild-to-moderate cognitive
will be discussed here.
impairment. However, limitations should be recognized as
Little will be said about short-term residential care
discussed in Section 28.3. Somatic items comprise 37% of
(respite) because of the dearth of relevant data and studies.
the Cornell. The prevalence of depression may be overes-
It has proved effective in reducing the subjective burden and
timated in patients with physical problems. Kurlowicz et
depression experienced by family caregivers of people with
al. (2002) called for research to establish whether an intra-
dementia (Donath et al., 2009). However, studies around the
psychic sub-scale of the Cornell may be more useful than
world have shown low utilization of such care, even when
the global scale for measuring depression among those
caregivers were well aware of its availability.
with dementia, physical illness and/or disability. Greenberg
et al. (2004) found that in palliative care cases the psychia-
trist’s diagnostic assessments did not correspond well with 28.5.1 PHYSICAL AND SOCIAL
Cornell Scale scores and concluded that there was no scale ENVIRONMENT AND STAFF
valid and reliable enough to effectively ascertain depres- TRAINING
sion in the most severely demented patients. A short form
of the Geriatric Depression Scale (GDS-15), validated by First it is appropriate to re-emphasize the importance of
Herrmann et al. (1996), has been found useful in screening environment and of care arrangements in relation to pre-
for depression among residents with only mild-to-moderate vention or development of mood and behaviour distur-
cognitive impairment (Gerety et al., 1994), and in such cases bance. Guidelines are available (e.g. American Geriatrics
correlates more closely than does the Cornell with psychia- Society and American Association for Geriatric Psychiatry,
trist assessments on other depression rating scales, such as 2003b; Conn and Gibson, 2007) to advise on how, and in
the Montgomery Åsberg Depression Rating Scale (MADRS) what settings, optimal care can be provided. Koopmans
(Montgomery and Åsberg, 1979; Snowdon and Fleming, (2005) recommends accommodating residents at similar
2008). Even if such scales are not valid in all cases, use of stages of dementia together, but notes that a drawback is
the GDS-15 and/or Cornell, as appropriate, in LTC facili- the need to move them when their dementia progresses.
ties, is strongly recommended, with the intention that, if Others advocate ageing in place. There is also an argument
pointers to depression are identified, staff and doctors will for housing people with early-onset dementia together. The
be prompted to discuss possible causation and appropriate major need surely is to look at the needs of the individual
interventions. and what suits that person best.
The challenge of preventing and identifying depression in As stated earlier, funding varies between jurisdictions,
this population has been highlighted by several recent stud- both in relative amount and how it is allocated. However,
ies. A Dutch study evaluated the effects of a 2-year stepped Conn and Gibson (2007) pointed out that countries that
care programme designed to prevent the onset of major have residential care facilities for older people commonly
depression. After 2 years, the incidence of depression was not face challenges that include ‘inadequate staffing levels, lack
significantly lower in the intervention group than in the con- of staff training regarding mental health issues, aging and
trol group although there was some evidence that completers poorly designed LTC homes, failure to identify and assess
had benefitted. Of note, the drop-out rate was very high at residents in a timely fashion, inappropriate use of psycho-
44% (van Schaik et al., 2014). In an Australian study of six tropic medications and limited availability of mental health
sessions of staff training to improve recognition of depres- consultants’.
sion, the trained group performed no better than controls Management of dementia in LTC settings is optimized
(McCabe et al. 2013). In another study, use of the Cornell by having available a multidisciplinary team that can
Scale for depression in dementia had very limited impact on coordinate care. Facility-based staff members may take
296 Dementia

on mental health interest roles, such as recognition and The Green House model of care was developed by Dr
coordination of management of depression, and may be William Thomas, who had introduced the Eden Alternative
designated to have a role liaising with doctors and visit- in the 1990s. This model aimed to reduce loneliness, bore-
ing mental health professionals. Some LTC facilities make dom and helplessness among nursing home residents. To
arrangements for a consultation-liaison old age mental enhance quality of life, nursing home staff were encouraged
health service to be accessible to work with facility staff and to integrate plants, animals and children into the daily life
primary care doctors in dealing with the range of psychi- of residents.
atric and behavioural problems associated with dementia. A review of the literature on the impact of environmen-
An Australian model for dealing with challenging behav- tal design for LTC concluded that designers may confidently
iours blended clinical consultation with staff training and use unobtrusive safety measures; vary ambience, size and
educational interventions (Turner and Snowdon, 2009). A shape of spaces; provide single rooms; maximize visual
UK study found that few LTC facilities had visits from psy- access and control levels of stimulation. There was less evi-
chologists or social workers, and only one-third received dence on the usefulness of signage, homelikeness, provision
visits from old age psychiatrists (Purandare et al., 2004). for engagement in ordinary activities, small size and the
Snowdon (2010) reviewed models of mental health service provision of outside space (Fleming and Purandare, 2010).
delivery to LTC homes concluding that prompt recognition Impetus towards developing the above ideas may have
of mental health problems among residents is required, and been accelerated by reports such as that from Sloane et
this is facilitated by availability of a team working within al. (1991). They found that the development of special-
the facility to deal with these problems. Provision of opti- ized dementia units (whether areas within nursing homes
mal mental health care in LTC settings is dependent on or entire facilities) apparently led to less use of physi-
adequate funding, availability of expertise and education, cal restraints but no reduction in use of pharmacological
positive and caring attitudes, recognition of needs and sup- restraint.
portive teamwork. However, a Cochrane review concluded that there are
Citing Canadian national guidelines relating to men- no identified RCTs investigating the effects of Special
tal health issues in LTC homes, Conn and Gibson (2007) Care Units on behavioural symptoms in dementia, and no
discussed organizational and systems issues. The first con- strong evidence of benefit from the available non-RCTs.
cerned LTC homes developing the physical and social envi- The authors concluded that it is probably more important to
ronment as a therapeutic milieu through the intentional implement best practices than to provide a specialized care
use of design principles. Verbeek et al. (2009) declared that environment (Lai, 2009).
traditional institutional care has been arranged according
to the medical model, with emphasis given to the treat-
ment of the underlying pathology. They contended that this
model has become outmoded. They said that a shift towards 28.5.2 PSYCHOTHERAPEUTIC
a psychosocial model has occurred, this approach being APPROACHES AND PERSON-
person centred and aimed at supporting the well-being and CENTRED CARE
remaining strengths of older people with dementia.
It has been recognized that a facility’s architectural and A person-centred approach argues that much of what is
physical environment influences dementia care, and studies described as ‘challenging behaviour’ is a rational response
have suggested that small, domestic-style environments are to an untenable situation and can be a response to unmet
beneficial for older people with dementia. Verbeek et al. (2009) physical or social needs (Downs et al., 2005). Cohen-
reviewed and compared 11 concepts, from various countries, Mansfield (2001) suggested an environmental vulner-
that have adopted a home-like philosophy of care in a small- ability model and a behavioural and learning model.
scale context. They referred to CADE (confused and disturbed Non-pharmacological treatments for behavioural distur-
elderly) units in Australia, the Cantou in France, Group Living bance, based on these models, are discussed in Chapters
and Small-scale Living in Sweden and the Netherlands and 23 and 24. Snoezelen and aroma therapy are examples of
Belgium, Green Houses in the United States and comparable strategies used by nursing home staff to reduce agitation.
concepts developed in Japan, Canada, the United Kingdom Chenoweth et al. (2009) have presented evidence of the
and United States. In all of the 11, residents are encouraged effectiveness of dementia-care mapping and person-centred
to participate in the household as far as possible, with activi- care in reducing agitation among residents of LTC facilities.
ties (such as cooking) being planned according to residents’ A recent review of non-pharmacological interventions for
wishes. Residents are stimulated, encouraged and supported, neuropsychiatric symptoms of dementia found that 16 of 40
with an emphasis on autonomy and choice. Care staff function included studies reported statistically significant results in
as part of the household, and thus need to have skills and atti- favour of the interventions (Seitz et al., 2012). These inter-
tudes that suit the philosophy. In Green Houses, staff (certified ventions included staff training, mental health consultation
nursing assistants) receive 120 hours of additional training for and treatment planning, exercise, recreational activities
an expanded universal role, that includes cooking, cleaning, and music therapy or other forms of sensory stimulation.
laundry, shopping and more (Ragsdale and McDougall, 2008). Many of the studies had methodological limitations that
Residential care for people with dementia 297

placed them at potential risk of bias. Most interventions availability of consultants to do this work. They suggested
required significant resources from services outside of LTC the possibility of psychiatric nurse practitioners being
or significant time commitments from LTC nursing staff for trained as depression care managers or masters-level mental
implementation. A study of the barriers to providing non- healthcare specialists, who could also provide staff training
pharmacological interventions concluded that awareness in management of ‘difficult’ patients and coordinate group
of this information provides a tool by which to tailor inter- therapy. Another model (Restore, Empower, Mobilize) for
ventions so as to anticipate or circumvent barriers, thereby brief individual psychotherapy to treat depression in long-
maximizing intervention delivery (Cohen-Mansfield et al., term care residents with mild-to-moderate dementia has
2012). been presented by Carpenter et al. (2002).
It has been stated that most agitated behaviours of people
with dementia are manifestations of unmet needs (Camp 28.5.3 PSYCHOTROPIC MEDICATIONS
et al., 2002). The goals of treatment should be to uncover and
address such needs; the person has been unable to fulfil them Use of antipsychotic medication to suppress behavioural
because of a combination of perceptual problems, commu- disruption has varied between countries and over time.
nication difficulties and an inability to manipulate the envi- Extensive comparisons will not be presented here. However,
ronment through appropriate channels (Camp et al., 2002). a major catalyst for discussions leading to OBRA-87 was
Cohen-Mansfield (2001) categorized non-­pharmacological concern about misuse of psychotropic medication in U.S.
interventions, but Cody et al. (2002) detailed some of the nursing homes. Introduction of federal antipsychotic drug
challenges to their use in nursing homes. They even sug- regulations led to a 36% reduction in prescriptions for
gested (and this was long after OBRA-87 was introduced) neuroleptics in Baltimore nursing homes over 6 months
that staff members often see the sedating effects of psycho- (Rovner et al., 1992), and other studies (cited by Hughes
tropic drugs as desirable – and might prefer their use to et al., 2000) showed comparable results. Data from various
resident activity. Attitudes and the quality of care in LTC countries showed that in the mid-1990s over 20% of resi-
facilities are largely determined by the leadership, example dents in Iceland, Italy and Sweden, but only 14.4% of U.S.
and training provided in those facilities, in addition to their nursing home residents, were taking antipsychotic drugs
design and philosophy. (Hughes et al., 2000).
There is good reason to examine factors that might be In Sydney nursing homes in 1993, 27.4% of residents were
responsible for demoralization among LTC residents who taking typical antipsychotic medication regularly, while 15
become depressed. Higher levels of mastery and greater years later, 28% were given antipsychotics regularly, 7.4%
satisfaction with available social support have been shown typical and 21.8% atypical (Snowdon et al., 2011). Two-
to have effects in lessening depression and in buffering the thirds of those given antipsychotics had dementia or cere-
adverse impact of disability (Jang et al., 2002). Blanchard bral disease and not schizophrenia (Snowdon et al., 2005).
et al. (2009) have discussed the processes of adjustment The percentage of residents regularly taking anxiolytics fell
needed to overcome the multiple losses of function, abili- progressively from 1993 to 2003 (8.6% to 4.1%) then rose
ties, roles and relationships that may occur in old age. They to 4.7%, and similarly there was a fall in regular hypnotic
referred to construction and consolidation of a new under- use (26.6% to 11.3% and then 11.1%) (Snowdon et al., 2006,
standing of oneself, with emergence of meaning to life, 2011). Use of antidepressants increased, but not as much as
goals and something to hope for. Losses in one domain are in U.S. nursing homes (Snowdon et al., 2006). The changes
compensated for through strengths in other domains. The have been attributed to increased availability of best prac-
relevance to LTC facility care is striking. tice information in Sydney. In the early 2000s in Norway,
Testad et al. (2014) provided a systematic review of evi- 21.8% of nursing home residents were taking antipsychotics,
dence for the value of psychosocial interventions for BPSD. 24.5% anxiolytics, 30.6% hypnotics and 39.5% antidepres-
There is good evidence that personalized pleasant activities sants (Barca et al., 2009). Antipsychotics were more com-
are effective in the treatment of agitation, and that reminis- monly prescribed for those with severe dementia, reflecting
cence therapy can improve mood. Van Bogaert et al. (2013) the fact that psychotic and aggressive symptoms increase
found that six to eight sessions of reminiscence therapy with severity of dementia (Selbaek et al., 2007). In Sweden,
reduced depressive symptoms in this population. A ran- 26.3% of residents of LTC facilities for older people were tak-
domized trial of humour therapy provided by professional ing antipsychotics, their use being higher among those with
clowns, supported by trained staff, has been carried out in aggressive, verbally disruptive and wandering behaviour
Australia with evidence of significant reductions in agita- (Lövheim et al., 2006). In Helsinki, in 2003, a remarkable
tion as well as improved mood (Low et al., 2014). 43.3% of residents with dementia and 41% of those with-
Bharucha et al. (2006) reviewed 18 studies of psycho- out were given antipsychotics, while 41.4% of those with
therapy provided in LTC settings. A majority showed ben- dementia and 51.9% of those without were given antide-
efits on instruments measuring depression, hopelessness, pressants (Hosia-Randell and Pitkala, 2005), and there was
self-esteem, perceived control and on other variables. They a high use of anxiolytics and hypnotics. In Dutch nursing
commented that a perception that psychotherapy is a ‘medi- homes, antipsychotic and antidepressant drugs were more
cal’ intervention is a serious obstacle because of the lack of often prescribed for residents with severe dementia, while
298 Dementia

anxiolytics and sedatives had no association with dementia (Alldred et al., 2013). The approaches included medication
stage (Nijk et al., 2009). Both antipsychotic and anxiolytic review, multidisciplinary case conferencing, staff education
use were associated with agitation. and use of clinical decision support technology. The authors
Several studies have shown considerable variation between concluded that the interventions did lead to the identi-
nursing homes in the extent of prescribing of antipsychotic fication and resolution of medication-related problems.
medication. Kleijer et al. (2014) found that facilities with a However, evidence of an effect on resident-related outcomes
high prevalence of such prescribing had higher proportions was not clearly established. Ideally LTC home leaders will
of residents who were physically dependent and cognitively develop quality improvement projects focused on how to
impaired, though levels of BPSD were not much greater than optimize psychotropic prescribing.
those recorded in the other facilities. They were larger, their
residents were more depressed and consumers (relatives and
residents) were less satisfied with staffing and personal care.
Concern about overprescribing of psychotropic medications 28.6 STANDARDS OF CARE IN LTC
in LTC settings persists and in some countries, e.g. England, FACILITIES
national targets have been set for reduced utilization with a
focus on antipsychotics (Banerjee, 2009). There is some evi- Useful advice on standards to be achieved in nursing home
dence that significant reductions have already occurred care has been provided by Koopmans (2005). Physicians
(Martinez et al., 2013). Evidence that antipsychotic medication specially trained in nursing home medicine are employed
is associated with increased rates of death and cerebrovascular by Dutch nursing homes, with a ratio of one doctor per
events as well as other adverse effects underline the need for 100 patients. Availability of doctors and specialists to visit
fully informed consent prior to initiation of treatment. or work in LTC facilities varies between jurisdictions, as
Use of other types of medication in managing scream- does the availability of psychologists. Standards of care are
ing, hypersexuality and other behavioural problems has largely dependent on staffing, and how well the staff work
been discussed elsewhere in this book, as has the use of anti- with residents. Standards regarding regular screening of
cholinesterase inhibitors. Frequency of use of the latter in mood, behaviour, physical function and use of medica-
nursing homes varies markedly between countries. tion are desirable, and capacity to respond to special needs
It is relevant to note that the evidence showing effective- should be one of the standards. Dufey and Hope (2005) have
ness of antidepressants in both community (Roose et al., listed a number of standards for nurses to achieve when car-
2004) and LTC (Snowden et al., 2003) settings is limited, ing for people with dementia.
and that a systematic review found no clear evidence for A Dutch-led multidisciplinary and multinational strat-
efficacy of antidepressants in dementia (Bains et al., 2009). egy resulted in the publication of a set of unique quality
Depression levels reduced in only one third of depressed indicators (QIs) that aims exclusively at assessing the qual-
residents treated with antidepressants for 2 months (Boyle ity of psychosocial dementia care. It is hoped that following
et al., 2004). Several studies showed that residents with implementation, these QIs will assist dementia care profes-
dementia responded less well than those without dementia sionals to individualize and tailor psychosocial interven-
(Snowden et al., 2003). Depression among residents with tions (Vasse et al., 2012).
dementia develops in response to a complex mix of factors,
some ongoing, so that non-response to antidepressants may
seem understandable. When symptoms are common to
both depression and dementia, difficulty suppressing symp-
28.7 PALLIATIVE CARE
toms may be because antidepressants have no effect on the
underlying dementia (Purandare et al., 2001). Hertogh (2005a) has discussed issues to be addressed dur-
ing end-of-life care, which in due course applies to most
28.5.4 OPTIMAL PSYCHOTROPIC people living in nursing homes. Quality of life needs to be
PRESCRIBING APPROACHES assessed. Provision of high-quality palliative care in the
nursing home setting is essential (Engel et al., 2006); next of
Many factors combine during the decision-making process kin are generally opposed to use of feeding tubes. Advance
related to prescribing of a new psychotropic medication in directives should be discussed with a care provider at the
the LTC home. Although the physician makes the final deci- time of admission. However, Hertogh (2005a) pointed out
sion other team members play a critical role as well. A vari- that euthanasia in cases of dementia is almost an unthink-
ety of approaches have been tested to determine how best to able option due to impaired cognitive skills and inability
optimize overall prescribing patterns. A Cochrane review of such patients to provide informed consent. Opiates are
of psychosocial interventions for reducing antipsychotic use frequently used in Dutch nursing homes for symptom con-
found that reductions can be achieved through education trol, but the administration of medications specifically
and training of staff and/or multidisciplinary team meet- intended to cause death was found to be uncommon (2%)
ings (Richter et al., 2012). Another Cochrane review focused (van der Steen et al., 2005). These issues are discussed in
on interventions to optimize prescribing in LTC homes Chapter 37 too.
Residential care for people with dementia 299

REFERENCES
28.8 USE OF TECHNOLOGIES IN
RESIDENTIAL CARE Access Economics (2009). Making choices. Future demen-
tia care: Projections, problems and preferences.
A range of technologies are beginning to be incorporated Report for Alzheimer’s Australia. https://fightdemen-
into LTC settings. These include a variety of monitoring tia.org.au/sites/default/files/20090400_Nat__AE_
devices; tools to enhance activities of daily living; robots; FullMakingChoices.pdf. Accessed 3 June 2016.
provision of care via telemedicine and approaches designed Alldred, D.P., Raynor, D.K., Hughes, C. et al. (2013).
to enhance social connectedness. Monitoring devices Interventions to optimise prescribing for older
include falls detection and prevention systems, mobility people in care homes. Cochrane Database Systematic
aids, activity and sleep monitors and medication compli- Reviews, (2): CD009095.
ance systems (Czaja et al., 2013). Videoconferencing can Alexopoulos, G.S., Abrams, R.C., Young, R.C. and
connect residents in LTC with family and friends, result- Shamoian, C.A. (1988). Cornell Scale for depression in
ing in reduced isolation and loneliness. Telepsychiatry has dementia. Biological Psychiatry, 23: 271–284.
proven valuable in the care of nursing home residents, American Geriatrics Society and American Association
providing virtual assessment and follow-up (Rabinowitz for Geriatric Psychiatry (2003a). Consensus statement
et al., 2010). The use of video-simulated presence (with on improving the quality of care in US nursing homes:
pre-recorded video by a family member) appears to assist Management of depression and behavioral symptoms
in reducing resistance to care (O’Connor et al., 2011). There associated with dementia. Journal of the American
is some evidence that companion robots can have a posi- Geriatrics Society, 51: 1287–1298.
tive effect on quality of life of people with mid-to-late stage American Geriatrics Society and American Association for
dementia. Two randomized trials looked at the use a com- Geriatric Psychiatry (2003b). The American Geriatrics
panion robot and suggested benefits on scores of pleasure Society and American Geriatric Psychiatry recommen-
(Moyle et al., 2013) and loneliness (Robinson et al., 2013). dations for policies in support of quality mental health
The introduction of new technologies can be challenging care in U.S. nursing homes. Journal of the American
and Niemeijer et al. (2010) have provided useful commen- Geriatrics Society, 51: 1299–1304.
tary on the ethical and practical concerns associated with Bagley, H., Cordingley, L., Burns, A. et al. (2000).
the introduction of surveillance technologies in residential Recognition of depression by staff in nursing and resi-
care as well as an ethnographic field study on how staff actu- dential homes. Journal of Clinical Nursing, 9: 445–450.
ally incorporate these technologies into practice (Niemeijer Bains, J., Birks, J. and Dening, T. (2009). Antidepressants
et al., 2015). for treating depression in dementia. The Cochrane
Database of Systematic Reviews, (3): CD003944.
Ballard, C.G., Margallo-Lana, M., Fossey, J. et al. (2001). A
1-year follow-up study of behavioral and psychological
28.9 GOOD CARE symptoms in dementia among people in care environ-
ments. Journal of Clinical Psychiatry, 62: 631–636.
Hertogh (2005b) has discussed elements of an ‘ethic of Banerjee, s. (2009). The Use of Antipsychotic Medication
care’ in LTC for people with dementia. Caregivers are for People with Dementia: A Time for Action. England:
encouraged to move beyond a simple custodial approach Department of Health, National Health Service.
to caring and focus on the residual abilities and subjec- Barca, M.L., Selbaek. G., Laks, J. and Engedal, K. (2008).
tive experiences rather than functional deficits of people The pattern of depressive symptoms and factor analy-
with dementia. The ethic of care ‘allows criticism of a soci- sis of the Cornell Scale among patients in Norwegian
ety that is too unilaterally oriented towards the maxims nursing homes. International Journal of Geriatric
of individual autonomy and independence’. Central to the Psychiatry, 23: 1058–1065.
ethic are safety and self-esteem. Caregivers should afford Barca, M.L., Selbaek, G., Laks, J. and Engedal, K. (2009).
people with dementia meaningful choices that allow them Factors associated with depression in Norwegian nurs-
to enhance their self-esteem and personhood. Good care ing homes. International Journal of Geriatric Psychiatry,
recognizes the fear and anxiety experienced by people with 24: 417–425.
dementia due to loss of control and progressive threaten- Beck, C.K. and Shue, V.M. (1994). Interventions for treat-
ing of their identity. They need a safe haven where they can ing disruptive behavior in demented elderly people.
feel ‘at home’ and secure. Nursing Clinics of North America, 29: 143–155.
‘Bringing together of functional caring and empathetic Bharucha, A.J., Dew, M.A., Miller, M.D. et al. (2006).
treatment is exactly what defines the specific competency Psychotherapy in long-term care: A review. Journal of
needed for caregiving in dementia’. There is also a need the American Medical Directors Association, 7: 568–580.
to care for the carers and attention to the morale of those Blanchard, M., Serfaty, M., Duckett, S. and Flatley, M.
working and living in LTC facilities. Adequate funding for (2009). Adapting services for a changing society: A
well-integrated care is vital. reintegrative model for old age psychiatry (based on
300 Dementia

a model proposed by Knight and Emanuel, 2007). Long-Term Care Home. A Handbook for Staff, Third
International Journal of Geriatric Psychiatry, 24: 202–206. Edition, Chapter 16. Toronto, Ontario: Hogrefe &
Borson, S., Loebel, J.P., Kitchell, M. et al. (1997). Huber, pp. 267–278.
Psychiatric assessments of nursing home residents Cummings, J.L., Mega, M., Gray, K. et al. (1994). The
under OBRA-87: Should PASARR be reformed? Journal Neuropsychiatric Inventory: Assessing psychopathol-
of the American Geriatrics Society, 45: 1173–1181. ogy in dementia patients. Neurology, 44: 2308–2314.
Boyle, V.L., Roychoudhury, C., Beniak, R. et al. (2004). Czaja, S., Beach, S., Charness, N. and Schulz, R. (2013).
Recognition and management of depression in skilled- Older adults and the adoption of healthcare technol-
nursing and long-term care settings. Evolving targets ogy: Opportunities and challenges. In A. Sixsmith
for quality improvement. American Journal of Geriatric and G. Gutman (eds.), Technologies for Active Aging,
Psychiatry, 12: 288–295. Chapter 3. New York, NY: Springer, pp. 27–46.
Brodaty, H., Draper, B., Saab, D. et al. (2001). Psychosis, Davison, T.E. Snowdon, J., Castle, N. et al. (2012). An eval-
depression and behavioural disturbances in Sydney uation of a national program to implement the Cornell
nursing home residents: Prevalence and predictors. Scale for Depression in Dementia into routine practice
International Journal of Geriatric Psychiatry, 16: 504–512. in aged care facilities. International Psychogeriatrics,
Camp, C.J., Cohen-Mansfield, J. and Cepezuti, E.A. 24: 631–641.
(2002). Use of nonpharmacologic interventions among Donath, C., Winkler, A. and Grabel, E. (2009). Short-term
nursing home residents with dementia. Psychiatric residential care for dementia patients: Predictors for
Services, 53: 1397–1401. utilization and expected quality from a family care-
Carpenter, B., Ruckdeschel, K., Ruckdeschel, H. and Van giver’s point of view. International Psychogeriatrics, 21:
Haitsma, K. (2002). R-E-M psychotherapy: A manu- 703–710.
alized approach for long-term care residents with Downs, M., Brooker, D. and Bruce, E. (2005). The practice
depression and dementia. Clinical Gerontologist, 25: of person-centred dementia care: UK. In A. Burns (ed.),
25–49. Standards in Dementia Care, Chapter 2. Abingdon,
Chenoweth, L., King, M.T., Jeon, Y.-H. et al. (2009). Caring England: Taylor & Francis, pp. 13–19.
for aged dementia care resident study (CADRES) of Dufey, A.-F. and Hope, K. (2005). Recognizing and deliver-
person-centred care, dementia-care mapping, and ing quality nursing care for people with dementia. In
usual care in dementia: A cluster-randomised trial. A. Burns (ed.), Standards in Dementia Care, Chapter
Lancet Neurology, 8: 317–325. 21. London: Taylor & Francis, pp. 155–163.
Cody, M., Beck, C. and Svarstad, B.L. (2002). Challenges Engel, S.E., Kiely, D.K. and Mitchell, S.L. (2006).
to the use of nonpharmacologic interventions in nurs- Satisfaction with end-of-life care for nursing home
ing homes. Psychiatric Services, 53: 1402–1406. residents with advanced dementia. Journal of the
Cohen, C.I., Hyland, K. and Kimhy, D. (2003). The utility of American Geriatrics Society, 54: 1567–1572.
mandatory depression screening of dementia patients Evers, M.M., Samuels, S.C., Lantz, M. et al. (2002). The
in nursing homes. American Journal of Psychiatry, 160: prevalence, diagnosis and treatment of depression in
2012–2017. dementia patients in chronic care facilities in the last
Cohen-Mansfield, J. (2001). Nonpharmacologic inter- six months of life. International Journal of Geriatric
ventions for inappropriate behaviors in dementia: A Psychiatry, 17: 464–472.
review, summary and critique. American Journal of Ferri, C.P., Prince, M., Brayne, C. et al.; Alzheimer’s
Geriatric Psychiatry, 9: 361–381. Disease International (2005). Global prevalence of
Cohen-Mansfield, J., Marx, M.S. and Rosenthal, A.S. dementia: A Delphi consensus study. Lancet, 366:
(1989). A description of agitation in a nursing home. 2112–2117.
Journal of Gerontology, 44: M77–M84. Fleming, R. and Purandare, N. (2010). Long-term care for
Cohen-Mansfield, J., Thein, K., Marx, M.S. and Dakheel- people with dementia: Environmental design guide-
Ali, M. (2012). What are the barriers to performing lines. International Psychogeriatrics, 22: 1084–1096.
nonpharmacological interventions for behavioral Gerety, M.B., Williams, J.W., Mulrow, C.D. et al. (1994).
symptoms in the nursing home? Journal of the Performance of case-finding tools for depression in
American Medical Directors Association, 13: 400–405. the nursing home: Influence of clinical and functional
Conn, D.K. (2007). Mental health issues in long-term care characteristics and selection of optimal threshold
facilities. In D.K. Conn et al. (eds.), Practical Psychiatry in scores. Journal of the American Geriatrics Society, 42:
the Long-Term Care Home. A Handbook for Staff, Third 1103–1109.
Edition. Toronto, Ontario: Hogrefe & Huber, p. 11. Greenberg, L., Lantz, M.S., Likourezos, A. et al. (2004).
Conn, D.K. and Gibson, M. (2007). Guidelines for the Screening for depression in nursing home pallia-
assessment and treatment of mental health issues. tive care patients. Journal of Geriatric Psychiatry and
In D.K. Conn et al. (eds.), Practical Psychiatry in the Neurology, 17: 212–218.
Residential care for people with dementia 301

Gruber-Baldini, A.L., Boustani, M., Sloane, P.D. and Kurlowicz, L.H., Evans, L.K., Strumpf, N.E. and Maislin, G.
Zimmerman, S. (2004). Behavioural symptoms in resi- (2002). A psychometric evaluation of the Cornell Scale
dential care/assisted living facilities: Prevalence, risk for depression in dementia in a frail, nursing home
factors, and medication management. Journal of the population. American Journal of Geriatric Psychiatry,
American Geriatrics Society, 50: 1610–1617. 10: 600–608.
Gruber-Baldini, A.L., Zimmerman, S., Boustani, M. et al. Lai, C.K., Yeung, J.H., Mok, V. and Chi, I. (2009). Special
(2005). Characteristics associated with ­depression care units for dementia individuals with behavioural
in long-term care residents with dementia. problems. Cochrane Database Systematic Reviews, (4):
Gerontologist, 45 (Special Issue 1): 50–55. CD006470.
Harrington, C. (2001). Residential nursing facilities in the Lövheim, H., Sandman, P.-O., Kallin, K. et al. (2006).
United States. British Medical Journal, 323: 507–510. Relationship between antipsychotic drug use and
Herrmann, N., Mittmann, N., Silver, I.L. et al. (1996). A vali- behavioral and psychological symptoms of dementia in
dation study of the Geriatric Depression Scale short old people with cognitive impairment living in geriatric
form. International Journal of Geriatric Psychiatry, 11: care. International Psychogeriatrics, 18: 713–726.
457–460. Low, L.F., Goodenough, B., Fletcher, J. et al. (2014). The
Hertogh, C.M.P.M. (2005a). End-of-life care and medi- effects of humor therapy on nursing home residents
cal decision-making in patients with dementia. In A. measured using observational methods: The SMILE
Burns (ed.). Standards in Dementia Care, Chapter 45. cluster randomized trial. Journal of the American
London: Taylor & Francis, pp. 339–353. Medical Directors Association, 15: 564–569.
Hertogh, C.M.P.M. (2005b). Towards a more adequate Margallo-Lana, M., Swann, A., O’Brien, J. et al. (2001).
moral framework: Elements of an ‘ethic of care’ in Prevalence and pharmacological management of
nursing home care for people with dementia. In A. behavioural and psychological symptoms amongst
Burns (ed.), Standards in Dementia Care, Chapter 48. dementia sufferers living in care environments.
London: Taylor & Francis, pp. 371–377. International Journal of Geriatric Psychiatry, 16: 39–44.
Hosia-Randell, H. and Pitkälä, K. (2005). Use of psycho- Martinez, C., Jones, R.W. and Rietbrock, S. (2013). Trends
tropic drugs in elderly nursing home residents with in the prevalence of antipsychotic drug use among
and without dementia in Helsinki, Finland. Drugs and patients with Alzheimer’s disease and other dementias
Aging, 22: 793–800. including those treated with antidementia drugs in the
Hughes, C.M., Lapane, K.L., Mor, V. et al. (2000). The community in the UK: A cohort study. British Medical
impact of legislation on psychotropic drug use in nurs- Journal Open, 3(1): e002080.
ing homes: A cross-national perspective. Journal of Matthews, F.E. and Dening, T.R. (2002). Prevalence of
the American Geriatrics Society, 48: 931–937. dementia in institutional care. Lancet, 360: 225–226.
Jang, Y., Haley, W.E., Small, B.J. and Mortimer, J.A. (2002). McCabe, M.P., Karantzas, G.C., Mrkic, D. et al. (2013). A
The role of mastery and social resources in the asso- randomized control trial to evaluate the beyond blue
ciations between disability and depression in later life. depression training program: Does it lead to better
Gerontologist, 42: 807–813. recognition of depression? International Journal of
Jones, R.N., Marcantonio, E.R. and Rabinowitz, T. (2003). Geriatric Psychiatry, 28: 221–226.
Prevalence and correlates of recognized depression in Menon, A.S., Gruber-Baldini, A.L., Hebel, J.R. et al.
U.S. nursing homes. Journal of the American Geriatrics (2001). Relationships between aggressive behaviors
Society, 51: 1404–1409. and depression among nursing home residents with
Katz, I.R. and Parmelee, P.A. (1994). Depression in elderly dementia. International Journal of Geriatric Psychiatry,
patients in residential care settings. In L.S. Schneider 16: 139–146.
et al. (eds.), Diagnosis and Treatment of Depression Montgomery, S.A. and Åsberg, M. (1979). A new depres-
in Late Life, Chapter 24. Washington, DC: American sion scale designed to be sensitive to change. British
Psychiatric Press, pp. 437–461. Journal of Psychiatry, 134: 382–389.
Kaup, B.A., Loreck, D., Gruber-Baldini, A.L. et al. (2007). Moyle, W., Cooke, M., Beattie, E. et al. (2013). Exploring
Depression and its relationship to function and the effect of companion robots on emotional expres-
medical status, in nursing home admissions. American sion in older adults with dementia: A pilot randomized
Journal of Geriatric Psychiatry, 15: 438–442. controlled trial. Journal of Gerontological Nursing, 39:
Kleijer, B.C., van Marum, R.J., Frijters, D.H.M. et al. 46–53.
(2014). Variability between nursing homes in preva- National Center for Health Statistics, United States (2004).
lence of antipsychotic use in patients with dementia. National Nursing Home Survey, 2004. Available at:
International Psychogeriatrics, 26: 363–371. http://www.cdc.gov/nchs/nnhs/. Accessed 3 June 2016.
Koopmans, R.T.C.M. (2005). Standard of care for dementia Niemeijer, A.R., Depla, M.F., Frederiks, B.J. and Hertogh,
in nursing homes: The Dutch experience and views. In C.M. (2015). The experiences of people with dementia
A. Burns (ed.). Standards in Dementia Care, Chapter and intellectual disabilities with surveillance technolo-
20. London: Taylor & Francis, pp. 143–153. gies in residential care. Nursing Ethics, 22 (3): 307–320.
302 Dementia

Niemeijer, A.R., Frederiks, B.J., Riphagen, I.I. et al. (2010). Roose, S.P., Sackheim, H., Krishnan, K.R.R. et al. (2004).
Ethical and practical concerns of surveillance technolo- Antidepressant pharmacotherapy in the treatment of
gies in residential care for people with dementia or depression in the very old: A randomized, placebo-
intellectual disabilities: An overview of the literature. controlled trial. American Journal of Psychiatry, 161:
International Psychogeriatrics, 22: 1129–1142. 2050–2059.
Nijk, R.M., Zuidema, S.U. and Koopmans, R.T.C.M. (2009). Rosenblatt, A., Samus, Q.M., Steele, C.D. et al. (2004).
Prevalence and correlates of psychotropic drug The Maryland assisted living study: Prevalence, recog-
use in Dutch nursing-home patients with dementia. nition, and treatment of dementia and other psychi-
International Psychogeriatrics, 21: 485–493. atric disorders in the assisted living population of
O’Connor, C.M., Smith, R., Nott, M.T. et al. (2011). Using Central Maryland. Journal of the American Geriatrics
video simulated presence to reduce resistance to care Society, 52: 1618–1625.
and increase participation of adults with dementia. Rovner, B.W., Edelman, B.A., Cox, M.P. and Shmuely, Y.
American Journal of Alzheimers Disease and Other (1992). The impact of antipsychotic drug regulations on
Dementias, 26: 317–325. psychotropic prescribing practices in nursing homes.
Organization for Economic Cooperation and American Journal of Psychiatry, 149: 1390–1392.
Development (OECD) (2011). Help Wanted – Providing Scocco, P., Rapattoni, M. and Fantoni, G. (2006). Nursing
and Paying for Long Term Care. Available at http:// home institutionalization: A source of eustress or dis-
www/oecd.org/els/healthsystems/help_wanted.htm; tress for the elderly? International Journal of Geriatric
Accessed 29 April 2015. Psychiatry, 21: 281–287.
Parmelee, P.A., Katz, I.R. and Lawton, M.P. (1993). Anxiety Selbaek, G., Kirkevold, O. and Engedal, K. (2007). The
and its association with depression among institution- prevalence of psychiatric symptoms and behavioural
alized elderly. American Journal of Geriatric Psychiatry, disturbances and the use of psychotropic drugs in
1: 46–58. Norwegian nursing homes. International Journal of
Payne, J.L., Sheppard, J.-M., Steinberg, M. et al. (2002). Geriatric Psychiatry, 22: 843–849.
Incidence, prevalence, and outcomes of depression in Seitz, D.P., Brisbin, S., Herrmann, N. et al. (2012). Efficacy
residents of a long-term care facility with dementia. and feasibility of nonpharmacological interventions for
International Journal of Geriatric Psychiatry, 17: 247–253. neuropsychiatric symptoms of dementia in long term
Purandare, N., Burns, A., Challis, D. and Morris, J. (2004). care: A systematic review. Journal of the American
Perceived mental health needs and adequacy of Medical Directors Association, 13: 503–506.
service provision to older people in care homes in the Seitz, D., Purandare, N. and Conn, D. (2010). Prevalence
UK: A national survey. International Journal of Geriatric of psychiatric disorders among older adults in long-
Psychiatry, 19: 549–553. term care homes: A systematic review. International
Purandare, N., Burns, A., Craig, S. et al. (2001). Depressive Psychogeriatrics, 22: 1025–1039.
symptoms in patients with Alzheimer’s disease. Sloane, P.D., Mathew, L., Scarborough, M. et al. (1991).
International Journal of Geriatric Psychiatry, 16: 960–964. Physical and pharmacologic restraint of nursing home
Rabinowitz, T., Murphy, K.M., Amour, J.L. et al. (2010). patients with dementia. Impact of specialized units.
Benefits of a telepsychiatry consultation service for Journal of the American Medical Association, 265:
rural nursing home residents. Telemedicine Journal 1278–1282.
and E-Health, 16: 34–40. Smalbrugge, M., Jongenelis, L., Pot, A.M. et al. (2006).
Ragsdale, V. and McDougall, G.J. (2008). The changing Incidence and outcome of depressive symptoms in
face of long-term care: Looking at the past decade. nursing home patients in the Netherlands. American
Issues in Mental Health Nursing, 29: 992–1001. Journal of Geriatric Psychiatry, 14: 1069–1076.
Reisberg, B., Borenstein, J., Salob, S.P. et al. (1987). Snowden, M., Sato, K. and Roy-Byrne, P. (2003).
Behavioral symptoms in Alzheimer’s disease: Assessment and treatment of nursing home residents
Phenomenology and treatment. Journal of Clinical with depression or behavioral symptoms associated
Psychiatry, 48 (Suppl.): 9–15. with dementia: A review of the literature. Journal of
Ribbe, M.W., Ljunggren, G., Steel, K. et al. (1997). Nursing the American Geriatrics Society, 51: 1305–1317.
homes in 10 nations: A comparison between countries Snowdon, J. (2010). Mental health service delivery in long-
and settings. Age and Ageing, 26 (Suppl. 2): 3–12. term care homes. International Psychogeriatrics, 22:
Richter, T., Meyer, G., Möhler, R. and Köpke, S. (2012). 1063–1071.
Psychosocial interventions for reducing antipsy- Snowdon, J., Day, S. and Baker, W. (2005). Why and how
chotic medication in care home residents. Cochrane antipsychotic drugs are used in 40 Sydney nursing
Database Systematic Reviews (12): CD008634. homes. International Journal of Geriatric Psychiatry, 20:
Robinson, H., Macdonald, B., Kerse, N. and Broadbent, E. 1146–1152.
(2013). The psychosocial effects of a companion robot: Snowdon, J., Day, S. and Baker, W. (2006). Current
A randomized controlled trial. Journal of the American use of psychotropic medication in nursing homes.
Medical Directors Association, 14: 661–667. International Psychogeriatrics, 18: 241–250.
Residential care for people with dementia 303

Snowdon, J. and Fleming, R. (2008). Recognising depres- Van der Steen, J.T., van der Wal, G., Mehr, D.R. et al.
sion in residential facilities: An Australian challenge. (2005). End-of-life decision making in nursing home
International Journal of Geriatric Psychiatry, 23: residents with dementia and pneumonia: Dutch physi-
295–300. cians’ intentions regarding hastening death. Alzheimer
Snowdon, J., Galanos, D. and Viswani, D. (2011). Patterns Disease and Associated Disorders, 19: 148–155.
of psychotropic medication use in nursing homes: van Schaik, D.J., Dozeman, E., van Marwijk, H.W. et al.
Surveys in Sydney, allowing comparisons over time and (2014). Preventing depression in homes for older
between countries. International Psychogeriatrics, 23: adults: Are effects sustained over 2years? International
1520–1525. Journal of Geriatric Psychiatry, 29: 191–197.
Teresi, J., Abrams, R., Holmes, D. et al. (2001). Prevalence Vasse, E., Moniz-Cook, E., Rikkert, M.O. et al. (2012). The
of depression and depression recognition in nursing development of quality indicators to improve psycho-
homes. Social Psychiatry and Psychiatric Epidemiology, social care in dementia. International Psychogeriatrics,
36: 613–620. 24: 921–930.
Testad, I., Corbett, A., Aarsland, D. et al. (2014). The value Verbeek, H., van Rossum, E., Zwakhalen, S.M.G.
of personalized psychosocial interventions to address et al. (2009). Small, homelike care environments for
behavioral and psychological symptoms in people with older people with dementia: A literature review.
dementia living in care home settings: A systematic International Psychogeriatrics, 21: 252–264.
review. International Psychogeriatrics, 26, 1083–1098. Wancata, J., Benda, N., Meise, U. and Windhaber, J.
Turner, J. and Snowdon, J. (2009). An innovative approach (2003). Non-cognitive symptoms of dementia in nurs-
to behavioral assessment and intervention in residen- ing homes: Frequency, course and consequences.
tial care: A service evaluation. Clinical Gerontologist, Social Psychiatry and Psychiatric Epidemiology, 38:
32: 260–275. 637–643.
Van Bogaert, P., Van Grinsven, R., Tolson, D. et al. (2013). Wittenberg, R., Comas-Herrera, A., Pickard, L. and
Effects of SolCos model-based individual reminis- Hancock, R. (2004). Future Demand for Long-Term
cence on older adults with mild to moderate demen- Care in the UK: A Summary of Projections of Long-
tia due to Alzheimer disease: A pilot study. Journal Term Care Finance for Older People to 2051. Joseph
of the American Medical Directors Association, 14: Rowntree Foundation. Future Costs of Long-Term Care
528.e9–528.e13. for Older People, Findings Ref: 944; JRF, York, United
Van der Linde, R.M., Stephan, B.C., Dening, T. and Kingdom.
Brayne, C. (2014). Instruments to measure behav- Yesavage, J.A., Brink, T.L., Rose, T.L. et al. (1983).
ioural and psychological symptoms of dementia. Development and validation of a geriatric depres-
International Journal of Methods in Psychiatric sion screening scale: A preliminary report. Journal of
Research, 23: 69–98. Psychiatric Research, 17: 37–49.
29
Design and dementia

JUNE ANDREWS

29.1 WHY ATTENTION TO BUILDING 29.2 HOW THE BUILT ENVIRONMENT


DESIGN AND TECHNOLOGY IS AFFECTS THE PERSON WITH
VITAL FOR THE PERSON WITH DEMENTIA
DEMENTIA
29.2.1 IMPAIRED MEMORY
Good design and technology promotes independence for
When you do not remember where you are, it is as if you
people with dementia and decreases the burden of care for
are in the building for the first time. You may ask people
families or health and social care workers. It can reduce the
where you are, and why, and try to leave to identify any
cost of care by postponing institutionalization and decreas-
familiar landmark. If people stop you from getting out, you
ing adverse incidents. In addition, because the dementia-
may become stressed and redouble your efforts to exit. The
friendly recommendation may even be less expensive than
dementia-friendly building aims to compensate for this
what the architect or planner would otherwise propose, the
problem by requiring you to remember less. Every location
financial savings for care providers can start even before the
important to you is obvious and easy to find.
project is functioning.
Dementia is a symptom of a primary disease process.
29.2.2 IMPAIRED REASONING
The ‘dementia’ is the extent to which that process (e.g.
Alzheimer’s disease or vascular disease) renders the per- Anyone can momentarily forget where they are or how
son less able to remember or learn or work things out and to use an everyday object. Usually, we can work it out. A
leaves them with a reduced ability to cope with the stress dementia-friendly building makes this easier. For example,
caused by reduction in that capacity. The dementia symp- the soap, tap, flush handle and the paper holder in the toilet
toms are not necessarily in direct proportion to the extent will be very easy to understand, an objective best achieved
of the disease process. Most people with dementia are by using familiar or traditional designs.
older, or very old, and their capacity to deal with the com-
mon changes of aging is reduced by environmental stress, 29.2.3 IMPAIRED LEARNING
the behaviour of other people, other physical illness and
fatigue. Because of acquired learning problem, the person with
There are three levels of support for design recommen- dementia has difficulty with any new environment. Good
dations: design provides cues to support residual capacity, reducing
distraction that impairs concentration. Design cues can help
●● Research involving people with dementia with orientation and prevent the person straying into unhelp-
●● Generalizing from sensory and physical impairment ful situations. An example is the hazard of frequent exiting
research through the fire escape door in the absence of a fire. Design
●● Sharing examples of good practical solutions based on can make this less likely. We need to balance the continuous
an understanding of cognitive impairment and expert risk of random hazardous exiting with the theoretical risk of
consensus the exit not being found by staff and visitors in a fire.

304
Design and dementia 305

29.2.4 ORDINARY CHANGES OF AGEING in to an alert system or an adapted plug that allows water to
drain away by being triggered by the weight of water above
The majority of people with dementia are old or very old and can help reduce the risk of a costly and potentially perma-
have age-related difficulties. The building needs to compen- nent move.
sate on their behalf. The person may need help, but forget
to use the call assist button. Technology such as a passive 29.3.2 IN HOSPITALS AND CARE HOMES
infrared beam can alert carers to the fact that the person
is on the move and so that they can offer timely assistance It is never too soon in a new build to consider incorporat-
even if no help is requested. ing dementia-friendly features, as simple features in the
architectural plan can improve the building at no extra cost
29.2.5 HIGH LEVELS OF STRESS (Fuggle, 2013). Examples of what design might do include
the following.
The stress created by never knowing where you are or why
you are there and never being even vaguely familiar with the 29.3.2.1 Night care
environment in which you find yourself is unfathomable.
This is made worse by meaningless noise and activity. Good Problem – The person might not sleep well and may fall,
building design can conceal background jobs like deliv- giving rise to injury, disruption of others and organi-
ery of laundry and reduce necessary noises. Incorporating zational consequences.
outdoor spaces for leisure, gardening and exercise helps to Solutions – Due to the yellowing of the ageing eye,
reduce stress. the day/night cycle can be disrupted because of poor
daylight exposure during daylight hours (Koncelik,
2003). Morning access every day to a garden or
bright daylight can help to reset the body clock.
29.3 SOME PRACTICAL IMPLICATIONS
Space and quiet available throughout the day can
support meaningful activity to create healthy fatigue
Staff or lay people who are consulted about design of facili- by bedtime. Reduced noise levels and slowed activity at
ties for people with dementia need an understanding of bedtime can promote falling off to sleep (Kerr et al.,
dementia and of planning in order to prevent their involve- 2008). Glass panels in bedroom doors in conjunction
ment becoming tokenistic, or even unhelpful. The use of with extraneous light can wake the person up and
an audit tool to inform design decisions can help ensure unhelpfully tempt them out of their room. Passive
that evidence rather than fashion informs decisions. It infrared sensors could better warn staff if necessary
is important that any architect or designer is trained in that the person is awake and moving about and can
dementia-friendly principles to ensure best value in the switch on lights to cue the person to use the toilet
project and secure reduced running costs for the build- and go back to bed. The positioning of the bed head
ing. Such expertise can be used at every stage – planning, so that the lavatory pan can be seen is crucial for
building or refurbishment. this. The use of vibrating pagers for call alerts can
reduce the meaningless noise of buzzers in the night
29.3.1 AT HOME (and day). One simple design change to assist people
with dementia at night is the use of an analogue
A familiar home environment with least alteration is best. clock set to the right time. The current generation
Poor short-term memory means that the person may not of older people with dementia may have forgotten
remember endorsing any ‘­improvement’. People often as- what a digital clock is and therefore be unable to
sume they have been robbed, or that someone is trying to tell the time (Fleming et al., 2008). An appropriately
infuriate them by moving belongings round, or that they positioned and well lit clock can reduce the need
are in the wrong house. So you have to balance the potential for someone with dementia to get up to orientate
benefit of change against the more certain risk that it will be themselves.
perplexing. The most productive change is to improve the
level of lighting (McNair et al., 2013). You must increase the
power of all light fittings, clear shrubs from outside win- 29.3.2.2 Hygiene
dows, clean the glass and make sure drapes can be opened
wide. Low-energy light bulbs must be replaced regularly Problem – The person with dementia may not use the toi-
when their luminosity fades. Assistive technology is increas- let appropriately and require extra help with hygiene.
ingly available and affordable (Kerr et al., 2010) and can keep Solutions – Incontinence can be caused by failing to
the person in their own home longer. For example, a person reach the toilet in time, so make sure that the floor
with dementia may forget a running tap and the flood then coverings are smooth, matte and self-coloured to
causes them to be displaced to an unfamiliar setting while encourage the person with dementia to walk more
home repairs are undertaken. Use of a flood detector built swiftly to the right place (Perritt et al., 2005). Make
306 Dementia

the door easy to find by using bright contrasting Exercise is of great value, and designing a space where
colour to the surrounding wall and signage at low dance, or music and movement can happen, in addition
height, so that the person does not have to look up to outside activities, may reduce the pacing behaviour
(Dementia Services Development Centre, 2011). The (Forbes et al., 2013). If the purpose of the pacing is a futile
sign should include words and pictures that make attempt by the person to find their own room, there are
sense to the generation who is using the toilet, i.e. the many design solutions that will help. A lack of everyday
matchstick man/woman sign is a recent identifier for household spaces and objects can lead to anxious old peo-
toilets. The fixtures and fittings should be culturally ple, with no motivation to do anything or who spend the
appropriate and recognizable in traditional designs whole day running their hands across tabletops, having
and contrasting colours. This could currently mean nowhere familiar to do what they have always done to pass
offering a bar of coloured soap and a cotton towel, the time. Given a kitchen, they can happily spend hours
against strict infection control guidance. Some inde- keeping the surfaces clean or mopping the sink. In some
pendent hand-washing is better than dependence on units, the kitchen is reserved for occupational therapy
staff, or worse, no hand-washing. activity and only available to certain patients at certain
times, which is a missed opportunity. Washing poles and a
Shiny bathroom surfaces and floors can cause visual prob- line in the garden with an adjacent sink, some grass to cut
lems leading to falls. Mirrors may cause distress if the per- or an old car in the garden to tinker with can be planned
son does not recognize their reflection. If the bathroom has into the design.
to be used as a store for shiny metal lifting equipment and
boxes of continence pads, it reduces the capacity of the per-
son to work out where they are and why they are there. That
could give rise to resistance to the idea of being undressed, 29.4 CONCLUSIONS
making bath times stressful and difficult for staff. Laying
out of clothes in the order they are to be put on to aid inde- Buildings can help people with dementia to function at their
pendent dressing is easier if there is a place to do that in the best (Dementia Services Development Centre, 2012 and
bathroom. 2013). In dedicated facilities, dementia care, which is com-
plex and challenging, can be made easier by design, which
29.3.2.3 Unwanted exiting and walking also helps families to feel more comfortable when visiting
and supporting their relative. The building can demonstrate
Problem – There is a perceived danger that the person will that people with dementia and the staff are valued and
walk into danger or cause a nuisance. respected. Providing people with dementia with the chance
Solution – The design solution depends on the reason why to make good decisions with helpful outcomes through
it’s a problem. Do staff need to know where everyone design environments that compensate for their impair-
is all the time? Do residents need to burn off energy to ments will enhance the quality of their lives.
reduce their own stress? Is the problem that families
need to feel that their relative is safe? REFERENCES
Discreet electronic equipment can assist with monitor- Dementia Services Development Centre (2011). Dementia
ing movement, minimizing the use of staff time and use of Design Audit Tool, Second Edition. Stirling, United
restraint. Unwanted exiting requires all hazardous exits to Kingdom: Dementia Services Development Centre,
be designed to be less attractive to the casual observer. This University of Stirling.
includes the use of lighting and floor covering and paint Dementia Services Development Centre (2012 and 2013).
work to conceal them and to make the approach to these Virtual Environments. Available at http://dementia.stir.
areas less interesting. Vision panels in exit doors literally ac.uk/design/virtual-environments. Accessed 27 June
offer a window into a more interesting world than where the 2016.
resident is. Turn their attention the other way. A safe garden Fleming, R., Crookes, P. and Sum, S. (2008). A review of the
is a good alternative for the inquisitive, as long as it is fulfill- empirical literature on the design of physical environ-
ing and interesting. ments for people with dementia. Australia: Dementia
Any design features that reduce stress will help. Making Collaborative Research Centres. Available in print
the purpose of the room clear and obvious and keeping the in the United Kingdom as part of Design for People
size at a domestic scale is useful. High levels of noise and low with Dementia: Audit tool. Stirling, United Kingdom:
levels of light have been shown to make people with demen- Dementia Services Development Centre, University
tia restless. Designing in the ability to locally control the of Stirling.
temperature allows staff to support the person to find one Forbes, D., Forbes, S.C., Blake, C.M. et al. (2013).
individual place that suits them. Pain may be a root cause of Exercise programs for people with dementia.
restlessness and a comfortable quiet place to sit would help The Cochrane Database of Systematic Reviews
(McClean and Cunningham, 2007). (12): CD006489.
Design and dementia 307

Fuggle, L. (2013). Designing Interiors for People with Koncelik, J. (2003). The human factors of ageing. In R.J.
Dementia, Fourth Edition. Stirling, United Kingdom: Scheidt and P. Windley (eds.), Physical Environments and
Dementia Services Development Centre, University of Ageing. Canada: Hawthorn Press Canada.
Stirling. McClean, W. and Cunningham, C. (2007). Pain in Older
Kerr, B., Cunningham, C. and Martin, S. (eds.) (2010). People with Dementia: A Practice Guide. Stirling,
Telecare and Dementia: Using Telecare Effectively in United Kingdom: Dementia Services Development
the Support of People with Dementia. Stirling, United Centre, University of Stirling.
Kingdom: Dementia Services Development Centre, McNair, D., Cunningham, C., Pollock, R. and McGuire,
University of Stirling. B. (2013). Light and Lighting Design for People with
Kerr, D., Wilkinson, H. and Cunningham, C. (2008). Dementia, Third Edition. Stirling, United Kingdom:
Supporting Older People in Care Homes at Dementia Services Development Centre, University
Night. York, United Kingdom: Joseph Rowntree of Stirling.
Foundation. Website Download Correct as of Perritt, M., McCune, E.D. (ed) and McCune, S.L. (2005).
31.09.09. Available at http://www.jrf.org.uk/publica- Research informs design: Empirical findings suggest
tions/supporting-older-people-care-homes-night. recommendations for carpet pattern and texture.
Accessed 27 June 2016. Alzheimer’s Care Quarterly, 6: 300–305.
30
Legal issues and dementia

HUGH SERIES AND ROBIN JACOBY

Many countries have statute law about capacity, which


30.1 JURISDICTIONS is usually separate from legislation about the compulsory
treatment of those with mental disorders. Northern Ireland
Law varies across the world. Anglophone jurisdictions is unusual in considering the introduction of a combined
(including the United Kingdom, the Commonwealth, the mental health/mental capacity act. In England and Wales,
United States and Hong Kong) incorporate much com- the relevant statute is the Mental Capacity Act 2005 (MCA),
mon law – law made by judicial precedent – in addition which sets out what is meant by capacity, how decisions
to statute law made by legislatures. However, many other may be made for people lacking capacity, what steps a per-
countries have a civil law legal system which is based on a son may take to retain a degree of autonomy for his own
code, derived from Roman law, in which judges apply the future care, as well as the legal framework for the Court
code rather than develop binding case precedents. This of Protection, which oversees the care of people who lack
chapter focuses mainly on the anglophone system. capacity. Its application is not by any means restricted to
people with dementia, but it is critical legislation for them.

30.2 CAPACITY 30.2.1 GENERAL REQUIREMENTS FOR


CAPACITY
Many of the legal issues that arise in dementia focus on To make a decision with capacity a person must be able to:
the affected person’s mental capacity or competence. In
the United Kingdom, ‘competence’ tends to be used in a ●● Understand the information relevant to the decision
­non-legal context and ‘capacity’ in a legal one, whereas in ●● Retain that information
the United States it is the other way round. In this chapter, ●● Use or weigh that information as part of the process of
the words will be considered interchangeable and should be making the decision
understood to refer to mental capacity. ●● Communicate his decision (whether by talking, using
Capacity is specific to a particular decision. Having the sign language or any other means)
capacity to make one kind of decision does not necessarily
mean having the capacity to make another. For example, a The MCA begins with five general principles:
person with dementia may no longer be competent to man-
age his financial affairs, but may still be competent to donate 1. A person must be assumed to have capacity unless it is
power of attorney (see Section 30.3.1 ). established otherwise.
Capacity is also time specific. A person must have 2. Everything possible must be done to assist the person
capacity to do the task at the time he does it, irrespec- to be able to make the decision himself.
tive of whether he lacked capacity before or afterwards. 3. A person should not be treated as lacking capacity
For example, someone with dementia could be in hospital simply because he makes an unwise decision.
with a serious infection that causes delirium and renders 4. An act done for a person lacking capacity must be
him incompetent to make a will. He is given antibiotics, done in his best interests.
the delirium remits, and he regains the capacity to make 5. An act done for a person lacking capacity must be the
the will. least restrictive act possible.

308
Legal issues and dementia 309

It seems that the first principle has not always been can speak on behalf of the person, an Independent Mental
appreciated by doctors. In a survey of 2100 physicians, sur- Capacity Advocate (IMCA) may be appointed. An IMCA is
geons and psychiatrists, Markson et al. (1994) found that not the decision maker, but speaks on behalf of the person
72% erroneously considered that dementia automatically who lacks capacity to ensure that his perspective is prop-
conferred incapacity, 66% thought the same for depression erly considered. If a decision is significant and cannot be
and 71% for psychosis, although psychiatrists were more reached informally it may be necessary to refer the matter
likely than others to respond correctly. It is not correct to to the Court of Protection.
assume that a person lacks capacity simply because he has
dementia – or any other condition or diagnosis. 30.2.1.1 Maintaining autonomy
A person with borderline capacity for a task may be
tilted towards having capacity by optimizing the situa- Much capacity legislation is built on the principle of enhanc-
tion and providing assistance. Allowing adequate time for ing a person’s autonomy, that is, allowing him to make deci-
an assessment and sitting opposite the person so that he sions for himself wherever possible. This may mean, as we
can lip-read and hear better are not only standard require- have seen, supporting him to make decisions in the present.
ments for any examination of an older person but may also But it also means setting up mechanisms to allow the per-
maximize the chance that he will be competent to make son to make decisions in advance. These may be in the form
the decision that is needed. It is necessary to ensure that of specific advance decisions, or the person may prefer to
the person can understand the language used (simple lan- appoint someone whom he trusts to make decisions on his
guage, use an interpreter if necessary) and can hear prop- behalf. In England and Wales, this is known as a Lasting
erly (a quiet side room instead of a noisy general hospital Power of Attorney (LPA). It is ‘lasting’ because, unlike a
ward, a hearing aid). general power of attorney, the power continues after the
It can be very tempting to assume that if a person makes person loses capacity to understand the power. LPAs may
an unwise decision then that shows that he lacks capacity, relate to property and financial affairs, or to personal wel-
so that, instead of accepting the decision, it can be assumed fare matters. A given type of LPA does not give authority
that he lacks capacity and a best interests decision can be to make decisions over matters which are not included in
made on his behalf. On the third principle in the MCA, set the power. In the United States, similar arrangements exist
out above, this would not be correct. However, an unwise but may vary by state. Lasting powers are usually referred
decision may well flag up the possibility of lack of capacity, to in the United States as ‘durable’, and the attorney as an
which would require further assessment (or ‘anxious scru- ‘attorney-in-fact’. Some powers cover financial affairs only,
tiny’ as one judge eloquently put it). others cover health and welfare matters.
In English law, and in many other jurisdictions, an act Many people think that the next of kin or nearest rela-
done for a person lacking capacity must be done in his best tive has a particular right to make decisions on behalf of
interests (principle 4), and must be the least restrictive step an incapacitated person. This is not usually the case, except
possible (principle 5). This may still leave a very wide set in very specific circumstances, such as when a person is
of possible decisions. It is not always clear who the relevant detained in a psychiatric hospital under the Mental Health
decision maker might be, but the MCA sets out what mat- Act for treatment of mental disorder. Close family members
ters he should take into account in making the decision can have a very important role in decision making as they
(MCA section 4). Very often this will be clear from the situ- have detailed knowledge of the person’s previous wishes and
ation: if the decision is whether the person should have a views, and their views will need to be very carefully consid-
bath, the decision maker will be the carer offering the bath. ered. However, they will not generally be the legal decision
If the decision is whether or not to have a particular medical maker where statutory authorities (health or social services)
procedure, the decision maker will usually be the doctor in are involved, unless they have been appointed under an LPA.
charge of the treatment. However, if the decision is whether
or not the person should be removed from his home to go
into a care home, the decision is likely to involve many
people, and the local authority may well be involved. The 30.3 SPECIFIC CAPACITIES
MCA requires that appropriate people should be consulted
before a complex best interests decision is made, and a help- 30.3.1 MANAGEMENT OF FINANCIAL
ful way to do this is by means of a best interests meeting, AFFAIRS
held to bring together the key parties such as family and
professionals. Such a meeting should be carefully set up, Capacity is relative to the complexity of the matter to be
chaired, and minutes taken. It may only go ahead once it decided. Consider the theoretical case of two people with
has been confirmed that the person lacks capacity. There is exactly the same degree of dementia. The first has an occu-
no requirement that the same person who confirms the lack pational pension from which his rent and utility bills are
of capacity should make the relevant decision. For decisions paid by direct debit from his bank account, and a small
about major medical treatment, or about where a person amount of money in a savings account. He is assessed by an
should live, where there is no close friend or relative who old age psychiatrist who judges him competent to manage
310 Dementia

his limited financial affairs. The second person is a mil- 30.3.2.1 Power of attorney
lionaire with a variety of share portfolios, joint trusts and
other complex financial instruments. The same psychiatrist Some jurisdictions allow a person to make a lasting power
judges him to be incompetent to manage his affairs. of attorney that covers health and treatment decisions. In
For people who lack the capacity to manage their financial England, this is called a health and welfare LPA. It works
affairs most countries have legislated for some sort of power in much the same way as a property and affairs LPA, except
of attorney, such as the LPA for property and affairs dis- that it only comes into effect when the donor lacks capac-
cussed above. ‘Attorney’ here and elsewhere in this chapter ity (by contrast, an English property and affairs LPA, unless
means the person nominated in a power of attorney, and not otherwise restricted, can be used as soon as it is registered,
necessarily a lawyer; attorneys are most commonly spouses which may well be while the donor still has capacity). At
or adult offspring. A person who makes an LPA (the donor) the time the health and welfare LPA is drawn up, the donor
must have the legal capacity to do so at the time he makes must decide whether or not he wishes the attorney to have
it. In essence, the donor of the power must understand that, the authority to refuse life sustaining treatment on his
if he loses capacity, the named attorney will have the power behalf. Both forms of LPA allow the donor to include guid-
to do anything with the donor’s finances that he himself ance for the attorney indicating the way in which he would
could have done prior to losing capacity. Once executed, an like the attorney to act. It is possible to appoint more than
LPA must be registered before it becomes effective. In some one attorney; in that case, the donor must specify whether
jurisdictions this usually happens immediately after the LPA the attorneys are always to act together (jointly) or may act
is signed, and does not indicate that the donor lacks capac- individually (jointly and severally).
ity. In others, the power is only registered once the donor is
regarded as having lost capacity (this is the case with English 30.3.2.2 Advance decisions
Enduring Powers of Attorney [EPA], which were used before
the current Mental Capacity Act came into effect in 2007). An advance decision (sometimes called a living will or
A person who retains the capacity to donate an power advance directive) is a decision made by a person who has
may at the same time lack the capacity to manage his affairs. capacity to refuse a specified medical treatment at some time
This was endorsed in an English High Court ruling in the in the future should he lose his mental capacity to decide
landmark case of two elderly women with dementia (Re K about it. Since no person can dictate to a doctor what treat-
and Re F). The case concerned EPAs, but the implication of ment he must give (a patient can request a treatment but not
the ruling is that in those circumstances the attorney should demand it), advance directives are in effect advance refusals
take over management of the affairs as soon as the power is of treatment. Very importantly, advance decisions are only
executed by the donor and registered. binding if they are valid and applicable to the actual future
For people with dementia who have not donated LPA when circumstances that arise. Although several organizations
competent to do so and who are no longer competent to man- have drafted forms of words that can be used, most juris-
age their affairs or to make an LPA, different countries have dictions do not provide any particular form or process for
different mechanisms for appointing someone to take over making an advance decision. Where there is doubt about
management on their behalf. Typically, the process involves validity or applicability, most doctors are likely to proceed
applying to a court either for a specific decision or to appoint on the basis of best interests rather than follow a path that
a deputy to make decisions on behalf of the incapacitated per- may lead to the person’s death, unless the advance decision
son. Court applications are often more complex, slower and is absolutely clear, unambiguous and applicable.
more expensive than appointing an attorney in advance.
An LPA has the advantage over a court-appointed deputy 30.3.2.3 Values directives
that the donor chooses who does it. However, there is little
doubt that LPAs are sometimes abused, often by adult off- A third way in which a competent person may wish to continue
spring, who cream off the assets for their own advantage. to exercise a degree of autonomy even after he has lost capacity
For a review of elder maltreatment, please see Hirsch and is by means of a values directive in which the person making
Vollhardt (2008). it gives a statement of his values and asks that his doctors and
others adhere to it if he becomes incompetent. Although this
30.3.2 MEDICAL TREATMENT may be helpful in guiding future decision makers, it is very
difficult to write one in such a way that it is binding.
People with dementia are mostly old and are therefore at
high risk of co-morbid physical illness, often requiring 30.3.3 CAPACITY TO MAKE A WILL
consideration of potentially hazardous interventions, such
as surgery. Any medical treatment given against his will In some civil law countries such as France, the disposition of
to a person competent to decide for himself constitutes an one’s estate after death is somewhat restricted, and offspring
assault in law. It is, therefore, essential in such a situation cannot normally be excluded, although this was modified
to determine whether a person with dementia retains the by European Union legislation on cross-border inheritance
capacity to decide whether to accept or refuse treatment. in 2015. In anglophone jurisdictions there is much greater
Legal issues and dementia 311

testamentary freedom, provided the testator is mentally com- that is most frequently contested after the testator’s death.
petent to make a will. Spouses or children may be disinher- Common scenarios are the disinheritance of one child in
ited, although in some jurisdictions, for example, England favour of another, or of a caregiver or charity. The grounds
and Wales, legislation allows provision to be made for those on which such wills are challenged clearly vary, but allega-
spouses or children who are still financially dependent on the tions include the following: inability to recall relatives; false
testator, even if they are excluded from the will. beliefs about relatives or others; and delirium supervening
In most anglophone jurisdictions, the leading case on on dementia.
testamentary capacity is Banks v. Goodfellow the key part A will may also be challenged on grounds of undue
of which states: influence, although this can be extremely difficult to prove
in some jurisdictions, notably the United Kingdom and
It is essential that a testator shall understand some Commonwealth countries, because it is necessary to
the nature of the act [of making a will] and its prove, not just that the testator was influenced in making
effects; shall understand the extent of the prop- a particular disposition of his estate, but that his will was
erty of which he is disposing; shall be able to overborne, against his real wishes, for example by threat or
comprehend and appreciate claims to which he force. The issue of undue influence is discussed in detail by
ought to give effect; and with a view to the lat- Peisah et al. (2009).
ter object no disorder of mind shall poison his Paradoxically it is not necessary to prove coercion in the
affections, pervert his sense of right and pre- equity jurisdiction that deals with contracts made when peo-
vent the exercise of his natural faculties – that ple are alive. In such transactions courts may accept a pre-
no insane delusion shall influence his will in sumption of undue influence, which theoretically, at least,
­disposing of the property and bring about a makes it easier to prove than in contested wills. However,
­disposal of it which, if the mind had been sound, case law in England and Wales (Re Beaney) has established
would not have been made. that where the effect of a lifetime gift is to dispose of the
donor’s only asset of value, the degree of understanding
This judgment can be simplified into four limbs, which required is as high as it is for a will. One might be forgiven
are set out in Box 30.1. for thinking that the law here seeks to avoid disputes over
It is not necessary for a testator (female, testatrix) to undue influence in favour of those over mental capacity.
appreciate the extent of his estate down to the last penny It is the common experience of old age psychiatrists that
(cent), but he must have a general grasp of his affairs. their dementia patients are not only vulnerable to undue
However, the more complex the estate, the higher the influence but also actually are so influenced. However, it is
threshold of capacity on this limb of the legal test. For another thing to prove it in a court of law. In the United
example, let us take again our theoretical case of two indi- States, where the difference between probate and equity
viduals with exactly the same degree of dementia. The first does not exist, it is much easier to prove undue influence.
may be able to understand that he owns his house and has a Psychiatrists are called upon to assess testamentary
few thousand dollars in the bank, and would therefore have capacity in two situations: prospective, i.e. before the patient
sufficient appreciation of his estate. The second may, how- dies; and retrospective, i.e. after death. There would be fewer
ever, lack the capacity to understand the extent of his estate retrospective cases if lawyers were to observe the ­so-called
because it consists of live business interests, complex trusts, golden rule formulated by the late Lord Templeman
joint insurance policies, cross-national share portfolios and (Kenward v. Adams), which (paraphrased) recommends
foreign properties. Nevertheless, it is possible to maximize that lawyers drawing up wills for elderly persons should
capacity on this limb of the Banks v. Goodfellow test, as dis- always seek a medical opinion on the testator’s capacity,
cussed below under assessment of testamentary capacity. however embarrassing it may be to propose it to their client.
The capacity to appreciate the ‘moral claims’ of those who Golden rule assessments are discussed by Jacoby and Steer
might expect to benefit from the testator’s bounty is the one (2007) and Shulman et al. (2009). As with any assessment
of capacity, examinations of prospective testators should
attempt to maximize capacity by an unhurried approach in
BOX 30.1: The Banks v. Goodfellow test a nonthreatening atmosphere. If the potential testator can-
not give a full enough account of his estate, it is legitimate to
A testator must be able to: give him the information, the examiner having been briefed
●● Understand the nature and consequences of the beforehand by the lawyer, and to ask him to commit it to
act of making a will memory. If he can do this for a sufficient length of time
●● Understand the extent of his estate to make competent decisions fulfilling the other limbs of
●● Appreciate who might have a claim on his bounty the Banks v. Goodfellow test, then his understanding of his
(both those to be included and excluded) estate may be adequate. The decision on whether this or any
●● Have no mental disorder and no insane delusion other aspect of the Banks v. Goodfellow test is fulfilled rests
directly affecting the above ultimately with the court. The doctor’s role is to give his
opinion in order to assist the court in reaching its decision.
312 Dementia

Retrospective assessment of the testamentary capac- 30.3.5 CAPACITY TO LITIGATE AND


ity of testators whose wills are subject to legal challenge STAND TRIAL
is more complex because opinions have to be given on
the basis of surviving documents and not examination of Few people with dementia are likely to litigate. In the United
the testator himself. Furthermore, there are often witness Kingdom, the test for capacity to litigate is based on the case
statements from the opposing sides that appear to contra- of Masterman-Lister v. Brutton & Co. [2002], together with
dict each other. It is within neither the remit nor the exper- the MCA. In England and Wales, if a person with dementia
tise of a psychiatrist to decide which statements are true lacks the capacity to litigate, he or she may be represented by
or not, for this is the prerogative of the court. Although the Official Solicitor. Other countries have their own agents
the psychiatrist may be instructed by lawyers acting for one for such purposes.
or other party, his or her duty is exclusively to the court Fitness to plead or to stand trial is a more important
to provide an independent, unbiased opinion of matters issue than capacity to litigate because a small but signifi-
within his or her expertise. Prospective and retrospective cant number of dementia sufferers commit offences, some
assessments of testamentary capacity are discussed in full ­serious, including homicide. In England and Wales, the cri-
by Jacoby (2013). teria for fitness to plead are derived from the leading case of
Most jurisdictions require a testator to have capac- R v. Pritchard and are explained more fully in R v. John M.
ity when he or she gives instructions for drawing up his They are summarized in Box 30.2.
or her will and when he or she executes it, i.e. signs it In the United States, the criteria for competence to
in the presence of witnesses. There are sometimes cases stand trial are based on the case of Dusky v. United States,
where the testator loses the necessary capacity between which states that the defendant requires ‘sufficient pres-
giving instructions and executing the will. In such cases, ent ability to consult with his attorney with a reasonable
the will might be valid if the testator can appreciate degree of rational understanding’ and ‘whether he has a
that he is signing a will drawn up on his or her previous rational as well as factual understanding of the proceed-
instructions, known as the Rule in Parker v. Felgate: see ings against him’.
Jacoby (2013). It is difficult to make general statements about what hap-
pens in practice because of the variation in worldwide juris-
dictions. In the United Kingdom, the authorities are keen
30.3.4 CAPACITY TO DECIDE RESIDENCE to divert dementia sufferers away from the criminal justice
system at least for less serious offences, such as shop lifting.
Problems arise when people with dementia either lack the For more serious offences, fitness to stand trial does become
insight to determine or deny the risks attendant on inde- an issue. Grubin (1991) studied 286 people in England and
pendent life at home. Different countries have different Wales who had been declared unfit to plead between 1976
laws on involuntary committal to another place of resi- and 1988. Ten out of 286 (0.3%) were found to be suffer-
dence, but the psychiatrist, social worker or community ing from dementia, six of whom died in hospital while
nurse may need to take a variety of factors into account still considered unfit to plead. Heinik et al. (1994) studied
when dealing with such situations. Some individuals or a group of elderly offenders referred by courts to a foren-
authorities may lean towards ensuring safety at all costs sic unit in Israel for psychiatric evaluation. Of the 17 with
(public sector authorities are often highly risk-averse), a diagnosis of dementia, several of whom had committed
while others may lean towards taking high levels of risk. violent offences, 9 were considered incompetent to stand
While some families provide a selfless and unstinting level trial, and the same number were considered incompetent
of support to the person with dementia, others may not, or to be sentenced, which is not a disposal available to British
may not be in a position to do so. Motives vary very widely courts. So, of the small proportion of people with demen-
indeed, and may include a wish to keep care costs low, tia who commit serious criminal offences, many are unfit
possibly to protect an inheritance, or a strong p ­ reference
for the perceived safety of a care home. However, as Lord
Justice Munby has put it, ‘What good is it making someone BOX 30.2: Fitness to stand trial in England
safer if it merely makes them miserable?’ (Munby, 2010).
and Wales
Disagreements can often be resolved by patient d ­ iscussion
with the various parties and mutual acceptance of risk.
To be fit to stand trial a defendant must be able to:
Some jurisdictions allow for objections to home care
on the part of the person with dementia to be o ­ vercome ●● Understand the nature of the charge
legally so that, if necessary, the patient’s home can be ●● Decide whether to plead guilty or not
entered without the patient’s permission. Authority to ●● Exercise the right to challenge jurors
enter the house is not the same as authority to insist on ●● Be able to instruct lawyers
providing personal care, and in any case, even with legal ●● Give evidence in his own defence
authority, it is in practice very difficult to provide care to ●● Follow the proceedings in court
an unwilling person in his or her own home.
Legal issues and dementia 313

or incompetent to stand trial and few, if any, recover suf- Hirsch, R.D. and Vollhardt, B.R. (2008). Elder maltreat-
ficiently to face trial. In England and Wales, the effect of a ment. In R. Jacoby, C. Oppenheimer, T. Dening and
finding of unfitness to stand trial is that a trial of the facts A. Thomas (eds.), The Oxford Textbook of Old Age
takes place in which a court will determine whether or not Psychiatry. Oxford, United Kingdom: Oxford University
the events alleged happened. If the person is found to have Press, pp. 731–745.
carried out the act alleged, the court has a wide range of Jacoby R. (2013). Testamentary capacity. In T. Dening and
options for disposal, including the making of a hospital or A. Thomas (eds.), The Oxford Textbook of Old Age
supervision order. Psychiatry, Second Edition. Oxford, United Kingdom:
Oxford University Press, pp. 797–804.
Jacoby, R. and Steer, P. (2007). How to assess capacity to
make a will. British Medical Journal, 335: 155–157.
30.4 DEPRIVATION OF LIBERTY Markson, L.J., Kern, D.C., Annas, G.J. and Glantz, L.H.
(1994). Physician assessment of patient compe-
In European countries that have ratified the European tence. Journal of the American Geriatrics Society,
Convention on Human Rights, article 5, the right to lib- 42: 1074–1080.
erty and security, creates an obligation on the state to Munby, J. (2010). Keynote address. Paper presented at
ensure that, ‘No one shall be deprived of his liberty save Taking Stock: The Mental Health & Capacity Reforms
… in accordance with a procedure prescribed by law’, and conference, Manchester, United Kingdom.
asserts that anyone who is deprived of his or her liberty has Peisah, C., Finkel, S., Shulman, K. et al. (2009). The wills
a right ‘to take proceedings by which the lawfulness of his of older people: Risk factors for undue influence.
or her detention shall be decided speedily by a court and International Psychogeriatrics, 21: 7–15.
his or her release ordered if the detention is not lawful’. Shulman, K.I., Peisah, C., Jacoby, R. et al. (2009).
A very large number of people with dementia (and those Contemporaneous assessment of testamentary capac-
with intellectual disabilities and many other conditions) ity. International Psychogeriatrics, 21: 433–439.
are deprived of their liberty because they are in care homes
or hospitals. In a very important case in the UK Supreme
FURTHER READING
Court (P v. Cheshire West and others), it has been clarified
that a person will be deprived of his or her liberty if he or Harrop-Griffiths, T., Cowen, J., Cooper, C. and Hadden, R.
she is under continuous supervision and control and is not (2014). Dementia and the Law. London: Jordans.
free to leave. It is now necessary in England and Wales that Foster, C., Herring, J. and Doron, I. (eds.) (2014). The Law
if such a person lacks the capacity to decide whether or and Ethics of Dementia. Oxford, United Kingdom: Hart
not to stay there, a formal assessment process must be fol- Publishing.
lowed. If he or she is found to lack capacity, is deprived of Frost, M., Lawson, S. and Jacoby, R. (2015). Testamentary
his liberty, and does not fall within the scope of the Mental Capacity: Law, Practice and Medicine. Oxford: Oxford
Health Act 1983, then formal authorization for the depri- University Press.
vation of liberty must be sought from the local authority.
The regulations governing this process are complex, but are
now established in English law, although the law on this is LEGAL CASES
currently (2016) under review. Other European countries
Banks v. Goodfellow [1870] 5 LR QB 549.
have different approaches to deprivation of liberty, and the
Beaney v. Beaney [1978] 1 WLR 770.
issue does not arise in the United States and other coun-
Dusky v. United States (1960) 362 U.S. 402.
tries, which have not ratified the European Convention on
Kenward v. Adams [1975] The Times (London) 29
Human Rights.
November.
Masterman-Lister v. Brutton & Co. [2002] EWHC 417 (QB).
REFERENCES P v. Cheshire West and Chester Council and another
(Respondents); P and Q (by their litigation friend, the
Grubin, D.H. (1991). Unfit to plead in England and Wales, Official Solicitor) (Appellants) v. Surrey County Council
1976–1988: A survey. British Journal of Psychiatry, (Respondent) [2014] UKSC 19.
158: 540–548. Parker v. Felgate [1883] 8 PD. 171.
Heinik, J., Kimhi, R. and Hes, J. (1994). Dementia and R v. John M [2003] EWCA Crim 3452.
crime: A forensic psychiatry unit study in Israel. Re K and Re F [1988] All ER 358.
International Journal of Geriatric Psychiatry, 9: 491–494. R v. Pritchard (1836) 7 C & P 303.
31
Driving and dementia

AOIFE FALLON AND DESMOND O’NEILL

and cognition (Keay et al., 2009), neurological disease


31.1 INTRODUCTION (Lafont et al., 2008) and cardiovascular disease and reduced
functional status (MacLeod et al., 2014). In addition, licens-
Driving has become an almost indispensable part of ing policies have an important impact: accelerated renewal,
­everyday life for older people worldwide, with falling use mental testing, peripheral vision testing, renewal in person
of public transportation and increasing personal affluence at age 70+ (as opposed to renewal by mail or online) and
leading to greater access to the personal car (OECD, 2001). restricted licensing – have a significant effect on an older
These trends are particularly noticeable in rural areas, driver’s decision to reduce or cease driving (Kullkov and
where the proportion of older people tends to be higher and Kulikov, 2010). While the knowledge base is still slender,
where public transport is less accessible. It is increasingly the issues involved have major practical, ethical and societal
clear that the private car and walking are the predominant implications for patients, family carers and health profes-
forms of transport for not only the general public but also sionals. There are widely differing agendas on the part of
older people in the developed world: even if usage of public those involved: the patient, the carers, the physician, the
transport is increasing among older U.S. citizens, it is from statutory licensing authority and the insurance companies.
a very low baseline and still below 3% of trips (Lynott and Dementia is a largely age-related disorder, and the devel-
Figueiredo, 2011). oped world is experiencing an exponential rise in the pro-
Despite increasing levels of age-related disease and dis- portion of older drivers among the driving population.
ability, older drivers in general compensate and adapt to their In the United States, there were 35 million licensed older
changing circumstances and are the safest demographic drivers in 2011, a 21% increase from 2002. In contrast, the
group on the roads, with even the oft-quoted increased total number of licensed drivers increased by only 9% from
crash rate per kilometre disappearing when allowance is 2002 to 2011. Older drivers made up 16% of all licensed
made for distance travelled (Langford et al., 2008b). This, drivers in 2011, compared with 15% in 2002 (National
and increasing awareness of the negative effects of driv- Highway Traffic Safety Administration, 2014). Over one-
ing cessation on not only the older person (Marottoli et al., third of those aged over 80 in 1990 drove at least once a
1997; Marottoli et al., 2000) but also on others (Curl et al., year in Ontario, Canada (Chipman et al., 1998). Although
2015), has led to an attitudinal shift among clinicians to most drive personal automobiles, this is not exclusively the
consider mobility as the primary concern in driving assess- case. On the one hand, licensing regulations for drivers of
ment, with due regard for safety to the same extent as other public service vehicles and heavy transport vehicles are
demographic groups (Dickerson et al., 2007). nearly always more restricted and use a more algorithmic
The U.S. Transportation Research Board, the approach than for drivers of personal automobiles. On the
Organization for Economic Cooperation and Development other hand, those who develop dementia at a younger age
and the European Conference of Ministers for Transport are more likely to be driving heavy goods or public service
have all prepared reports on ageing and transport (European vehicles, and the abolition of mandatory retirement age in
Conference of Ministers of Transport, 2001; OECD, 2001; the United States has resulted in school bus drivers being
U.S. Department of Transportation, 2003). Poor health is able to continue into their mid-80s. There has been litiga-
strongly correlated with driving cessation, including vision tion concerning this group.

314
Driving and dementia 315

interviews with families did not reveal significant prob-


31.2 WHAT PUBLIC HEALTH ISSUES lems with memory or activities of daily living (Lundberg
ARISE WITH OLDER DRIVERS? et al., 1999). It has been shown that patients with demen-
tia are more likely to restrict their driving in order to avoid
One of the most important, and under-recognized, public complex situations than older patients with normal cogni-
health hazards arising from dementia is the consequence tion (O’Connor et al., 2013). Early retrospective studies of
of loss of mobility for those who stop driving (Taylor and dementia and driving from dementia clinics tend to show a
Tripodes, 2001). One key question about the age of the driv- high risk (Friedland et al., 1988; Lucas-Blaustein et al., 1988;
ing population is whether they add significantly to hazard O’Neill, 1992), whereas those that are prospective and look
on the roads. This question is symptomatic of an ageist at the early stages of dementia show a less pronounced risk
approach to older driver issues; it is likely that a more signif- pattern. In the first 2 years of dementia, the risk approxi-
icant problem is that older people give up driving without mates that of the general population (Drachmann, 1991;
sufficient remediation (Siren et al., 2004). The crash rate for Carr et al., 2000; Eby and Molnar, 2012). The most care-
older drivers for a given period of time is considerably lower fully controlled study of crashes and dementia showed no
than for the driving population as a whole (Langford et al., increase in crash rates for drivers with dementia (Trobe
2008b): in some road tests healthy older drivers perform bet- et al., 1996). Likely causes of this finding include lower
ter than younger controls (Gebers et al., 1993). Older drivers annual mileage and restriction of driving by the patient,
with increasing comorbities are more likely to self-regulate family and physicians.
their driving, whereas comorbities in younger drivers were Extrapolating from special populations may skew risk
shown to be associated with an increase in crash rates (Papa predictions. For example, epilepsy, for which most coun-
et al., 2014). The increased crash rate per distance driven tries have relatively clear-cut guidelines, would seem to
noted in older populations in comparison to middle-aged pose a clear threat to driving ability as viewed from a
controls is not only academic while older people continue clinic setting. However, population-based studies indi-
to drive a lower mileage; it also is a product of driving a low cate that the increased risk is relatively low (Hansotia,
mileage, which is itself intrinsically risky. If younger and 1993; Drazkowski et al., 2003). For cognitive function, in
older people who drive a low mileage are compared, this a population renewing their licences in North Carolina,
apparent increase disappears (Hakamies-Blomqvist et al., the lowest decile had a very modest relative crash risk of
2002). Low mileage exposes them to more dangers per mile 1.5 in the 3 years previous to the cognitive testing (Stutts
than high-mileage drivers as they encounter disproportion- et al., 1998). A somewhat reassuring finding from this
ately more intersections, congestion, confusing visual envi- cohort is that those with the poorest scores for visual and
ronments, signs and signals (Janke, 1991), yet older drivers cognitive function also drove less and avoided high-risk
cope at least as well with this. situations (Stutts, 1998). A reasonable conclusion from
Crashes involving older people are more likely to be fatal these studies is that dementia among drivers is not yet a
by a factor of 3.5 in two-car accidents (Li et al., 2003), reflect- public health problem. Although increasing numbers of
ing the increased frailty and reduced reserve of older adults, older drivers may change this situation, it is also possible
while raising suspicions that automobile design may not be that ‘Smeed’s law’ will operate, whereby increasing num-
tailored for maximum safety of this group (Yoganandan bers of drivers among a defined population are associated
et al., 2007). However, there was a significant decline in fatal with a drop in fatality rates per car (Hakamies-Blomqvist
crash involvement in the United States in those aged 70 and et al., 2005).
older between 1997 and 2008, when compared with driv- There is no evidence that mild cognitive impairment
ers aged 35–54 (Cicchino and McCartt, 2014). In addition to (MCI, termed ‘mild neurocognitive disorder’ under the
this, accident rates for young adults often arise from behav- Diagnostic and Statistical Manual of Mental Disorders,
iour that leads to high-risk situations; older drivers tend to 5th Edition [DSM-5] classification) represents an increased
avoid high-risk situations (Carmel et al., 2014) and have the risk of crash for road users as might be expected from a
lowest proportion of crashes while under the influence of syndrome whose definition includes an absence of func-
alcohol (McGwin and Brown, 1999). tional impairment. Self-assessed driving skills in MCI
are preserved (Okonkwo et al., 2009): of the two studies
that seem to suggest some problems, one study combined
dementia and MCI together, which makes interpreta-
31.3 IS DRIVING WITH DEMENTIA A
tion of the analysis difficult (Frittelli et al., 2009), and the
PUBLIC HEALTH HAZARD? other showed less than optimal performance rather than
substantive impairment (Wadley et al., 2009). The key
The precise contribution of dementia to overall crash haz- issue is to arrange for regular review of patients with MCI
ard is uncertain. Although Johansson et al. (1997) sug- to track for the development of dementia, at which stage
gested a major role for dementia as cause of crashes among the assessment and processes should be adopted for this
older drivers on neuropathological grounds, subsequent condition.
316 Dementia

2015), this ‘failure’ by doctors to acquaint themselves with the


31.4 IS THERE A ROLE FOR ­regulations may reflect distrust of the current official medi-
SCREENING OLDER cal fitness criteria. There may also be an element of ageism by
POPULATIONS OF DRIVERS? which doctors may assume that older patients do not drive.
Drivers may not only be unaware but also may wilfully
Despite the lack of convincing evidence for an older driver ignore medical advice and regulations. In many countries,
‘problem’, ageist policies in many jurisdictions have led to they continue to drive despite failing to comply with regu-
screening programmes for older drivers. In the absence of reli- lations for diabetes, visual disease and automatic implant-
able and sensitive assessment tools, this approach is flawed, as able cardioverter defibrillators (Eadington and Frier, 1988;
illustrated by data from Scandinavia and Australia (Hakamies- McConnell et al., 1991; Finch et al., 1993). However, there
Blomqvist et al., 1996; Langford et al., 2008a). There is no is no evidence that this under-reporting results in any
reduction in the number of older people dying in car crashes increased crash risk!
in jurisdictions with routine screening but an increase in the We have little information on advice given to drivers
number of those dying as pedestrians and cyclists, possibly due with dementia by family physicians or physicians at spe-
in part to removing drivers from their cars unnecessarily. In cialist clinics for the evaluation of dementia. This would be
New Zealand, a biennial on road driving test was introduced of interest because of the wide range of answers given for
for all drivers aged 80 and over in 1999 and remained in place patients with life-threatening arrhythmias (Strickberger
until 2006. Analysis of data over 20 years, from 1989, showed et al., 1991). It has been suggested that family physicians
that there was no significant change in either licensing rates, are becoming more confident in managing uncomplicated
driving rates, injuries or pedestrian activity at the time the test dementia (Hum et al., 2014) but have reported a number of
was in place (Keall and Woodbury, 2014). Denmark added barriers in addressing driving with patients with demen-
a cognitive test to older driver screening that was associated tia, including a lack of resources and caregiver resistance
with an increase in deaths among older unprotected road users (Moorhouse and Hamilton, 2014). In any event, a major
(Siren and Meng, 2012). A more minimalist and less medical shift of emphasis is required by healthcare professionals
approach using very simple measures, such as a vision test and to consider driving ability in the functional assessment of
a written skill examination, may be more helpful (Levy et al., older people.
1995); unfortunately this approach is also associated with a The procedures for intervention after the opportunistic
reduction in the number of older drivers, a possible negative detection of illnesses relevant to driving vary widely. In the
health impact (Levy, 1995). Another approach is opportunis- United Kingdom, the doctor’s duty is to inform the patient
tic health screening, perhaps of those older drivers with traf- that he/she must contact the Driver and Vehicle Licencing
fic violations (Johansson et al., 1996). It remains to be seen Authority (DVLA); direct contact by the doctor with the
whether these and other screening policies reduce mobility DVLA is only allowed if there is evidence of continued driv-
among older people, a practical and civil rights issue of great ing that constitutes a hazard to others and if persuasion
importance. Another problem is the uncertainty about the through other family members and carers has been unsuc-
outcome of screening, as there is currently a limited repertoire. cessful. This contrasts with the position in several states in
the United States and provinces in Canada where the doc-
tor is bound by law to report patients with certain illnesses
to the licensing authorities. This disparity is confusing, but
cross-national comparisons may prove a boon to research-
31.5 PHYSICIANS AND DRIVING
ers who wish to establish the most appropriate methods for
ASSESSMENT screening and reporting of age-related diseases.

Another problem is the relative ignorance of physicians about


the effects of illness on driving. Some of this relates to the pre-
dominantly negative tone of much of the medical regulations 31.6 ASSESSMENT PROCEDURES
for driving (White and O’Neill, 2000), and it is likely that doc-
tors are insufficiently aware of healthcare interventions that A systematic inclusion of a question on transport and driv-
have been shown to improve driving comfort and safety: ing is now mandatory in the comprehensive assessment
examples exist for arthritis, sleep apnoea and cataract (Jones of patients with memory problems (Adler and Silverstein,
et al., 1991; George 2001; Monestam and Wachtmeister, 1997). 2008), and it is necessary to identify whether or not the
Doctors are unaware of the driving habits of their patients patient drives (Bradley et al., 2000); this is important in
when prescribing drugs that may affect driving (Cartwright, view of the relatively low assessment of driving in primary
1990) and also have a patchy knowledge of medical regula- care (Pimlott et al., 2006). In recent years, there has been
tions for driving (O’Neill et al., 1994). As the regulations, an introduction of resources for physicians in an effort to
while improving in content in recent decades, rarely fully increase awareness of this issue. This has been reported to
present the origins of their evidence base (Rapoport et al., reduce avoidance of the topic of driving cessation in the
Driving and dementia 317

primary care setting (Moorhouse and Hamilton, 2014). The is barred from driving throughout the EU and referral to
placing of driving issues in an appropriate therapeutic con- a social worker to plan alternative transportation is appro-
text is the most important task. Rather than focussing on priate. Equally, a mild cognitive defect may only require a
the difficult case of patients who present late with impaired review by the physician and occupational therapist (OT).
driving ability and insight, we must recognize that the The overall interdisciplinary assessment should attempt to
assessment of dementia provides the potential for a range provide solutions to both maintaining activities and explor-
of interventions, one of the most important of which is the ing transport needs. An on-road test is likely to be needed for
establishment of a framework for advance planning in a all but the most mild and the most severe, given the inability
progressive disease. We need to start a process that encom- of off-road tests to accurately predict driving ability. It may
passes an assessment, a commitment to maximizing mobil- demonstrate deficits to a patient or carer who is ambivalent
ity and also an awareness-raising process for the patient about the patient stopping driving. At a therapeutic level,
and carers that the progression of the disease will inevitably team members may be able to help patients come to terms
result in a loss of driving capacity. with losses associated with stopping driving. The OT may be
This latter component has been termed a modified Ulysses able to maximize activities and function and help focus on
contract, after the hero made his crew tie him to the mast preserved areas of achievement, whereas the social worker
on the condition that they did not heed his entreaties to be can advise on alternative methods of transport.
released when seduced by the song of the sirens (Howe, The assessment of fitness to drive should only take place
2000). This process has been incorporated in practice and is after a thorough evaluation of the underlying medical
now widely accepted in dementia care, that is, the diagnostic condition(s). Dementia may coexist with other conditions
disclosure in at least general terms – the patient who drives that affect driving ease and ability. Important components
needs to be told that he/she has a memory problem that is of the history and examination of theoretical relevance to
likely to progress and hamper driving ability. In general, car- the driving task are medication and alcohol use (Doege
ers are fearful of diagnosis disclosure, but older people seem and Engelburg, 1986), perception, cognitive status and psy-
to want to be told if they have this illness. There is evidence chomotor ability. Perception is probably more important
that such a process may facilitate driving cessation by enhanc- than vision. Cognition may be usefully measured by the
ing a therapeutic dimension to disease diagnosis and advance physician using one of the many brief mental status sched-
planning (Bahro et al., 1995). It forms the basis of a useful ules (O’Neill, 1993), and the elements of general clinical
patient and carer brochures from the Hartford Foundation assessment of older drivers for medical fitness to drive are
(Hartford Foundation, 2000) and the CanDRIVE Programme now described on both sides of the Atlantic (Breen et al.,
(Byszewski et al., 2013), which are available online. This type 2007; Carr et al., 2010). Although the Mini-Mental State
of approach also seems to have worked with older, visually Examination (MMSE; see Chapter 5, Screening and assess-
impaired drivers (Owsley et al., 2003). ment instruments for the detection and measurement of
The most useful model of assessment is that of an assess- cognitive impairment) correlates with driving performance
ment cascade (see Figure 31.1) and a helpful scheme has (Odenheimer et al., 1994; Fox et al., 1997), it is not suffi-
been described in the context of the driver licencing system ciently sensitive or specific to be used as a determinant of
in the United Kingdom, which can inform the process for driving ability (Joseph et al., 2014).
other jurisdictions (Carter et al., 2015), although the medi- The basic thrust of the assessment is to catalogue and
cal staffing, pathways and procedures in the UK DVLA are remedy pathologies that may be relevant to driving: vision,
formalized to a degree not seen in most other driver licenc- cognitive function, neurological and musculoskeletal disor-
ing agencies. ders, as well as conditions that may give rise to transient
Not all levels will be required by all patients: except as an loss of consciousness such as diabetes and syncope. In any
exceptional case, a patient with a homonymous hemianopia one illness there may be multiple facets that affect driving:
in Parkinson’s disease, this includes motor, cognitive and
Physician
affective aspects. Each component needs to be maximally
remedied before a final decision is made.
The patient’s own assessment of driving should be
Occupational therapist assessed and a promising approach is the Adelaide Self-
Efficacy Scale (George et al., 2007), particularly if used in
terms of a proxy assessment by a family member (George
Neuropsychologist et al., 2007). A history of any previous crashes or traffic
violations is also significant, as it has been shown that a
Specialist driver crash within the past 2 years is a predictor of future crashes
assessor (Joseph et al., 2014). A collateral (witness) history of driving
abilities is important, given the often collaborative nature of
Socialworker driving in later life (Vrkljan and Polgar, 2007). Caregivers
may have concerns about a patient’s driving and conse-
Figure 31.1 An assessment cascade. quently have put driving restrictions in place for the patient
318 Dementia

at an early stage (Mauri et al., 2014), but physicians should controls, information processing and error theory. At a mini-
be cognizant of the conflict of interest of a spouse who does mum, a test battery should contain an informant history of
not drive (Adler et al., 2000). A more formalized informant driving behaviour, a general measure of overall cognitive
driving history, the Fitness to Drive Screening Measure, function, a dementia grading and include tests of attention,
also shows promise (Winter et al., 2015). particularly visual attention, information processing and
Medication review is important: some medications may perception. Some matching to dementia type will be appro-
improve driving skills (antidepressants, anti-inflammatories priate (e.g. attention in subcortical dementias, judgement in
and possibly cholinesterase inhibitors [ChEIs]; Daiello et al., frontotemporal dementia).
2008), while others pose hazards – in the case of neurolep- Specific tests that show statistical (but not clinically use-
tics, almost certainly as a marker of the nature and severity ful!) correlation with driving ability in more than one study
of the underlying illness (Brunnauer et al., 2004). The signifi- include the MMSE, the Trail Making Test (Staplin et al.,
cance of benzodiazepines is uncertain (O’Neill, 1998). 2014) and a range of tests of visual attention, including the
Useful Field of View (Myers et al., 2000), a composite mea-
sure of pre-attentive processing, incorporating speed of
visual information processing, ability to ignore distractors
31.7 FURTHER ASSESSMENT (selective attention) and ability to divide attention (Reger
et al., 2004). In a retrospective study, the Assessment of
In cases of very mild or very severe cognitive impairment, the Driving Related Skills (ADReS) as recommended for use
judgement may be relatively easy and require little supplemen- in older drivers by the American Medical Association was
tary testing. For those falling in between, the precise nature shown not to be associated with a history of crash in the
of the further evaluation is not yet standardized, but neither previous 2 years, though it was acknowledged that prospec-
is the neuropsychological evaluation of dementia. A helpful tive analysis is required (Woolnough et al., 2013). A range of
nosology of the type of the skills required at different levels of other tests has been assessed in single studies (including traf-
specialization of the driving assessment has been described fic sign recognition [Carr et al., 1991]) and a comprehensive
in 2014 (Lane et al., 2014). Physicians may need to invoke review is available from the U.S. National Highway Traffic
the assistance of a driving specialist centre, the components Safety Administration (Staplin et al., 1999). In conjunction
of which are medical, OT, sometimes neuropsychology and with the clinical assessment and collateral history, these tests
­specialist driving assessors. A Canadian study has suggested will help to decide which patients require on-road testing, as
that there is much variability between fitness to drive centres, well as those who are likely to be dangerous to test.
both in relation to testing, numbers of assessments completed The use of simulators to assess the driving capabilities of
and cost (Vrkljan et al., 2013) and so it is important for doctors patients with dementia has not been widely accepted, but
to be familiar with what is available in their local area. A first is a potentially useful research tool (Uc and Rizzo, 2008).
effort may be made with a suitably trained OT: postgraduate Simulators may have a more important role in driver reha-
courses in driver assessment are now organized in a number of bilitation of those with neurological disease (Akinwuntan
countries (Unsworth, 2007). et al., 2005). Older people may be more prone to motion
The choice of tests will be relatively arbitrary and, as in any sickness in driving simulators (Golding, 2006).
memory clinic where doctors will familiarize themselves with
the battery of tests carried out by the OT or psychologist in the
local area, it is likely that good communication between pro-
fessionals and familiarity with the chosen tests is as important
31.8 ON-ROAD TESTING
as the precise tests chosen. The relatively poor clinical utility of
cognitive scales in determining fitness to drive (Molnar et al., Given the poor correlation of nearly all psychometric tests
2007; Hoggarth et al., 2013) relates to their lack of congruity with driving ability, a low threshold for an on-road assess-
to current models of driving behaviour and capabilities, and ment is advisable. In the United Kingdom, assessments are
in particular the concept of risk homeostasis (Fuller, 2005). available from the Forum group of driver assessment cen-
This does not mean that cognition should not be assessed, but tres, with a much broader range of options through state,
rather that such measures should be integrated with other fac- not-for-profit and private organizations in the rest of Europe
tors, including an assessment of insight/anosagnosia, judge- (Sanders et al., 2006). In the United States, the Association
ment skills and strategic thinking. of Driver Rehabilitation Specialists (ADED, www.aded.net)
While a number of screening and evaluation test batteries can provide a list of suitably qualified driving assessors. It
have been proposed, it makes more sense to choose some is important to emphasize to the patient that this test is not
core areas of interest, and to allow some room for clinical the driving test used for learner drivers. Rather, this is an
judgements on areas such as judgement and impulsiveness. assessment to gain insight into both the capabilities and dif-
The task is rendered more complex by the sophistication of ficulties of the driver. A good relationship with a specialist
models of driving, which do not conform easily to traditional driving assessor is important to the assessment process. The
cognitive test batteries. At least five main types of model have assessor will require a full clinical report, and may use a
been explored: psychometric, motivational, hierarchical scoring system for on-road testing of patients with dementia
Driving and dementia 319

(Davis et al., 2012). These include the Washington University have a big effect on lifestyle. Forty-two per cent of older
Road Test (Hunt et al., 1997) and the Alberta Road Test people think that driving is a right as opposed to 27% who
(Dobbs et al., 1998). The authors of the latter have classified think that it is a privilege (AA Foundation for Road Safety
the errors made by drivers with dementia into categories of Research, 1988). However, normal older drivers accept
increasing significance, providing a basis for future studies. that their physician’s advice would be very influential
in deciding to give up driving (AA Foundation for Road
Safety Research, 1988) and many patients with dementia
respond to advice from families or physicians (Adler and
31.9 DECISION MAKING Kuskowski, 2003).
AND INTERVENTIONS For those who resist persuasion, removal of a driving
licence represents a potential breach of civil rights (Reuben
et al., 1988). The way we deal with driving reflects how we
If the assessment points to safe driving practice, the deci- help patients to deal with the reality of the deficits caused
sion to continue driving entails several components. These by dementia. A more positive approach has been sug-
are as follows: gested, whereby the issue of driving is treated as a part of
a therapeutic programme. A case was described whereby
●● Duration before review the patient’s feelings and fears about giving up driving were
●● Possible restriction explored with him (Bahro et al., 1995). The intervention was
●● Driving accompanied designed with the patient as collaborator rather than patient
●● Licensing authority reporting relationship and by dealing with the events at an emotional rather than
●● Insurance reporting responsibility at an intellectual level. The patient was able to grieve about
the disease and in particular about the loss of his car. This
As dementia is progressive, any declaration of fitness to in turn enabled him to redirect his attention to other mean-
drive should be subject to regular review; one study sug- ingful activities that did not involve driving. Although this
gests that a review period of 6 months is probably reason- approach may be hampered by the dementia, it reflects a
able (Duchek et al., 2003); sooner if any deterioration is more widespread trend towards sharing the diagnosis of
reported by the carer. It is also important to advise patients dementia with the patient.
and their families of the predicted decline in driving ability If this approach is unsuccessful, confidentiality may have
on an individual basis, although 3 years from when the dis- to be broken for a small minority of cases. Most professional
ease becomes clinically obvious may be a reasonable group physician associations accept that the principle of confiden-
average (Breen et al., 2007). Nevertheless, some patients tiality is covered to a degree by a ‘common good’ principle
with mild and slowly progressive dementia appear to retain of protecting third parties when direct advice to the patient
driving skills for longer than this (Ott et al., 2008). Given
is ignored (General Medical Council, 1985; Retchin and
evidence that the crash rate is reduced if the driver is accom-
Anapolle, 1993). Removal of the driving licence is not likely to
panied (Bédard et al., 1996), it may be sensible to advise
have much effect on these patients, and the vehicle may need
driving to when there is someone else in the car, not as a
to be disabled or removed (Donnelly and Karlinsky, 1990) or
codriver but as both an observer and as a means of improv-
contact made with the local police force (some driver licenc-
ing vigilance (Shua-Haim and Gross, 1996). There is prelim-
ing agencies have good linkage with traffic police).
inary evidence that drivers with restricted driving licences
In the event of a decision to advise cessation of driving,
have lower crash rates (Caragata Nasvadi and Wister, 2009).
advice from a social worker is helpful in planning strategies
Patients should be advised to avoid traffic congestion as well
for alternative modes of travel. The availability of transport
as driving at night and in bad weather. The patient and car-
resources within the family or of high-quality alternative
ers should acquaint themselves with local driver licensing
transportation is helpful (Freund and Vine, 2010), as may be
authority requirements as well as the policy of their motor
provision of support groups for driving cessation (Gustafsson
insurance company. All this should be recorded in the med-
et al., 2012). This may be more challenging in rural settings.
ical notes. Except for jurisdictions where there is mandatory
reporting of drivers with dementia, there is no obligation on
the doctor to break medical confidentiality in these cases.
31.11 THE FUTURE

Several developments may attenuate the problems of the


31.10 WHEN DRIVING IS NO LONGER older driver with dementia. At a population level, preven-
POSSIBLE tive strategies for dementia may lessen the burden of cog-
nitive disability and strictures on the prescribing of agents
If the assessment supports driving cessation, patients and such as long-acting benzodiazepines may reduce crash sus-
carers should be told and a social worker consulted to help ceptibility. Improvements in highway design, signage and
maximize transportation options. Giving up driving can Intelligent Transportation Systems may make travel safer
320 Dementia

for all. Improved assessment procedures may also main- dementia toolkit – For persons with dementia (PWD)
tain transport options (Martin et al., 2009), for instance, and caregivers – A practical resource. BMC Geriatrics,
in-vehicle data acquisition technology has been used to 13: 117.
compare drivers with early cognitive impairment to those Caragata Nasvadi, G. and Wister, A. (2009). Do restricted
without over a period of 1–2 months (Eby et al., 2012). driver’s licenses lower crash risk among older driv-
For the individual, we do not know whether cholin- ers? A survival analysis of insurance data from British
esterase inhibitor therapy can help driving skills in early Columbia. Gerontologist, 49 (4): 474–484.
Alzheimer’s disease. There is also a suggestion that cogni- Carmel, S., Rechavi, T.B. and Ben-Moshe, Y. (2014).
tive training with useful field of view can help driving skills Antecedents of self-regulation in driving among older
in non-demented older adults (Ball and Owsley, 1994); some drivers. Ageing and Society, 34 (7): 1097–1117.
form of cognitive rehabilitation may be useful. Finally, older Carr, D.B., Duchek, J. and Morris, J.C. (2000).
drivers can benefit from environmental and technological Characteristics of motor vehicle crashes of drivers with
cueing (Dingus et al., 1997); developments in this technol- dementia of the Alzheimer type. Journal of American
ogy may benefit drivers with cognitive impairment. Geriatric Society, 48 (1): 18–22.
Carr, D., Madden, D., Cohen, H.J. and Jackson, T.W.
(1991). The use of traffic identification signs to identify
REFERENCES drivers with dementia. Journal of American Geriatric
Society, 39: A62.
AA Foundation for Road Safety Research (1988). Motoring Carr, D.B., Schwartzberg, J.G., Manning, L. and
and the Older Driver. Basingstoke, England: AA Sempek, J. (2010). Physician’s Guide to Assessing
Foundation for Road Safety Research. and Counseling Older Drivers, Second Edition.
Adler, G. and Kuskowski, M. (2003). Driving cessation Washington, DC: NHTSA.
in older men with dementia. Alzheimer Disease and Carter, K., Monaghan, S., O’Brien, J. et al. (2015).
Associated Disorders, 17 (2): 68–71. Driving and dementia: A clinical decision pathway.
Adler, G., Rottunda, S., Rasmussen, K. and Kuskowski, M. International Journal of Geriatric Psychiatry, 30 (2):
(2000). Caregivers dependent upon drivers with demen- 210–216.
tia. Journal of Clinical Geropsychology, 6 (1): 83–90. Cartwright, A. (1990). Medicine taking by people aged 65
Adler, G. and Silverstein, N.M. (2008). At-risk drivers with or more. British Medical Bulletin, 46 (1): 63–76.
Alzheimer’s disease: Recognition, response, and refer- Chipman, M.L., Payne, J. and McDonough, P. (1998). To
ral. Traffic Injury Prevention, 9 (4): 299–303. drive or not to drive: The influence of social factors on
Akinwuntan, A.E., De Weerdt, W., Feys, H. et al. (2005). the decisions of elderly drivers. Accident Analysis and
Effect of simulator training on driving after stroke: Prevention, 30 (3): 299–304.
A randomized controlled trial. Neurology, 65 (6): Cicchino, J.B. and McCartt, A.T. (2014). Trends in older
843–850. driver crash involvement rates and survivability in
Bahro, M., Silber, E., Box, P. and Sunderland, T. (1995). the United States: An update. Accident Analysis and
Giving up driving in Alzheimer’s disease – An integra- Prevention, 72: 44–54.
tive therapeutic approach. International Journal of Curl, A.L., Proulx, C.M., Stowe, J.D. and Cooney, T.M.
Geriatric Psychiatry, 10 (10): 871–874. (2015). Productive and social engagement following
Ball, K. and Owsley, C. (1994). Predicting vehicle crashes driving cessation: A couple-based analysis. Research
in the elderly: Who is at risk? In K. Johansson and on Aging, 37 (2): 171–199.
C. Lundberg (eds.), Aging and Driving. Stockholm, Daiello, L.A., Festa, E.K., Ott, B.R. and Heindel, W.C.
Sweden: Karolinska Institutet, pp. 1–2. (2008). Cholinesterase inhibitors improve visual atten-
Bédard, M., Molloy, M. and Lever, J. (1996). Should tion in drivers with Alzheimer’s disease. Alzheimer’s
demented patients drive alone? Journal of American and Dementia, 4 (Suppl. 1): T498.
Geriatric Society, 44: S9. Davis, J.D., Papandonatos, G.D., Miller, L.A. et al. (2012).
Bradley, E.H., Bogardus, S.T., Jr., van Doorn, C. et al. Road test and naturalistic driving performance in
(2000). Goals in geriatric assessment: Are we measur- healthy and cognitively impaired older adults: Does
ing the right outcomes? Gerontologist, 40 (2): 191–196. environment matter? Journal of American Geriatric
Breen, D.A., Breen, D.P., Moore, J.W. et al. (2007). Driving Society, 60 (11): 2056–2062.
and dementia. British Medical Journal, 334 (7608): Dickerson, A.E., Molnar, L.J., Eby, D.W. et al. (2007).
1365–1369. Transportation and aging: A research agenda for
Brunnauer, A., Laux, G., Geiger, E. and Moller, H.J. (2004). advancing safe mobility. Gerontologist, 47 (5): 578–590.
The impact of antipsychotics on psychomotor per- Dingus, T.A., Hulse, M.C., Mollenhauer, M.A. et al.
formance with regards to car driving skills. Journal of (1997). Effects of age, system experience, and
Clinical Psychopharmacology, 24 (2): 155–160. navigation technique on driving with an advanced
Byszewski, A., Aminzadeh, F., Robinson, K. et al. (2013). traveler information system. Human Factors, 39 (2):
When it is time to hang up the keys: The driving and 177–199.
Driving and dementia 321

Dobbs, A.R., Heller, R.B. and Schopflocher, D. (1998). A Professional Conduct and Discipline: Fitness to Practice.
comparative approach to identify unsafe older drivers. London, England: General Medical Council.
Accident Analysis and Prevention, 30 (3): 363–370. George, C.F. (2001). Reduction in motor vehicle collisions
Doege, T.C. and Engelburg, A.L. (eds.) (1986). Medical following treatment of sleep apnoea with nasal CPAP.
Conditions Affecting Older Drivers. Chicago, IL: Thorax, 56 (7): 508–512.
American Medical Association. George, S., Clark, M. and Crotty, M. (2007). Development
Donnelly, R.E. and Karlinsky, H. (1990). The impact of of the Adelaide driving self-efficacy scale. Clinical
Alzheimer’s disease on driving ability: A review. Rehabilitation, 21 (1): 56–61.
Journal of Geriatric Psychiatry Neurology, 3 (2): 67–72. Golding, J.F. (2006). Motion sickness susceptibility.
Drachmann, D.A. (1991). Driving and Alzheimer’s disease. Autonomic Neuroscience Basic and Clinical, 129: 67–76.
Annals of Neurology, 28: 591. Gustafsson, L., Liddle, J., Liang, P. et al. (2012). A driving
Drazkowski, J.F., Fisher, R.S., Sirven, J.I. et al. (2003). cessation program to identify and improve transport
Seizure-related motor vehicle crashes in Arizona and lifestyle issues of older retired and retiring drivers.
before and after reducing the driving restriction International Psychogeriatric, 24 (5): 794–802.
from 12 to 3 months. Mayo Clinic Proceedings, Hakamies-Blomqvist, L., Johansson, K. and Lundberg, C.
78 (7): 819–825. (1996). Medical screening of older drivers as a traf-
Duchek, J.M., Carr, D.B., Hunt, L. et al. (2003). fic safety measure – A comparative Finnish–Swedish
Longitudinal driving performance in early-stage evaluation study. Journal of American Geriatric Society,
dementia of the Alzheimer type. Journal of American 44: 650–653.
Geriatric Society, 51 (10): 1342–1347. Hakamies-Blomqvist, L., Raitanen, T. and O’Neill, D.
Eadington, D.W. and Frier, B.M. (1988). Type 1 diabetes (2002). Driver ageing does not cause higher accident
and driving experience: An eight-year cohort study. rates per km. Transportation Research Part F: Traffic
Diabetic Medicine, 6: 137–141. Psychology and Behaviour, 5 (4): 271–274.
Eby, D.W. and Molnar, L.J. (2012). Cognitive impairment Hakamies-Blomqvist, L., Wiklund, M. and Henriksson, P.
and driving safety. Accident Analysis and Prevention, (2005). Predicting older drivers’ accident involve-
49: 261–262. ment – Smeed’s law revisited. Accident Analysis and
Eby, D.W., Silverstein, N.M., Molnar, L.J. et al. (2012). Prevention, 37 (4): 675–680.
Driving behaviors in early stage dementia: A study Hansotia, P. (1993). Seizure disorders, diabetes mellitus,
using in-vehicle technology. Accident Analysis and and cerebrovascular disease. Considerations for older
Prevention, 49: 330–337. drivers. Clinics in Geriatric Medicine, 9 (2): 323–339.
European Conference of Ministers of Transport (2001). Hartford Foundation (2000). At the Crossroads: A Guide
Report on Transport and Ageing of the Population. to Alzheimer’s Disease, Dementia and Driving.
Paris, France: CEMT. Hartford, CT: Hartford Foundation.
Finch, N.J., Leman, R.B., Kratz, J.M. and Gillette, P.C. Hoggarth, P.A., Innes, C.R., Dalrymple-Alford, J.C. and
(1993). Driving safety among patients with automatic Jones, R.D. (2013). Predicting on-road assessment
implantable cardioverter defibrillators. The Journal of pass and fail outcomes in older drivers with cognitive
the American Medical Association, 270: 1587–1588. impairment using a battery of computerized sensory-
Fox, G.K., Bowden, S.C., Bashford, G.M. and Smith, D.S. motor and cognitive tests. Journal of American
(1997). Alzheimer’s disease and driving: Prediction Geriatric Society, 61 (12): 2192–2198.
and assessment of driving performance. Journal of Howe, E. (2000). Improving treatments for patients who
American Geriatric Society, 45 (8): 949–953. are elderly and have dementia. The Journal of Clinical
Freund, K. and Vine, J. (2010). Aging, mobility, and the Ethics, 11 (4): 291–303.
model T: Approaches to smart community transporta- Hum, S., Cohen, C., Persaud, M. et al. (2014). Role expec-
tion. Generations, 34 (3): 76–81. tations in dementia care among family physicians and
Friedland, R.P., Koss, E., Kumar, A. et al. (1988). Motor specialists. Canadian Geriatrics Journal, 17 (3): 95–102.
vehicle crashes in dementia of the Alzheimer type. Hunt, L.A., Murphy, C.F., Carr, D. et al. (1997). Reliability
Annals of Neurology, 24 (6): 782–786. of the Washington University Road Test. A perfor-
Frittelli, C., Borghetti, D., Iudice, G. et al. (2009). Effects mance-based assessment for drivers with demen-
of Alzheimer’s disease and mild cognitive impairment tia of the Alzheimer type. Archives of Neurology,
on driving ability: A controlled clinical study by simu- 54 (6): 707–712.
lated driving test. International Journal of Geriatric Janke, M.K. (1991). Accidents, mileage and the exag-
Psychiatry, 24 (3): 232–238. geration of risk. Accident Analysis and Prevention, 23:
Fuller, R. (2005). Towards a general theory of driver behav- 183–188.
iour. Accident Analysis and Prevention, 37 (3): 461–472. Johansson, K., Bogdanovic, N., Kalimo, H. et al. (1997).
Gebers, M.A., Romanowicz, P.A. and McKenzie, D.M. Alzheimer’s disease and apolipoprotein E E4 allele in
(1993). Teen and Senior Drivers. Sacramento, CA: older drivers who died in automobile accidents. The
California Department of Motor Vehicles. Lancet, 349: 1143.
322 Dementia

Johansson, K., Bronge, L., Lundberg, C. et al. (1996). Can accidents. Paper presented at the The Older Driver,
a physician recognize an older driver with increased Health and Mobility, Dublin, Ireland, 18 September 1999.
crash risk potential? Journal of American Geriatric Lynott, J. and Figueiredo, C. (2011). How the Travel
Society, 44 (10): 1198–1204. Patterns of Older Adults are Changing: Highlights
Jones, J.G., McCann, J. and Lassere, M.N. (1991). Driving from the 2009 National Household Travel Survey.
and arthritis. British Journal of Rheumatology, 30: Washington, DC: AARP Public Policy Institute.
361–364. MacLeod, K.E., Satariano, W.A. and Ragland, D.R. (2014).
Joseph, P.G., O’Donnell, M.J., Teo, K.K. et al. (2014). The The impact of health problems on driving status
Mini-Mental State Examination, clinical factors, and among older adults. Journal of Transport and Health,
motor vehicle crash risk. Journal of the American 1 (2): 86–94.
Geriatrics Society, 62 (8): 1419–1426. Marottoli, R.A., Mendes de Leon, C.F., Glass, T.A. et al.
Keall, M.D. and Woodbury, E. (2014). An analysis of (1997). Driving cessation and increased depressive
changes in mobility and safety of older drivers associ- symptoms: Prospective evidence from the New Haven
ated with a specific older driver on-road licensing test: EPESE (Established Populations for Epidemiologic
A population study. BMC Public Health, 14: 165. Studies of the Elderly). Journal of the American
Keay, L., Munoz, B., Turano, K.A. et al. (2009). Visual Geriatric Society, 45 (2): 202–206.
and cognitive deficits predict stopping or restrict- Marottoli, R.A., Mendes de Leon, C.F.M., Glass, T.A.
ing driving: The Salisbury Eye Evaluation Driving et al. (2000). Consequences of driving cessation:
Study (SEEDS). Investigate Ophthalmology and Visual Decreased out-of-home activity levels. The Journal of
Science, 50 (1): 107–113. Gerontology Series B: Psychology Sciences and Social
Kullkov, E. and Kulikov, E. (2010). The social and policy Science, 55 (6): S334–S340.
predictors of driving mobility among older adults. Martin, A.J., Marottoli, R. and O’Neill, D. (2009). Driving
Journal of Aging Society Policy, 23 (1): 1–18. assessment for maintaining mobility and safety in driv-
Lafont, S., Laumon, B., Helmer, C. et al. (2008). Driving ers with dementia. Cochrane Database of Systematic
cessation and self-reported car crashes in older driv- Reviews (1): CD006222.
ers: The impact of cognitive impairment and demen- Mauri, M., Sinforiani, E., Cuzzoni, M.G. et al. (2014).
tia in a population-based study. Journal of Geriatric Driving habits in patients with dementia: A report from
Psychiatry Neurology, 21 (3): 171–182. Alzheimer’s disease assessment units in northern Italy.
Lane, A., Green, E., Dickerson, A.E. et al. (2014). Driver Functional Neurology, 29 (2): 107–112.
rehabilitation programs: Defining program models, McConnell, R.A., Spall, A.D., Hirst, L.W. and Williams, G.
services, and expertise. Occupational Therapy Health (1991). A survey of the visual acuity of Brisbane drivers.
Care, 28 (2): 177–187. The Medical Journal of Australia, 155 (2): 107–111.
Langford, J., Bohensky, M., Koppel, S. and Newstead, S. McGwin, G., Jr. and Brown, D.B. (1999). Characteristics
(2008a). Do age-based mandatory assessments reduce of traffic crashes among young, middle-aged, and
older drivers’ risk to other road users? Accident older drivers. Accident Analysis and Prevention, 31 (3):
Analysis and Prevention, 40 (6): 1913–1918. 181–198.
Langford, J., Bohensky, M., Koppel, S. and Newstead, S. Molnar, F.J., Marshall, S.C., Man-Son-Hing, M. et al. (2007).
(2008b). Do older drivers pose a risk to other road Acceptability and concurrent validity of measures
users? Traffic Injury Prevention, 9 (3): 181–189. to predict older driver involvement in motor vehicle
Levy, D.T. (1995). The relationship of age and state license crashes: An emergency department pilot case-control
renewal policies to driving licensure rates. Accident study. Accident Analysis and Prevention, 39 (5):
Analysis and Prevention, 27 (4): 461–467. 1056–1063.
Levy, D.T., Vernick, J.S. and Howard, K.A. (1995). Monestam, E. and Wachtmeister, L. (1997). Impact of cata-
Relationship between driver’s license renewal policies ract surgery on car driving: A population based study
and fatal crashes involving drivers 70 years or older. in Sweden. British Journal of Ophthalmology, 81 (1):
The Journal of the American Medical Association, 274 16–22.
(13): 1026–1030. Moorhouse, P. and Hamilton, L.M. (2014). Not if, but when:
Li, G., Braver, E.R. and Chen, L.H. (2003). Fragility versus Impact of a driving and dementia awareness and edu-
excessive crash involvement as determinants of high cation campaign for primary care physicians. Canadian
death rates per vehicle-mile of travel among older driv- Geriatric Journal, 17 (2): 70–75.
ers. Accident Analysis and Prevention, 35 (2): 227–235. Myers, R.S., Ball, K.K., Kalina, T.D. et al. (2000). Relation of
Lucas-Blaustein, M.J., Filipp, L., Dungan, C. and Tune, L. useful field of view and other screening tests to on-road
(1988). Driving in patients with dementia. Journal of driving performance. Perceptual and Motor Skills, 91
American Geriatric Society, 36 (12): 1087–1091. (1): 279–290.
Lundberg, C., Johansson, K., Bogdanovic, N. et al. (1999). National Highway Traffic Safety Administration (2014).
Follow-up of Alzheimer’s disease and apolipoprotein Traffic Safety Facts: Older Population. Washington,
E E4 allele in older drivers who died in automobile DC: National Highway Traffic Safety Administration.
Driving and dementia 323

O’Connor, M.L., Edwards, J.D. and Bannon, Y. (2013). Self- Shua-Haim, J.R. and Gross, J.S. (1996). The ‘co-pilot’
rated driving habits among older adults with clinically- driver syndrome. Journal of American Geriatric Society,
defined mild cognitive impairment, clinically-defined 44 (7): 815–817.
dementia, and normal cognition. Accident Analysis Siren, A., Hakamies-Blomqvist, L. and Lindeman, M.
and Prevention, 61: 197–202. (2004). Driving cessation and health in older women.
O’Neill, D. (1992). Physicians, elderly drivers, and demen- Journal of Applied Gerontology, 23 (1): 58–69.
tia. Lancet, 339 (8784): 41–43. Siren, A. and Meng, A. (2012). Cognitive screening of
O’Neill, D. (1993). Brain stethoscopes: The use and abuse older drivers does not produce safety benefits.
of brief mental status schedules. Postgraduate Medical Accident Analysis and Prevention, 45: 634–638.
Journal, 69: 599–601. Staplin, L., Gish, K.W. and Sifrit, K.J. (2014). Using cogni-
O’Neill, D. (1998). Benzodiazepines and driver safety. tive status to predict crash risk: Blazing new trails?
Lancet, 352 (9137): 1324–1325. Journal of Safety Research, 48: 19–25.
O’Neill, D., Crosby, T., Shaw, A. et al. (1994). Physician Staplin, L.S., Lococo, K.H., Stewart, J. and Decina, L.E. (1999).
awareness of driving regulations for older drivers. Safe Mobility for Older People Notebook. Washington,
Lancet, 344: 1366–1367. DC: National Highway Traffic Safety Administration.
Odenheimer, G.L., Beaudet, M., Jette, A.M. et al. (1994). Strickberger, S.A., Cantillon, C. and Friedman, R. (1991).
Performance-based driving evaluation of the elderly When should patients with lethal ventricular tachyar-
driver: Safety, reliability, and validity. Journal of rhythymias resume driving? Annals of Internal
Gerontolgy, 49 (4): M153–M159. Medicine, 115: 560–563.
OECD (2001). Ageing and Transport: Mobility Needs and Stutts, J.C. (1998). Do older drivers with visual and cogni-
Safety Issues. Paris, France: OECD. tive impairments drive less? Journal of American
Okonkwo, O.C., Griffith, H.R., Vance, D.E. et al. (2009). Geriatric Society, 46 (7): 854–861.
Awareness of functional difficulties in mild cognitive Stutts, J.C., Stewart, J.R. and Martell, C. (1998). Cognitive
impairment: A multidomain assessment approach. test performance and crash risk in an older driver
Journal of American Geriatric Society, 57 (6): population. Accident Analysis and Prevention, 30 (3):
978–984. 337–346.
Ott, B.R., Heindel, W.C., Papandonatos, G.D. et al. (2008). Taylor, B.D. and Tripodes, S. (2001). The effects of driving
A longitudinal study of drivers with Alzheimer disease. cessation on the elderly with dementia and their caregiv-
Neurology, 70 (14): 1171–1178. ers. Accident Analysis and Prevention, 33 (4): 519–528.
Owsley, C., Stalvey, B.T. and Phillips, J.M. (2003). The Trobe, J.D., Waller, P.F., Cook-Flannagan, C.A. et al. (1996).
efficacy of an educational intervention in promoting Crashes and violations among drivers with Alzheimer
self-regulation among high-risk older drivers. Accident disease. Archives of Neurology, 53 (5): 411–416.
Analysis and Prevention, 35 (3): 393–400. Uc, E.Y. and Rizzo, M. (2008). Driving and neurodegenera-
Papa, M., Boccardi, V., Prestano, R. et al. (2014). tive diseases. Current Neurology and Neuroscience
Comorbidities and crash involvement among younger Reports, 8 (5): 377–383.
and older drivers. PLoS One, 9 (4): e94564. Unsworth, C.A. (2007). Development and current status of
Pimlott, N.J., Siegel, K., Persaud, M. et al. (2006). occupational therapy driver assessment and rehabili-
Management of dementia by family physicians in tation in Victoria, Australia. Australian Occupational
academic settings. Canadian Family Physician, 52 (9): Therapy Journal, 54 (2): 153–156.
1108–1109. U.S. Department of Transportation (2003). Safe Mobility
Rapoport, M.J., Weegar, K., Kadulina, Y. et al. (2015). for a Maturing Society: Challenges and Opportunities.
An international study of the quality of national-level Washington, DC: U.S. Department of Transportation.
guidelines on driving with medical illness. Quarterly Vrkljan, B.H., Myers, A.M., Crizzle, A.M. et al. (2013).
Journal of Medicine, 108 (11): 859–869. Evaluating medically at-risk drivers: A survey of
Reger, M.A., Welsh, R.K., Watson, G.S. et al. (2004). The assessment practices in Canada. Canadian Journal of
relationship between neuropsychological function- Occupational Therapy, 80 (5): 295–303.
ing and driving ability in dementia: A meta-analysis. Vrkljan, B.H. and Polgar, J.M. (2007). Driving, navigation,
Neuropsychology, 18 (1): 85–93. and vehicular technology: Experiences of older driv-
Retchin, S.M. and Anapolle, J. (1993). An overview ers and their co-pilots. Traffic Injury Prevention, 8 (4):
of the older driver. Clinics in Geriatric Medicine, 403–410.
9 (2): 279–296. Wadley, V.G., Okonkwo, O., Crowe, M. et al. (2009). Mild
Reuben, D.B., Silliman, R.A. and Traines, M. (1988). The cognitive impairment and everyday function: An inves-
aging driver. Medicine, policy, and ethics. Journal of tigation of driving performance. Journal of Geriatric
American Geriatric Society, 36 (12): 1135–1142. Psychiatry Neurology, 22 (2): 87–94.
Sanders, N., Hendrix, M., Martin, B. et al. (2006). White, S. and O’Neill, D. (2000). Health and relicenc-
EU MEDRIL Project: Medical Testing for the Driving ing policies for older drivers in the European Union.
Licence. Brussels, Belgium: CIECA. Gerontology, 46 (3): 146–152.
324 Dementia

Winter, S.M., Classen, S. and Shanahan, M. (2015). User Yoganandan, N., Pintar, F.A., Stemper, B.D. et al.
evaluation of the fitness-to-drive screening measure. (2007). Biomechanics of side impact: Injury criteria,
Physical and Occupational Therapy in Geriatrics, aging occupants, and airbag technology. Journal of
33 (1): 64–71. Biomechanics, 40 (2): 227–243.
Woolnough, A., Salim, D., Marshall, S.C. et al. (2013).
Determining the validity of the AMA guide: A histori-
cal cohort analysis of the assessment of driving related
skills and crash rate among older drivers. Accident
Analysis and Prevention, 61: 311–316.
32
Quality of life in dementia: Conceptual and
practical issues

BETTY S. BLACK AND PETER V. RABINS

by both intrapersonal and social-normative criteria, of the


32.1 INTRODUCTION person–environment ­system of the individual’ (Lawton and
Herzog, 1991). He believed that there is a need for a frame
Maximizing quality of life is a primary goal of care for indi- of reference against which an individual’s subjective assess-
viduals who have irreversible dementia, given the current ment can be compared and that often there are consensual
lack of disease-modifying therapies. A major challenge for standards of quality (Lawton, 1997). Lawton suggested that
care providers is determining what makes life as good as subjective aspects of QoL (e.g. psychological well-being)
possible over the course of illness in the face of this chronic, and objective domains (e.g. physical safety) should be
progressive disorder that affects every aspect of life. The assessed in parallel to examine congruence and incongru-
importance of this goal is reflected by the growing research ence between the two approaches.
focus on quality of life (QoL) in dementia and the increase Health-related QoL (HRQoL) – a more narrow
in its use as an outcome measure in intervention studies. ­concept – focuses on aspects of life that are affected by
This chapter examines how QoL in dementia is conceptual- a person’s health conditions and the treatment of those
ized and measured, what we are learning about it and how conditions (Kane, 2003). Although more specific than
future research may advance our understanding of this QoL, HRQoL is also a multidimensional concept that
sometimes elusive concept. refers to one’s social, psychological and physical well-
QoL is an individual’s state of being that is determined being consonant with the individual’s values and culture.
by the evaluation of important aspects of life based on a This construct is broader than concepts such as mood
set of values, goals, experiences and culture. While there or function and is of interest because its primary focus
is general agreement that QoL is a broad multidimen- is on individuals’ subjective global assessment of their
sional concept, there is no consensus on how to define it or situation. Although HRQoL is also conceptualized as a
what aspects of life should be considered when appraising broad construct, most researchers have concluded that it
one’s QoL. For example, the World Health Organization is constructed of several domains, most commonly physi-
(WHO) defines QoL as ‘the individual’s perceptions of cal health, psychological well-being, function and social
their position in life in the context of the culture and activity (Patrick and Erickson, 1993).
value system in which they live, and in relationship to While HRQoL is conceptualized as a global construct
their goals, expectations, standards and concerns’ (WHO and often examined using generic instruments, such as
QoL Group, 1995). However, as Faden and German (1994) the Quality of Well-Being (QWB) scale (Kaplan and Bush,
note, there are multiple valid and competing conceptions 1982) or the Short Form (36) Health Survery (SF-36) (Ware
of the good life, the meaning of QoL changes over a life- and Sherbourne, 1992), many researchers have developed
time and adaptation to circumstances affects one’s per- disease-specific measures because the characteristics of
ception of QoL. diseases have a significant impact on measurement of the
Lawton (1997), perhaps the most influential force in construct. This approach is often advocated for dementia
how QoL is conceptualized and measured, argued that QoL because it ultimately robs a person of the ability to express
can and should be assessed both subjectively and objec- oneself due to the progressive, debilitating nature of most
tively. He defined QoL as ‘the multidimensional evaluation, dementing illnesses and because of the unique value placed

325
326 Dementia

on thought and cognition. Ettema et al. (2005b) defined


dementia-specific QoL as ‘the multidimensional evalua- 32.3 CHALLENGES IN MEASURING
tion of the person-environment system of the individual, QoL IN DEMENTIA
in terms of adaptation to the perceived consequences of the
dementia’. They contend that successful adaptation to the The assessment of QoL in persons with dementia presents
effects of the disease will lead to a sense of well-being. several conceptual and practical challenges (Rabins and
Examining QoL can contribute uniquely to assessing Kasper, 1997). The first is whether there are domains of
the effectiveness of interventions to treat persons with QoL that are unique to dementia. Some of the hallmarks
dementia. Agencies that approve pharmacological agents of dementia are that it impairs memory, it can affect atten-
emphasize cognitive improvement as a necessary attribute tion, insight, judgment, problem solving, behaviour, person-
for drugs approved to treat cognitive disorders. However, ality, communication skills and lead to other non-cognitive
since these diseases also universally impair function and symptoms (e.g. agitation, anxiety, depression, delusions,
social capacity, it is plausible that effective treatments hallucinations) (Rabins et al., 2006). This constellation of
might improve non-cognitive aspects of dementia more possible features unique to dementia can markedly influence
than the cognitive aspects and non-biological inter- QoL. Howard and Rockwood (1995) emphasize the need to
ventions may affect well-being more than medications employ QoL measures that discriminate between patterns
(Whitehouse 2006). This suggests that measuring QoL is of symptoms across the illness course. An individual’s envi-
potentially a unique approach for examining treatment ronment also influences functioning and opportunities for
outcomes, and improving QoL through effective interven- social interaction, which impact HRQoL. Measures of QoL
tions may ease some burdens for those with dementia and must be sensitive to the settings where people with demen-
their caregivers. tia reside or receive care, including at home in the commu-
nity, in day-care programs, in assisted living environments
and in nursing homes.
32.2 GOALS OF MEASURING QoL A second issue relates to the subjective nature of the
­construct. In the earlier stages of dementia when symptoms
are mild to moderate and individuals can conceptualize
Measuring QoL can serve several important purposes. this construct and express their opinions, obtaining self-
Fundamentally, determining QoL increases our basic rated QoL is most appropriate, informative and of primary
understanding of the impact that dementia has on indi- interest. There is great value in capturing the perspectives –
viduals over the course of their illness (Kerner et al., both quantitatively and qualitatively – of those who have
1998). Beyond specific aspects of the dementia syndrome, dementia; there is clear evidence that it is feasible to assess
such as cognitive and functional impairments, QoL pro- QoL directly from those in the mild-to-moderate stages
vides a more global indicator of how dementia influences of dementia (Logsdon and Teri, 1997; Brod et al., 1999;
well-being. Because dementia affects a large number of Logsdon et al., 2000; Selai et al., 2001a, 2001b; Trigg et al.,
people, has multiple causes and is non-linear in progres- 2007a); and there is some support for seeking self-rated QoL
sion, QoL provides a common language for evaluating the from individuals with more severe impairments (Hoe et al.,
effects of interventions (Mack and Whitehouse, 2001). As 2005; James et al., 2005). However, since individuals in late
new medications and other therapies become available, stage dementia usually lack the capacity to self-assess QoL,
reliable measures of QoL are important for comparing they must either be excluded from assessment or methods
pharmacological agents in terms of both benefits and side must be used whereby other knowledgeable individuals can
effects and for comparing non-pharmacological inter- provide an indication of the patient’s QoL.
ventions with each other or with pharmacological agents Reliance on others to determine an individual’s QoL
(Rabins and Kasper, 1997). QoL can be a key consider- raises important concerns and questions. Does another
ation in evaluating service programmes or care institu- person have the right to make judgements about an indi-
tions (Brooker, 2005; Kazui et al., 2008) and in developing vidual’s state of well-being? If so, who is the most appropri-
clinical guidelines (NICE, 2006). For those whose lives ate person to serve as a proxy rater of another’s QoL? One
have been altered significantly by moving to long-term of the most consistent findings in the literature on QoL
care facilities, Kane (2003) believes that we have a moral in dementia is that proxy ratings are generally lower than
responsibility to understand and improve QoL in nursing self-ratings (e.g. in Sheehan et al., 2012). A noted exception to
homes and other residential facilities. QoL is also a major this is the finding of Ready et al. (2006) that greater patient
consideration in advanced dementia when decision mak- insight is associated with higher caregiver ratings than
ers are faced with choosing between aggressive interven- patient ratings. Concordance between patients and proxy
tions or palliative approaches to care (Taylor et al., 2008). responses is influenced by the nature/quality of their rela-
Finally, we must keep issues of QoL in mind when allocat- tionship, the amount of time spent together, the objective-
ing scarce resources at both the individual and societal ness of the questions, the patient’s level of impairments and
levels (Brod et al., 1999; Selai and Trimble, 1999; Banerjee the proxy’s well-being (e.g. caregiver burden, depression)
and Wittenberg, 2009). (Brod et al., 1999; Logsdon et al., 2002; Snow et al., 2005;
Quality of life in dementia: Conceptual and practical issues 327

Banerjee et al., 2006; Karlawish et al., 2008b; Conde-Sala that provides subscale scores reflecting specific domains of
et al., 2009; Huang et al., 2009; Moyle et al., 2012; Huang QoL rather than one that provides only a single summary
et al., 2014). Proxy ratings have the drawback of filtering a score. Instruments also vary in how items are scored. In
subjective measure through the opinion of another person many cases, the items of an instrument contribute equally
who may or may not share relevant values. This limitation to either a subscale score and/or the total score, while other
may be unavoidable, especially for those in the later stages measures have weighted scores based on the assumption
of dementia. However, measures consisting of items that that the issues reflected by different items vary in their con-
describe observable behaviours and expressions can help to tribution to the individual’s QoL.
minimize this limitation (Rabins and Kasper, 1997). Characteristics of the study sample can influence the
A third major issue is establishing acceptable psycho- choice of methods and instruments. Some instruments are
metric properties of instruments that measure QoL in a designed to assess QoL across the course of illness. These
disorder in which some individuals are unable to concep- usually rely on proxy raters, such as family members or
tualize the construct. Establishing the validity of a measure formal health care providers, so that individuals with
(i.e. that it accurately measures what it purports to mea- advanced dementia can be included along with those who
sure) is a challenge when there is no universally accepted are less impaired. This approach is particularly useful for
definition of QoL and any definition is subject to an indi- minimizing missing data in longitudinal studies if partici-
vidual’s interpretation and values (Rabins and Kasper, pants are likely to lose the ability to self-assess as the illness
1997). While criterion validity may not be possible, most progresses. Proxy rated instruments can vary in whether
developers seek to establish face validity and content valid- the proxy is asked to respond based on what they think the
ity by demonstrating the relevance and comprehensive- person’s QoL is or based on substituted judgement (i.e. what
ness of their instrument. Reliability of a measure can more they think the person with dementia would say). Other
readily be established than validity by demonstrating that instruments have been developed specifically for those with
its items have internal consistency, that it has good repeat- mild-to-moderate dementia who are likely to be able to
ability across administrations and that its use by multiple self-assess their QoL or participate in the assessment pro-
observers produces similar results. Another psychometric cess along with a knowledgeable informant. Some measures
challenge for instruments measuring QoL in dementia is to focus exclusively on QoL in severe or late-stage dementia,
demonstrate its responsiveness or sensitivity to meaningful often relying on formal caregivers as proxy raters or observ-
change. This is a critical characteristic for its usefulness as ers since many of these individuals are cared for in residen-
an outcome measure. tial facilities.
Lawton (1997) rejected the idea that there can be only
one scale of QoL since life is composed of many facets.
Beyond the subjective measure of QoL involving self-report,
32.4 APPROACHES TO MEASURING he described two types of objective measures that deal with
QoL observable phenomena (Lawton, 1997). First are attri-
bute ratings, which are characteristics rated based on the
Numerous approaches have been used to assess QoL in rater’s familiarity with or observation of the person’s typi-
people with dementia. Differences relate to the general type cal behaviour over a period of time (e.g. the prior 2 weeks).
of instrument (i.e. generic vs. disease-specific), the scope of Instruments examining attributes (e.g. functional health,
the instrument (e.g. a single item vs. multiple domains), the behavioural symptoms, social interaction and affect) are
type of scores the measure produces and the data collec- typically rated by a family member or health care provider.
tion method. The approach selected by an investigator will The second type of objective measure is direct observation
depend on several factors. First, the purpose for measuring and coding of ongoing behaviour, which can include behav-
QoL may determine whether a generic or disease-specific iours of persons with dementia, their displays of affect and
instrument is most appropriate. While a disease-specific interactions with care providers, or it can include aspects
measure of QoL is most sensitive to the unique character- of the environment. Trained researchers typically complete
istics of dementia and its impact on people’s lives, it cannot observational measures.
be used to compare the well-being of individuals who have Concerns about the high cost of dementia care increas-
dementia with that of people who are cognitively normal or ingly have led to reliance on utility-based measures of QoL
who have other illnesses. Naglie (2007) notes that generic that are used to conduct cost-effectiveness analyses (Naglie
measures can be classified as health profiles or utility mea- et al., 2006). Generic utility instruments provide a global
sures and can be helpful for health policy decision making. measure of an individual’s preference for a health state
If QoL is being used as an outcome measure, it is impor- using a single number usually ranging from 0 (representing
tant to consider whether the instrument’s content will pro- death) to 1 (representing perfect health) and allow for evalu-
vide an appropriate measure of effectiveness of the therapy, ating interventions based on their cost per quality-adjusted
intervention or programme being examined. Investigators life year (QALY) (Naglie, 2007). Thus, QALY is a measure
wanting to examine relationships between an intervention of disease burden that incorporates both the quality and
and particular aspects of QoL may prefer an instrument quantity of life lived and is based on the relative desirability
328 Dementia

(‘utility’) of ­different outcomes (Knapp and Mangalore, used to determine the frequency of, ­opportunity for and
2007). Utilities can be obtained directly from individuals ­enjoyment of activities that take place either o ­ utside the
or from health indexes that incorporate a health state clas- home (5 items) or indoors (10 items). Affect is ­measured
sification ­system and a set of ­population-derived weights using Lawton’s Affect Rating scale (Lawton, 1994), which
that produce a utility score. Two frequently used indices consists of three positive affects (pleasure, interest,
for measuring health preferences are the EuroQOL Five ­contentment) and three negative effects (anger, anxiety,
Dimensions questionnaire (EQ-5D) (Rabin and de Charro depression). The frequency of each affect in the previous
2001) and the Health Utility Index (HUI) (Horsman et al., 2 weeks is determined based on a 5-point scale. A c­ omposite
2003), both of which have been used to assess QoL in p ­ ersons QoL indicator is then ­constructed by ­combining the affect
with ­dementia (Jonsson et al., 2006; Naglie et al., 2006; and activity i­ ndicators. While the developers have reported
Karlawish et al., 2008b; Kavirajan et al., 2009; Miller et al., that reliability and validity of the AAIQoL are acceptable
2009; Hounsome et al., 2011). The EQ-5D measures health (Albert et al., 1996) and that it can be used in clinical and
states according to five d ­imensions (mobility, s­elf-care, community-based samples, they suggest that the affect
usual activities, ­ pain/discomfort and anxiety/depres- measure, which is less clearly linked to observed behav-
sion), and a later version of the HUI (HUI3) includes eight iours, may be a less reliable indicator of QoL when reported
­attributes (vision, h­ earing, speech, a­ mbulation, ­dexterity, by proxies (Albert et al., 2000).
emotion, cognition and pain). Knapp and Mangalore (2007)
provide a ­discussion of the a­ dvantages and ­disadvantages of
using the QALY ­measurement approach. 32.5.2 ALZHEIMER’S DISEASE–RELATED
QUALITY OF LIFE (ADRQL)
The Alzheimer’s Disease-Related Quality of Life (ADRQL)
is a proxy rated measure of QoL of individuals who have
32.5 MEASURES OF QoL IN DEMENTIA AD or other types of dementia for use across all levels of
disease severity (Rabins et al., 2000). It was developed using
While many investigators use generic or utility-based an iterative process involving input from health care pro-
­measures to examine QoL in dementia, the nine i­ nstruments viders, informal caregivers and a national panel of experts
described below and summarized in Table 32.1 are ­examples in dementia research and treatment. The original ADRQL
of dementia-specific measures for ­assessing QoL in people contains 47 items reflecting five domains that describe pri-
with dementia. This is not an exhaustive list, but it includes marily observable behaviours. It is administered using a
many of the instruments represented in the current l­ iterature structured interview format to either a formal or informal
on QoL in dementia, and it illustrates the v­ ariability that care provider who has knowledge of the person with demen-
exists in the content (i.e. domains, number of items) and data tia in the previous 2 weeks, with response choices of ‘Agree’
collection methods. For example, other instruments used or ‘Disagree’. One assumption underlying the ADRQL is
to measure dementia-related QoL that are not described that the items and domains vary in their contribution to
in detail here are the Pleasant Events ­Schedule-Alzheimer’s the concept of QoL. These differences are incorporated
Disease (PES-AD; Logsdon and Teri, 1997), the Quality of into the measure with preference weights to assign a scale
Life Assessment Schedule (QoLAS; Selai et al., 2001b), the value to each ADRQL item. Summary scores ranging from
Quality of Life Questionnaire for Dementia (QoL-D, Terada 0 to 100 are calculated for each domain and for the overall
et al., 2002) and the Bath Assessment of Subjective Quality of QoL score, with higher scores reflecting a higher QoL. The
Life in Dementia (BASQID; Trigg et al., 2007a). For selected ADRQL has good reliability and validity (Black et al., 2000;
reviews of generic and dementia-specific instruments, Gonzalez-Salvador et al., 2000) and is responsive to change
Walker et al. (1998), Demers et al. (2000), Salek et al. (1998), (Lyketsos et al., 2003; Missotten et al., 2007; Kasper et al.,
Selai and Trimble (1999), Ready and Ott (2003), Ettema 2009). The 40-item version of the ADRQL is recommended
et al. (2005a), Schölzel-Dorenbos et al. (2007), Banerjee et al. based on analyses of data from three settings (community,
(2009), Perales et al. (2013) and Bowling et al. (2015). An assisted living, nursing home) that demonstrate improved
online database of QoL ­instruments is available at www.­ psychometric properties over the original version (Kasper
proqolid.org. et al., 2009). The 40-item ADRQL can be derived from the
original 47 items and reflects the same five domains of QoL.

32.5.1 ACTIVITY AND AFFECT


INDICATORS OF QUALITY OF LIFE 32.5.3 CORNELL-BROWN SCALE FOR
QoL IN DEMENTIA
Albert et al. (1996, 2000) developed the Activity and Affect
Indicators of Quality of Life (AAIQoL) by adapting two Ready et al. (2002) developed the Cornell-Brown Scale
existing measures to assess QoL related to the domains (CBS) by modifying the Cornell Scale for Depression
of activity and affect as rated by caregivers. The 53-item (Alexopoulos et al., 1988). The CBS was developed based
PES-AD (Logsdon and Teri, 1997) was reduced to 15 items, on the conceptualization that high QoL is indicated by the
Quality of life in dementia: Conceptual and practical issues 329

Table 32.1 Dementia-specific measures of quality of life (QoL)

Instrument Content/Domains Respondent Patient Population Items


Activity and Affect Indicators of Activity Proxy–formal or Mild to severe 21
Quality of Life (AAIQoL) Positive Affect informal
Negative Affect caregiver
Alzheimer’s Disease-Related Social interaction Proxy–formal or Mild to severe 40
Quality of Life (ADRQL) Awareness of self informal
Enjoyment of activities caregiver
Feelings and mood
Response to surroundings
Cornell-Brown Scale (CBS) for Negative affectivity Clinician with Mild to moderate 19
Quality of Life in Dementia Physical complaints patient’s and
Positive affectivity caregiver’s input
Satisfaction
Dementia Care Mapping (DCM) Well-being Trained observer Moderate to severe 24
Illbeing
Personal detractions
Dementia Quality of Life (DQoL) Self-esteem Patient Mild to moderate 29
Positive affect/humour
Negative affect
Feelings of belonging
Sense of aesthetics
DEMQoL Daily activities Patient Mild to moderate 28
DEMQoL-Proxy Health and well-being Proxy Mild to severe 31
Cognitive functioning
Social Relationships
Self-concept
QUALIDEM Care relationship Professional Mild to severe 37
Positive affect caregiver
Negative affect
Restless tense behaviour
Positive self-image
Social relations
Social isolation
Feeling at home
Having something to do
Quality of Life-Alzheimer’s Physical condition Patient and/or Mild to moderate 13
Disease (QoL-AD) Mood proxy
Interpersonal relationships
Ability to participate in
meaningful activities
Financial situation
Overall assessment of self
Life quality as a whole
Quality of Life in Late Stage Activity Proxy Severe 11
Dementia (QUALID) Affect

presence of positive affect, satisfaction, self-esteem and the month, each item is rated on a 5-point scale from −2 to +2.
relative absence of negative affect. It includes 19 bipolar Reliability and validity have been demonstrated in a sam-
items in four categories that yield a single QoL score. The ple of outpatients with either dementia or mild cognitive
CBS is completed by a clinician after conducting a joint impairment, with similar properties based on data from
semi-structured interview with the patient and caregiver. the mild and the more severely impaired halves of the sam-
Based on information obtained regarding the previous ple (Ready et al., 2002).
330 Dementia

32.5.4 DEMENTIA CARE MAPPING multi-staged process beginning with the development


of a conceptual framework that includes five domains
Kitwood and Bredin (1994) devised the Dementia Care (Smith et al., 2005a). The DEMQoL is a 28-item i­ nstrument
Mapping (DCM) method for evaluating quality of care ­administered to ­persons with mild-to-moderate ­dementia,
and the well-being of people with dementia in residen- and the DEMQoL-Proxy is a 31-item questionnaire for the
tial care settings. The DCM methodology is based on the caregiver to complete regarding patients with ­mild-to-severe
social–psychological theory of dementia care (Kitwood dementia, with higher scores i­ndicating b ­etter QoL.
and Bredin, 1992), which holds that much of the decline The authors, who report satisfactory ­ psychometric
among those with dementia is a consequence of social properties on these measures, recommend that both
­
and environmental factors. DCM is a structured, obser- instruments be used together, although the DEMQoL
­
vational method in which the well-being or illbeing (WIB) ­cannot be used with ­individuals who have severe cognitive
of patients is rated based on signs from the patient and impairment (i.e. MMSE scores <10).
staff behaviours toward the patient. Trained observers
also record any personal detractors (PDs), which are any
episodes that could lead to a reduction in self-esteem for 32.5.7 QUALIDEM
the patient, and Positive Enhancers (PEs). Typically, five to
Ettema et al. (2007a, 2007b) developed the QUALIDEM to
ten residents are rated every 5 minutes over a 6-hour time
assess QoL in individuals with dementia living in residen-
period using 24 Behaviour Category Codes (BCC). WIB
tial care settings. Content of the QUALIDEM is based on an
values, which range from −5 (illbeing) to +5 (well-being),
iterative process that involved a literature review, qualitative
can be aggregated to determine an overall WIB score for
research methods, field testing and psychometric analyses.
the group and for each individual. Literature reviews have
It is a multidimensional scale, rated by professional caregiv-
described the extensive use of DCM and summarized
ers, that consists of 37 items describing observable behaviour
evidence of its reliability, validity and responsiveness
and making up nine subscales (Koopmans et al., 2009). The
to change (Beavis et al., 2002; Brooker, 2005). DCM has
subscale scores are linearly transformed to range from 1 to
undergone several changes since its inception (Brooker and
100, with higher scores reflecting a better QoL. The devel-
Surr, 2006). Because of its complexity and required spe-
opers report that 18 of the QUALIDEM items comprising
cialist training, others have examined the use of ‘stream-
six subscales are applicable for measuring QoL in the final
lined’ approaches to mapping in an effort to simplify the
phase of dementia. The QUALIDEM has satisfactory reli-
process and reduce the resources required to implement
ability and validity for use in research and practice. The
this method (Fulton et al., 2006). Despite its limitations
authors strongly discourage the calculation of a total score
(Beavis et al., 2002; Thornton, 2004; Sloane, 2007), DCM is
since the subscales differ in content and distinct subscale
unique among the dementia-specific QoL measures in that
information would be lost.
it also serves to examine some elements of quality of care,
is a tool for practice development and has been used as a
intervention for improving QoL. 32.5.8 QUALITY OF LIFE-AD

32.5.5 DEMENTIA QUALITY OF LIFE The Quality of Life-AD (QoL-AD) was developed by


Logsdon et al. (2000) to assess QoL as perceived by both
Brod et al. (1999) developed the Dementia Quality of Life the patient and their caregiver. The content of the QoL-AD
(DQoL), which was designed to be administered to people was derived based on a review of relevant literature on QoL
with mild to moderately severe dementia. It is a 29-item of older adults and other chronically ill populations and
instrument developed through an iterative process i­ nvolving the input of patients, caregivers and experts in the fields
a literature review, focus groups, pilot testing and ­statistical of geriatrics and gerontology. The QoL-AD consists of
examination of its psychometric p ­roperties. Screening 13 items used to assess seven aspects of the patient’s QoL.
­questions are used to assess the individual’s ­comprehension An interviewer administers the QoL-AD to patients with
of the response format. The DQoL consists of items rated on mild-to-moderate dementia and caregivers complete the
a 5-point visual scale that form five ­subscales. The developers measure in the form of a questionnaire. Each item is rated
(Brod et al., 1999) have reported good internal consistency, by the respondent on a 4-point scale, with 1 being ‘Poor’
reliability and construct validity and suggest that reliable and 4 being ‘Excellent’, and total scores range from 13 to
data can be obtained from individuals with ­Mini-Mental 52. A weighted composite score is calculated by giving
State Examination (MMSE) scores >12. greater weight to the patient’s rating than the caregiver’s
rating. The developers (Logsdon et al., 2000, 2002) report
32.5.6 DEMQoL AND DEMQoL-PROXY that the QoL-AD has good reliability and validity, and it is
responsive to change (Spector et al., 2003; Karlawish et al.,
The DEMQoL and DEMQoL-Proxy were devised by Smith 2004). The QoL-AD is widely used and frequently serves
et al. (2007) to assess HRQoL in all stages of dementia as an indicator of concurrent validity for other dementia-
severity. These instruments were developed based on a specific and generic measures of QoL.
Quality of life in dementia: Conceptual and practical issues 331

32.5.9 QoL IN LATE-STAGE DEMENTIA but there are also exceptions to these findings. For exam-
ple, neither Fuh and Wang (2006) nor Matsui et al. (2006)
Weiner et al. (2000) developed the Quality of Life in found an association between measures of QoL and ADL
­Late-Stage Dementia (QUALID) for use in assessing QoL in impairments, while others report findings in which proxy
persons with late-stage dementia in institutional settings. The ratings of QoL are associated with functional status but self-
scale items were selected from the affect and activity measure ratings are not (Edelman et al., 2004b; Edelman et al., 2005).
(i.e. the AAIQoL) devised by Albert et al. (1996). A group of Based on multiple regression analyses, Giebel et al. (2015)
­clinicians experienced in assessing persons with AD selected found that ADL impairments had a significant impact on
the items by consensus. The QUALID, which is a­ dministered QoL only in moderate dementia. Thus, studies do not reveal
as a structured interview, contains 11 items reflecting a consistent pattern in the relationship between QoL and
­observable behaviours rated by frequency on a 5-point Likert functional status.
scale by a nursing home technician who has at least 30 hours One of the most consistent findings in persons with
of exposure to the resident in the previous week. Scores range dementia is the relationship between symptoms of depres-
from 11 to 55, with lower scores ­reflecting better QoL. The sion and lower QoL (e.g. Smith et al., 2005b; Banerjee et al.,
developers report that the QUALID has good internal consis- 2006; Vogel et al., 2006; Ettema et al., 2007b; Trigg et al.,
tency, adequate construct validity and is responsive to change 2007b; Sheehan et al., 2012; Clare et al., 2014). In some stud-
(Weiner et al., 2000; Martin-Cook et al., 2005). ies, this is reflected by both self-ratings and proxy ratings
of QoL (Logsdon et al., 2002; Snow et al., 2005; Black et al.,
2012). A partial exception to this relationship was reported
by Hoe et al. (2005) who found that individuals with severe
32.6 CORRELATES OF QoL IN
dementia had QoL-AD scores significantly associated with
DEMENTIA the mood scale on the Health Status Questionnaire but not
with the Cornell Scale for Depression in Dementia or the
Through numerous cross-sectional studies, researchers have depression or anxiety subscales of the Hospital Anxiety and
identified a few fairly consistent correlates of QoL, while Depression Schedule. The generally strong relationships
some relationships are less clear. For example, in a review among dementia, depression and QoL highlight the impor-
of findings based on dementia-specific measures, Banerjee tance of diagnosis and treatment of depression, especially
et al. (2009) suggest that there is little relationship between among residents of long-term care facilities where the risk
severity of cognitive impairment and HRQoL. They cite of depression is high.
several studies (Logsdon et al., 2002; Smith et al., 2005a; The relationship between behavioural disorders and
Fuh and Wang, 2006; Vogel et al., 2006) that found little or poor QoL in persons with dementia has been widely
no association between cognition and QoL, but they note reported (e.g. Samus, 2005; Banerjee et al., 2006; Samus,
exceptions to this as well, such as a study by Edelman et al. 2006; Hoe et al., 2007; Beer et al., 2010; Cordner et al.,
(2005) that found QoL-AD scores s­ ignificantly ­worsening as 2010; Black et al., 2012). In a review, Banerjee et al. (2009)
MMSE scores decreased. Findings elsewhere reveal v­ ariable cite studies in which behavioural disorders are associated
relationships between cognitive ­function and QoL, with with decreased QoL based on staff and caregiver ratings but
some investigators identifying significant c­ orrelations (e.g. not based on self-ratings (Logsdon et al., 2002; Hoe et al.,
Samus, 2005, 2006; Falk et al., 2007; Cordner et al., 2010; 2005; Banerjee et al., 2006; Hoe et al., 2006). They suggest
Mougias et al., 2011; Sheehan et al., 2012), others reporting that these ­differences may be due to either the patient’s lack
no significance between the two factors (e.g. Trigg et al., of insight or differential appraisals of the impact that these
2007b; Missotten et al., 2008; Karlawish et al., 2008a) and disorders have for those with dementia compared with
still others reporting that their findings differ based on the proxies. However, some investigators have found signifi-
person’s level of cognitive impairment (i.e. mild vs. moder- cant relationships between behavioural disturbances and
ate vs. severe) (Matsui et al., 2006; Missotten et al., 2009) or QoL as rated by both the caregiver and the patient (Matsui
on who rates the person’s QoL (i.e. self vs. proxy) (Edelman et al., 2006; Hurt et al., 2008). In a review of studies focused
et al., 2004a; Beer et al., 2010). Banerjee et al. (2009) note on persons with dementia in long-term care, Beerens et al.
that, while staff ratings of QoL usually show higher cor- (2013) found associations between proxy-rated QoL and
relations with cognitive ­f unction than ­self-reports or fam- behaviours – especially agitation – based on both ­bivariate
ily caregiver reports, the absolute ­correlation is generally and multivariate analyses. Researchers have also found
low, suggesting that QoL and cognition are independent significantly lower QoL in individuals with dementia who
constructs. are taking psychotropic medications (Albert et al., 1996;
Investigators often find that impairments in activities Gonzalez-Salvador et al., 2000; Ballard et al., 2001; Falk
of daily living (ADL) or greater physical dependency are et al., 2007; Mougias et al., 2011; Clare et al., 2014). However,
associated with diminished QoL in persons with dementia since these are cross-sectional studies, it is unclear whether
(Albert et al., 2000; Gonzalez-Salvador et al., 2000; Logsdon medications adversely affect QoL or whether lower QoL is
et al., 2002; Hoe et al., 2005; Ettema et al., 2007b; Conde- due to the psychiatric and behavioural problems that are
Sala et al., 2009; Black et al., 2012; Sheehan et al., 2012), being treated.
332 Dementia

An increasing number of longitudinal studies have replacement therapy improved QoL in AD, but had no sig-
examined change and correlates of change in QoL. Lyketsos nificant effect on cognition. Phase 1 of the CATIE-AD trial
et al. (2003) identified a small but significant mean change in of three atypical antipsychotics showed some improvement
ADRQL scores over 2 years in a residential setting, with lower in clinical symptoms, but there was no effect on cognition or
baseline scores being the only significant predictor of change QoL (Sultzer et al., 2008). After systematically reviewing 15
in QoL. In a 12-month follow-up examination of QoL-AD pharmacologic interventions that examined change in QoL
scores in a randomized controlled trial (RCT), Karlawish or well-being, Cooper et al. (2013) concluded that there is
et al. (2004) found that declines in functional status were no consistent evidence that any drug improves QoL in per-
more strongly associated with diminished QoL than with sons with dementia. Not included in their review, however,
declines in cognition. Zimmerman et al. (2005) conducted a was a study by Larsson et al. (2011) who conducted a sec-
6-month follow-up of QoL-AD scores in the Dementia Care ondary analysis of a 24-week RCT and found that meman-
project and found a small but significant worsening in QoL tine improved total QoL based on caregiver-rated QoL-AD
that was associated with greater cognitive impairment and scores. More recently, Caramelli et al. (2014) reported that
depression. Over 1 year, Kasper et al. (2009) found a signifi- a 24-week trial of galantamine improved self-rated QoL-AD
cant decline in ADRQL scores in a community-based sample scores in mixed dementia patients. Despite the limited posi-
of individuals who had increases in ADL impairments. Hoe tive findings, drug trials should continue to include QoL as
et al. (2009) showed that change in QoL-AD scores in long- an outcome measure.
term care homes was related to baseline QoL scores, depres- Studies examining a variety of non-pharmacologic
sion and anxiety symptoms and an increase in depressive ­interventions and their effects on QoL have focused on
symptoms and cognitive deterioration during the 20-week those with dementia in a range of settings – the ­community,
follow-up. More recently in a 10-month follow-up study of day-care centres and residential care (Cooper et al., 2012).
nursing home residents, Mjorud et al. (2014) determined A series of studies examined the impact of cognitive
that change in QUALID scores was associated with base- ­stimulation therapy (CST) (Spector et al., 2003; Knapp
line QoL scores and change in neuropsychiatric symptoms. et al., 2006; Woods, 2006). Spector et al. (2003) found that a
Beerens et al. (2014) reported that change in QoL-AD scores 14-session CST programme had a ­significant positive impact
for residents in long-term care facilities was associated with on MMSE and QoL-AD scores of ­persons with dementia.
baseline cognition, functional dependency and depression Other investigators have found that ­ community-based
and with increases in dependency and depressive symptoms occupational therapy can improve patients’ QoL and their
over time. Other investigators have found no significant informal caregivers’ well-being (Dooley and Hinojosa,
mean change in QoL after periods ranging from 6 months 2004; Graff et al., 2007; Kumar et al., 2014). Teri et al.
to 2 years (Hoe et al., 2005; Selwood et al., 2005; Missotten (2005) used the QoL-AD in an RCT to determine whether
et al., 2007; Oudman and Veurink, 2014). However, even in community consultants to family caregivers could reduce
studies that report little or no mean change in QoL, sizeable mood and behaviour problems in persons with AD. Their
proportions of their samples show increases, no change or STAR-C training program ­significantly reduced ­behaviour
decreases in QoL over time (Lyketsos et al., 2003; Selwood problems, improved QoL for the patients and benefited the
et al., 2005; Missotten et al., 2007; Beerens et al., 2014; Mjorud caregivers. Logsdon et al. (2010) found that an early stage
et al., 2014). Further research is needed better to understand memory loss ­support group improved patients’ QoL and
what leads to these fluctuations in QoL. depression. RCTs of ­dementia care ­management (Vickrey
et al., 2006) and care c­oordination (Samus et al., 2014)
models have ­ demonstrated improvements in QoL for
­community-dwelling persons with dementia. Using DCM
32.7 QoL AS AN OUTCOME MEASURE in an RCT, aroma therapy with Melissa oil improved QoL
and reduced agitation in a sample of individuals with severe
Relatively few intervention trials have examined QoL as an dementia (Ballard et al., 2002). DCM was also used by
outcome measure for persons with dementia (Takeda et al., Rokstad et al. (2013) in a 10-month trial to p
­ revent ­agitation
2006; Schölzel-Dorenbos et al., 2007; Banerjee et al., 2009), and other ­ neuropsychiatric symptoms that resulted in
particularly as a primary outcome or with d­ ementia-specific improved QUALID scores in the intervention group.
measures, but their numbers are increasing (Cooper et al., Lai et al. (2004) used DCM and found that r­eminiscence
2012, 2013) as interest grows in improving QoL. To date, ­t herapy had a significant impact on WIB scores for ­nursing
QoL has been examined as an outcome in pharmacologic home residents with ­dementia. Reminiscence therapy was
trials less often than in non-pharmacologic studies. also used by O’Shea et al. (2014), who found ­significantly
Among drug trials, studies conducted by Rogers et al. improved QoL-AD scores in n ­ursing home residents
(1996, 1998a, 1998b) on donepezil showed some improve- based on per-protocol analysis. Other investigators have
ment in QoL. However, the changes they observed in shown some positive effects on QoL for nursing home
QoL were not always significant or seen in the interven- residents with a therapeutic garden (Edwards et al., 2012),
tion group and the rating scales had not been validated for an ­a nimal-assisted intervention (Nordgren and Engstom,
use in dementia. Lu et al. (2006) showed that testosterone 2014) and an awareness-based staff training intervention
Quality of life in dementia: Conceptual and practical issues 333

involving persons with severe dementia (Clare et al., 2013). that impact self-rated QoL measures and what factors con-
The range of non-pharmacologic interventions shown to found QoL ratings provided by patients, informal caregivers
improve QoL in persons with dementia in various care and professional care providers.
settings demonstrates that there are multiple pathways to Most published research on QoL in dementia is based
increase patient well-being in the face of this disorder. on cross-sectional data, so little is known about the ­natural
­history of QoL in dementia (Banerjee et al., 2009) or how
change occurs in QoL (Selwood et al., 2005). Available
longitudinal studies show that decline in QoL is not
­
32.8 QUESTIONS AND ISSUES FOR ­inevitable with disease progression and that QoL may be as
THE FUTURE likely to improve with time as to diminish. More p­ rospective
studies are needed with larger, ­
­ representative samples
Examining QoL in dementia is an important means of that are conducted over longer periods to i­dentify causal
understanding the experience of people with these disor- ­relationships and mediating factors for ­individuals with
ders, determining the efficacy and effectiveness of phar- dementia (Banerjee et al., 2009). Because our k­ nowledge of
macological and non-pharmacological approaches and QoL near the end of life in dementia is limited (Koopmans
evaluating and improving care practices. The advancements et al., 2009), studies are needed to determine the impact
that have occurred over the past two decades in the study of that care settings, approaches to symptom management and
QoL in dementia serve as a foundation for further progress ­end-of-life care decision making have on QoL. Finally, while
that is needed in several areas. These include the measure- progress is being made on developing methods to improve
ment of QoL, identifying predictors of change in QoL and QoL in dementia, there is also a great need for ­replicating
in the development and dissemination of interventions to efficacious interventions and promoting ­ translational
improve patients’ well-being. research to spread the adoption of best practices in care
Reliable and valid dementia-specific instruments are quality that impact QoL for persons with dementia.
available to measure QoL across the spectrum of severity. We anticipate that interest in and the importance of
Many instruments are available in multiple languages and maximizing QoL will increase in the future as the number
some have been validated across cultures. They reflect dif- of individuals with dementia grows and as investigators
ferent conceptualizations of this construct, rely on varied seek to identify effective therapies to modify dementia’s
methods for data collection and obtain the perspectives of effects and ease the suffering of these individuals and
different types of respondents to measure QoL. For some their caregivers.
of these measures, limited or no data are available on their
responsiveness – a psychometric property that is critical REFERENCES
for determining the impact of therapeutic interventions.
Related to this are the questions of how users should inter- Albert, S., Castillo-Castanada, C., Jacobs, D. et al. (2000).
pret changes in QoL scores, and what is a minimally impor- Proxy-reported quality of life in Alzheimer’s patients:
tant difference or a clinically significant change in scores Comparison of clinical and population-based samples.
(Naglie, 2007; Schölzel-Dorenbos et al., 2007). Addressing In S. Albert and R. Logsdon (eds.), Assessing Quality
these issues in future research will facilitate greater reliance of Life in Alzheimer’s Disease. New York: Springer,
on QoL as an outcome measure in RCTs. pp. 69–80.
There is a clear consensus on the importance of obtain- Albert, S.M., Del Castillo-Castaneda, C., Sano, M. et al.
ing self-assessed QoL from individuals capable of concep- (1996). Quality of life in patients with Alzheimer’s
tualizing this construct and responding meaningfully to disease as reported by patient proxies. Journal of the
assessment items. The value of this approach is highlighted American Geriatrics Society, 44: 1342–1347.
by the consistent differences seen between self-rated and Alexopoulos, G.S., Abrams, R.C., Young, R.C. and
proxy-rated measures of QoL; differences that may be attrib- Shamoian, C.A. (1988). Cornell scale for depression in
utable to characteristics of the person with dementia, the dementia. Biological Psychiatry, 23: 271–284.
proxy informant and/or the QoL instrument. Correlates of Ballard, C., O’Brien, J., James, I. et al. (2001). Quality of
HRQoL often depend on who rates an individual’s QoL and life for people with dementia living in residential and
which measure is used (Black et al., 2012). Since the perspec- nursing home care: The impact of performance on
tives of the person with dementia and the carer often differ, activities of daily living, behavioral and psychological
proxy ratings cannot be substituted directly for self-ratings. symptoms, language skills, and psychotropic drugs.
Clarification of criteria is needed for including self-ratings to International Psychogeriatrics, 13: 93–106.
maximize the use and validity of self-assessment; and guid- Ballard, C., O’Brien, J., Reichelt, K. and Perry, E. (2002).
ance is needed to determine the most appropriate proxy for Aromatherapy as a safe and effective treatment for
those who lack the ability to self-assess (Naglie, 2007) since the management of agitation in severe dementia:
the perspectives of informal and formal caregivers can dif- The results of a double-blind, placebo-controlled
fer. Further research should determine the aspects of cog- trial with Melissa. The Journal of Clinical Psychiatry,
nitive impairment (e.g. insight, understanding, judgment) 63: 553–558.
334 Dementia

Banerjee, S., Samsi, K., Petrie, C.D. et al. (2009). What nimodipine on cognitive speed and quality of life in
do we know about quality of life in dementia? A mixed dementia: A 24-week, randomized, placebo-
review of the emerging evidence on the predictive controlled exploratory trial (the REMIX study).
and explanatory value of disease specific measures of Arquivos de Neuro-psiquiatria, 72: 411–417.
health related quality of life in people with dementia. Clare, L., Quinn, C., Hoare, Z. et al. (2014). Care staff
International Journal of Geriatric Psychiatry, 24: 15–24. and family member perspectives on quality of life in
Banerjee, S., Smith, S.C., Lamping, D.L. et al. (2006). people with very severe dementia in long-term care:
Quality of life in dementia: More than just cogni- A cross-sectional study. Health and Quality of Life
tion. An analysis of associations with quality of life Outcomes, 12: 175.
in dementia. Journal of Neurology, Neurosurgery, & Clare, L., Whitaker, R., Woods, R.T. et al. (2013).
Psychiatry, 77: 146–148. AwareCare: A pilot randomized controlled trial of an
Banerjee, S. and Wittenberg, R. (2009). Clinical and cost awareness-based staff training intervention to improve
effectiveness of services for early diagnosis and inter- quality of life for residents with severe dementia in
vention in dementia. International Journal of Geriatric long-term care settings. International Psychogeriatrics,
Psychiatry, 24: 748–754. 25: 128–139.
Beavis, D., Simpson, S. and Graham, I. (2002). A literature Conde-Sala, J.L., Garre-Olmo, J., Turro-Garriga, O. et al.
review of dementia care mapping: Methodological (2009). Factors related to perceived quality of life in
considerations and efficacy. Journal of Psychiatric and patients with Alzheimer’s disease: The patient’s per-
Mental Health Nursing, 9: 725–736. ception compared with that of caregivers. International
Beer, C., Flicker, L., Horner, B. et al. (2010). Factors associ- Journal of Geriatric Psychiatry, 24: 585–594.
ated with self and informant ratings of the quality Cooper, C., Makadam, N., Katona, C. et al. (2012).
of life of people with dementia living in care facili- Systematic review of the effectiveness of non-phar-
ties: A cross sectional study. PLoS ONE, 5: e15621. macological interventions to improve quality of life of
doi:10.1371/journal.pone.0015621 people with dementia. International Psychogeriatrics,
Beerens, H.C., Zwakhalen, S.M., Verbeek, H. et al. (2014). 24: 856–870.
Change in quality of life of people with dementia Cooper, C., Mukadam, N., Katona, C. et al. (2013).
recently admitted to long-term care facilities. Journal Systematic review of the effectiveness of pharmaco-
of Advanced Nursing, 71 (6): 1435–1447. doi:10.111/ logic interventions to improve quality of life and well-
jan.12570 being in people with dementia. American Journal of
Beerens, H.C., Zwakhalen, S.M., Verbeek, H. et al. (2013). Geriatric Psychiatry, 21: 173–183.
Factors associated with quality of life of people with Cordner, Z., Blass, D.M., Rabins, P.V. et al. (2010). Quality
dementia in long-term care facilities: A systematic of life in nursing home residents with advanced
review. International Journal of Nursing Studies, dementia. Journal of the American Geriatrics Society,
50: 1259–1270. 58: 2394–2400.
Black, B.S., Johnston, D., Morrison, A. et al. (2012). Quality Demers, L., Oremus, M., Perrault, A. et al. (2000). Review
of life of community-residing persons with dementia of outcome measurement instruments in Alzheimer’s
based on self-rated and caregiver-rated measures. Disease drug trials: Psychometric properties of func-
Quality of Life Research, 21: 1379–1389. tional and quality of life scales. Journal of Geriatric
Black, B.S., Rabins, P.V. and Kasper, J.D. (2000). Alzheimer Psychiatry and Neurology, 13: 170–180.
Disease Related Quality of Life (ADRQL) User’s Manual. Dooley, N.R. and Hinojosa, J. (2004). Improving ­quality
Baltimore, MD: DEMeasure. of life for persons with Alzheimer’s disease and
Bowling, A., Rowe, G., Adams, S. et al. (2015). Quality of their family caregivers: Brief occupational therapy
life in dementia: A systematically conducted narra- ­intervention. American Journal of Occupational
tive review of dementia-specific measurement scales. Therapy, 58: 561–569.
Aging and Mental Health, 19: 13–31. Edelman, P., Fulton, B.R. and Kuhn, D. (2004a).
Brod, M., Stewart, A.L., Sands, L. and Walton, P. (1999). Comparison of dementia-specific quality of life
Conceptualization and measurement of quality of life ­measures in adult day centers. Home Health Care
in dementia: The dementia quality of life instrument Services Quarterly, 23: 25–42.
(DQoL). The Gerontologist, 39: 25–35. Edelman, P., Fulton, B.R., Kuhn, D. and Chang, C.-H.
Brooker, D.J. (2005). Dementia care mapping: A review of (2005). A comparison of three methods of ­measuring
the research literature. The Gerontologist, 45: 11–18. dementia-specific quality of life: Perspectives of
Brooker, D.J. and Surr, C. (2006). Dementia care map- residents, staff, and observers. The Gerontologist,
ping (DCM): Initial validation of DCM 8 in UK field 45: 27–36.
trials. International Journal of Geriatric Psychiatry, Edelman, P., Kuhn, D. and Fulton, B.R. (2004b). Influence
21: 1018–1025. of cognitive impairment, functional impairment and
Caramelli, P., Laks, J., Palmini, A.L. et al. (2014). Effects care setting on dementia care mapping results. Aging
of galantamine and galantamine combined with and Mental Health, 8: 514–523.
Quality of life in dementia: Conceptual and practical issues 335

Edwards, C.A., McDonnell, C. and Meri, H. (2012). An Hoe, J., Katona, C., Orrell, M. and Livingston, G. (2007).
evaluation of a therapeutic garden’s influence on the Quality of life in dementia: Care recipient and care-
quality of life of aged care residents with dementia. giver perceptions of quality of life in dementia: The
Dementia, 12: 494–510. LASER-AD study. International Journal of Geriatric
Ettema, T.P., Droes, R.M., de Lange, J. et al. (2005a). A Psychiatry, 22: 1031–1036.
review of quality of life instruments used in dementia. Hoe, J., Katona, C., Roch, B. and Livingston, G. (2005). Use
Quality of Life Research, 14: 675–686. of the QoL-AD for measuring quality of life in people
Ettema, T.P., Droes, R.M., de Lange, J. et al. (2007a). with severe dementia – The LASER-AD study. Age and
QUALIDEM: Development and evaluation of a demen- Ageing, 34: 130–135.
tia specific quality of life instrument: Scalability, reli- Horsman, J., Furlong, W., Feeny, D. and Torrance, G.
ability and internal structures. International Journal of (2003). The health utilities index (HUI): Concepts,
Geriatric Psychiatry, 22: 549–556. measurement properties and applications. Health and
Ettema, T.P., Droes, R.M., de Lange, J. et al. (2007b). Quality of Life Outcomes, 1: 1–13.
QUALIDEM: Development and evaluation of Hounsome, N., Orrell, M. and Edwards, T. (2011). EQ-5D
a ­dementia specific quality of life instrument: as a quality of life measure in people with dementia
Validation. International Journal of Geriatric Psychiatry, and their carers: Evidence and key issues. Value in
22: 424–430. Health, 14: 390–399.
Ettema, T.P., Droes, R.M., de Lange, J. et al. (2005b). The Howard, K. and Rockwood, K. (1995). Quality of life in
concept of quality of life in dementia in the different Alzheimer’s disease. Dementia, 6: 113–116.
stages of the disease. International Psychogeriatrics, Huang, H.L., Chang, M.Y., Tang, J.S. et al. (2009).
17: 353–370. Determinants of the discrepancy in patient-
Faden, R. and German, P.S. (1994). Quality of life: and ­caregiver-rated quality of life for persons
Considerations in geriatrics. Clinics in Geriatric with ­dementia. Journal of Clinical Nursing,
Medicine, 10: 541–551. 18: 3107–3118.
Falk, H., Persson, L.O. and Wijk, H. (2007). A psychometric Huang, H.L., Weng, L.C., Tsai, Y.H. et al. (2014). Predictors
evaluation of a Swedish version of the quality of life of self- and caregiver-rated quality of life for
in late-stage dementia (QUALID) scale. International ­people with dementia living in the community and
Psychogeriatrics, 19: 1040–1050. in nursing homes in northern Taiwan. International
Fuh, J.L. and Wang, S.J. (2006). Assessing ­quality of Psychogeriatrics, 27 (5): 825–836.
life in Taiwanese patients with Alzheimer’s ­disease. Hurt, C., Bhattacharyya, S., Burns, A. et al. (2008). Patient
International Journal of Geriatric Psychiatry, and caregiver perspectives of quality of life in demen-
21: 103–107. tia. Dementia and Geriatric Cognitive Disorders,
Fulton, B.R., Edelman, P. and Kuhn, D. (2006). Streamlined 26: 138–146.
models of dementia care mapping. Aging and Mental James, B.D., Xie, S.X. and Karlawish, J.H.T. (2005). How
Health, 10: 343–351. do patients with Alzheimer disease rate their overall
Giebel, C.M., Sutcliffe, C. and Challis, D. (2015). Activities quality of life? American Journal of Geriatric Psychiatry,
of daily living and quality of life across different stage 13 (6): 484–490.
of dementia: A UK study. Aging and Mental Health, Jonsson, L., Andreasen, N., Kilander, L. et al. (2006).
19: 63–71. Patient- and proxy-reported utility in Alzheimer
Gonzalez-Salvador, T., Lyketsos, C.G., Baker, A. et al. disease using the EuroQoL. Alzheimer Disease and
(2000). Quality of life in dementia patients in ­long-term Associated Disorders, 20: 49–55.
care. International Journal of Geriatric Psychiatry, Kane, R.A. (2003). Definition, measurement, and
15: 181–189. ­correlates of quality of life in nursing homes: Toward a
Graff, M.J.L., Vernooij-Dassen, M.J.M., Thijssen, M. et al. reasonable practice, research, and policy agenda. The
(2007). Effects of community occupational therapy Gerontologist, 43: 28–36.
on quality of life, mood health status in dementia Kaplan, R.M. and Bush, J.W. (1982). Health-related quality
patients and their caregivers: A randomized ­controlled of life measurement for evaluation research and policy
trial. Journal of Gerontology Medical Sciences, analysis. Health Psychology, 1: 61–80.
62A: 1002–1009. Karlawish, J., Lu, M., Logsdon, R. et al. (2004). How
Hoe, J., Hancock, G., Livingston, G. and Orrell, M. much do 12 month changes in cognition and ­function
(2006). Quality of life of people with dementia in ­influence changes in mild to moderate patients
­residential care homes. British Journal of Psychiatry, and caregivers ratings of patient quality of life?
188: 460–464. Neurobiology Aging, 25: S325.
Hoe, J., Hancock, G., Livingston, G. et al. (2009). Changes Karlawish, J.H., Zbrozek, A., Kinosian, B. et al. (2008a).
in the quality of life of people with dementia living Preference-based quality of life in patients with
in care homes. Alzheimer Disease and Associated Alzheimer’s disease. Alzheimer’s and Dementia,
Disorders, 23: 285–290. 4: 193–202.
336 Dementia

Karlawish, J.H.T., Zbrozek, A., Kinosian, B. et al. (2008b). Lawton, M.P. (1994). Quality of life in Alzheimer dis-
Caregivers’ assessments of preference-based qual- ease. Alzheimer Disease and Associated Disorders,
ity of life in Alzheimer’s disease. Alzheimer’s and 8: 138–150.
Dementia, 4: 208–211. Lawton, M.P. and Herzog, R.A. (1991). Special research
Kasper, J.D., Black, B.S., Shore, A.D. and Rabins, P.V. methods for gerontology. International Journal of
(2009). Evaluation of the validity and reliability of Aging and Human Development, 33: 113–118.
the Alzheimer disease-related quality of life assess- Logsdon, R., Gibbons, L. and McCurry, S. (2000). Quality
ment instrument. Alzheimer Disease and Associated of life in Alzheimer’s disease patients. In S. Albert
Disorders, 23 (3): 275. and R. Logsdon (eds.), Assessing Quality of Life in
Kavirajan, H., Hays, R., Vassar, S. and Vickrey, B. (2009). Alzheimer’s Disease. New York: Springer Publishing
Responsiveness and construct validity of the Health Company.
Utilities Index in patients with dementia. Medical Care, Logsdon, R.G., Gibbons, L.E., McCurry, S.M. and Teri,
47: 651–661. L. (2002). Assessing quality of life in older adults
Kazui, H., Harada, K., Eguchi, Y.S. et al. (2008). Association with cognitive impairment. Psychosomatic Medicine,
between quality of life of demented patients and 64: 510–519.
professional knowledge of care workers. Journal of Logsdon, R.G., Pike, K.C., McCurry, S.M. et al. (2010).
Geriatric Psychiatry and Neurology, 21: 72–78. Early-stage memory loss support groups: Outcomes
Kerner, D.N., Patterson, T.L., Grant, I. and Kaplan, R.M. from a randomized controlled clinical trial. Journal of
(1998). Validity of the quality of well-being scale for Gerontology Psychological Sciences, 65B: 691–697.
patients with Alzheimer’s disease. Journal of Aging Logsdon, R.G. and Teri, L. (1997). The pleasant events
and Health, 10: 44–61. schedule-AD: Psychometric properties and relation-
Kitwood, T. and Bredin, K. (1994). Evaluating Dementia ship to depression and cognition in Alzheimer’s dis-
Care: The DCM Method, Sixth Edition. Bradford, UK: ease patients. The Gerontologist, 37: 40–45.
University of Bradford, Bradford Dementia Research Lu, P., Masterman, D., Mulnard, R. et al. (2006). Effects of
Group. testosterone on cognition and mood in male patients
Kitwood, T. and Bredin, K. (1992). Towards a theory of with mild Alzheimer disease and healthy elderly men.
dementia care: Personhood and well-being. Aging and Archives of Neurology, 63: 177–185.
Society, 12: 269–287. Lyketsos, C.G., Gonzalez-Salvador, T, Chin J.J. et al.
Knapp, M. and Mangalore, R. (2007). The trouble with (2003). A follow-up study of change in quality of life
QALYs…. Epidemiologia e Psichiatria Sociale, 16: among persons with dementia residing in a long-
289–293. term care facility. International Journal of Geriatric
Knapp, M., Thorgrimsen, L., Patel, A. et al. (2006). Psychiatry, 18: 275–281.
Cognitive stimulation therapy for people with demen- Mack, J.L. and Whitehouse, P.J. (2001). Quality of life in
tia: Cost-effectiveness. British Journal of Psychiatry, dementia: State of the art – Report of the international
188: 574–580. working group for harmonization of the dementia
Koopmans, R.T.C.M., van der Molen, M., Raats, M. and drug guidelines and the Alzheimer’s Society satellite
Ettema, T.P. (2009). Neuropsychiatric symptoms meeting. Alzheimer Disease and Associated Disorders,
and quality of life in patients in the final phase of 15: 69–71.
dementia. International Journal of Geriatric Psychiatry, Martin-Cook, K., Hynan, L.S., Rice-Koch, K. et al. (2005).
24: 25–32. Responsiveness of the quality of life in late-stage
Kumar, P., Tiwari, S.C., Goel, A. et al. (2014). Novel occu- dementia scale to psychotropic drug treatment in
pational therapy interventions may improve quality late-stage dementia. Dementia and Geriatric Cognitive
of life in older adults with dementia. International Disorders, 19: 82–85.
Archives of Medicine, 7: 26. doi:10.1186/1755-7682-7-26 Matsui, T., Nakaaki, S., Murata, Y. et al. (2006).
Lai, C., Chi, I. and Kayser-Jones, J. (2004). A randomized Determinants of the quality of life in Alzheimer’s
controlled trial of a specific reminiscence approach ­disease patients as assessed by the Japanese ­version
to promote the well-being of nursing home ­residents of the quality of life- Alzheimer’s disease scale.
with dementia. International Psychogeriatrics, Dementia and Geriatric Cognitive Disorders, 21:
16: 33–49. 182–191.
Larsson, V., Engedal, K., Aarsland, D. et al. (2011). Quality Miller, E., Schneider, L. and Resenheck, R. (2009). Assessing
of life and the effect of memantine in dementia with the relationship between health utilities, quality of
Lewy Bodies and Parkinson’s disease ­dementia. life, and health services use in Alzheimer’s disease.
Dementia and Geriatric Cognitive Disorders, International Journal of Geriatric Psychiatry, 24: 96–105.
32: 227–234. Missotten, P., Squelard, G., Ylieff, M. et al. (2008).
Lawton, M.P. (1997). Assessing quality of life in Alzheimer Relationship between quality of life and cognitive
disease research. Alzheimer Disease and Associated decline in dementia. Dementia and Geriatric Cognitive
Disorders, 11: 91–99. Disorders, 25: 564–572.
Quality of life in dementia: Conceptual and practical issues 337

Missotten, P., Thomas, P., Squelard, G. et al. (2009). Impact Rabins, P.V., Lyketsos, C.G. and Steele, C.D. (2006).
of place of residence on relationship between quality Practical Dementia Care, Second Edition. New York:
of life and cognitive decline in dementia. Alzheimer Oxford University Press.
Disease and Associated Disorders, 23: 395–400. Rabins, P.V. and Kasper, J.D. (1997). Measuring q ­ uality
Missotten, P., Ylieff, M., Di Notte, D. et al. (2007). of life in dementia: Conceptual and practical issues.
Quality of life in dementia: A 2-year follow-up Alzheimer Disease and Associated Disorders,
study. International Journal of Geriatric Psychiatry, 11: 100–104.
22 (12): 1201–1207. Ready, R.E. and Ott, B.R. (2003). Quality of life measures
Mjorud, M., Rosvik, J., Rokstad, A.M. et al. (2014). for dementia. Health and Quality of Life Outcomes,
Variables associated with change in quality of life 1: 1–9.
among persons with dementia in nursing homes: Ready, R.E., Ott, B.R. and Grace, J. (2006). Insight and
A 10 months follow-up study. PLoS One, 9: e115248. cognitive impairment: Effects on quality-of-life reports
doi:10.1371/journal.pone.0115248 from mild cognitive impairment and Alzheimer’s
Mougias, A.A., Politis, A., Lyketsos, C.G. et al. (2011). ­disease patients. American Journal of Alzheimer’s
Quality of life in dementia patients in Athens, Greece: Disease and Other Dementias, 21: 242–248.
Predictive factors and the role of caregiver-related Ready, R.E., Ott, B.R., Grace, J. and Fernandez, I. (2002).
­factors. International Psychogeriatris, 23: 395–403. The Cornell-Brown Scale for quality of life in ­dementia.
Moyle, W., Murfield, J.E., Griffiths, S.G. et al. (2012). Alzheimer Disease and Associated Disorders,
Assessing quality of life of older people with 16: 109–115.
­dementia: A comparison of quantitative self-report Rogers, S., Doody, R., Mohs, R. et al. (1998a). Donepezil
and proxy accounts. Journal of Advanced Nursing, improves cognition and global function in Alzheimer
68: 2237–2246. disease: A 15-week, double-blind, placebo-controlled
Naglie, G. (2007). Quality of life in dementia. Canadian study. Archives of Internal Medicine, 158: 1021–1031.
Journal of Neurology, 34: S57–S61. Rogers, S., Farlow, M., Doody, R. et al. (1998b). A
Naglie, G., Tomlinson, G., Tansey, C. et al. (2006). 24-week, double-blind, placebo-controlled trial
­Utility-based quality of life measures in Alzheimer’s of donepezil in patients with Alzheimer’s disease.
disease. Quality of Life Research, 15: 631–643. Neurology, 50: 136–145.
NICE (2006). Dementia: Supporting People with Dementia Rogers, S.L., Friedhoff, L.T. and Group TDS. (1996). The
and their Carers in Health and Social Care. London: efficacy and safety of donepezil in patients with
National Institute for Health and Clinical Excellence. Alzheimer’s disease: Results of a US multicentre,
Nordgren, L. and Engstrom, G. (2014). Animal-assisted randomized, double-blind, placebo-controlled trial.
intervention in dementia: Effects on quality of life. Dementia, 7: 293–303.
Clinical Nursing Research, 23: 7–19. Rokstad, A.M., Rosvik, J., Kirkevold, O. et al. (2013). The
O’Shea, E., Devane, D., Cooney, A. et al. (2014). The effect of person-centred dementia care to prevent
impact of reminiscence on the quality of life of agitation and other neuropsychiatric symptoms and
­residents with dementia in long-stay care. International enhance quality of life in nursing home patients: A
Journal of Geriatric Psychiatry, 29: 1062–1070. 10-month randomized controlled trial. Dementia and
Oudman, E. and Veurink, B. (2014). Quality of life in Geriatric Cognitive Disorders, 36: 340–353.
­nursing home residents with advanced dementia: Salek, S.S., Walker, M.D. and Bayer, A.J. (1998). A review
A 2-year follow-up. Psychogeriatrics, 14: 235–240. of quality of life in Alzheimer’s disease: Part 2: Issues
Patrick, D.L. and Erickson, P. (1993). Concepts of in assessing drug effects. Pharmacoeconomics,
­health-related quality of life. In D.L. Patrick and 14: 613–627.
P. Erickson (eds.), Health Status and Health Policy. Samus, Q., Johnston, D., Black, B.S. et al. (2014). A
New York: Oxford University Press, pp. 76–112. ­multidimensional home-based care coordination
Perales, J., Cosco, T.D., Stephan, B.C.M. et al. (2013). intervention for elders with memory disorders: The
Health-related quality-of-life instruments for maximizing independence at home (MIND) pilot
Alzheimer’s disease and mixed dementia. International ­randomized trial. American Journal of Geriatric
Psychogeriatrics, 25: 691–706. Psychiatry, 22: 398–414.
Rabin, R. and de Charro, F. (2001). EQ-5D: A measure Samus, Q., Rosenblatt, A., Onyike, C. et al. (2006).
of health status from the EuroQol Group. Annals of Correlates of caregiver-related quality of life in
Medicine, 33: 337–343. assisted living: The Maryland assisted living study.
Rabins, P., Kasper, J., Kleinman, L. et al. (2000). Concepts Journal of Gerontology Psychological Sciences,
and methods in the development of the ADRQL: An 61B: P311–P314.
instrument for assessing health-related quality of Samus, Q., Rosenblatt, A., Steele, C. et al. (2005). The
life in persons with Alzheimer’s disease. In S. Albert association of neuropsychiatric symptoms and envi-
and R. Logsdon (eds.), Assessing Quality of Life in ronment with quality of life in assisted living residents
Alzheimer’s Disease. New York: Springer, pp. 51–68. with dementia. Gerontologist, 45: 19–26.
338 Dementia

Schölzel-Dorenbos, C.J.M., Ettema, T.P., Bos, J. et al. (2007). Taylor, H.A., Black, B.S. and Rabins, P.V. (2008). Deciding
Evaluating the outcome of interventions on quality of in the best interest of clients with dementia: The
life in dementia: Selection of the appropriate scale. experience of public guardians. The Journal of Clinical
International Journal of Geriatric Psychiatry, 22: 511–519. Ethics, 19: 120–126.
Selai, C. and Trimble, M.R. (1999). Assessing quality of life Terada, S., Ishizu, H., Fujisawa, Y. et al. (2002).
in dementia. Aging and Mental Health, 3: 101–111. Development and evaluation of a health-related
Selai, C., Vaughan, A., Harvey, R.J. and Logsdon, R.G. ­quality of life questionnaire for the elderly with
(2001a). Using the QoL-AD in the UK. International dementia in Japan. International Journal of Geriatric
Journal of Geriatric Psychiatry, 16: 537–542. Psychiatry, 17: 851–858.
Selai, C.E., Trimble, M.R., Rossor, M.N. and Harvey, Teri, L., McCurry, S., Logsdon, R. and Gibbons, L. (2005).
R.J. (2001b). Assessing quality of life in dementia: Training community consultants to help family
Preliminary psychometric testing of the quality of life ­members improve dementia care: A randomized
assessment schedule (QoLAS). Neuropsychological ­controlled trial. The Gerontologist, 45: 802–811.
Rehabilitation, 11: 219–243. Thornton, A. (2004). Dementia care mapping
Selwood, A. Thorgrimsen, L. and Orrell, M. (2005). ­reconsidered: Exploring the reliability and validity
Quality of life in dementia – A one-year follow-up of the observational tool. International Journal of
study. International Journal of Geriatric Psychiatry, Geriatric Psychiatry, 19: 718–726.
20: 232–237. Trigg, R., Jones, R.W. and Skevington, S.M. (2007a). Can
Sheehan, B.D., Lall, R., Stinton, C. et al. (2012). Patient and people with mild to moderate dementia provide
proxy measurement of quality of life among general reliable answers about their quality of life? Age and
hospital in-patients with dementia. Aging and Mental Ageing, 36: 1–7.
Health, 16: 603–607. Trigg, R., Skevington, S. and Jones, R. (2007b). How
Sloane, P. (2007). Dementia care mapping as a research can we best assess the quality of life of people with
tool. International Journal of Geriatric Psychiatry, dementia? The Bath Assessment of Subjective Quality
22: 580–589. of Life in Dementia (BASQID). The Gerontologist,
Smith, S., Lamping, D., Banerjee, S. et al. (2005a). 47: 789–797.
Measurement of health-related quality of life for Vickrey, B.G., Mittman, B.S., Connor, K.I. et al. (2006).
­people with dementia: Development of a new The effect of a disease management intervention on
­instrument (DEMQoL) and an evaluation of current ­quality and outcomes of dementia care. Annals of
methodology. Health Technology Assessment, 9: 1–93. Internal Medicine, 145: 713–726.
Smith, S.C., Lamping, D.L., Banerjee, S. et al. (2007). Vogel, A., Mortensen, E., Hasselbalch, S. et al. (2006).
Development of a new measure of health-related Patient versus informant reported quality of life in the
quality of life for people with dementia: DEMQoL. earliest phases of Alzheimer’s disease. International
Psychological Medicine, 37: 737–746. Journal of Geriatric Psychiatry, 21: 1132–1138.
Smith, S.C., Murray, J., Banerjee, S. et al. (2005b). What Walker, M.D., Salek, S.S. and Bayer, A.J. (1998). A review
constitutes health-related quality of life in dementia? of quality of life in Alzheimer’s disease: Part 1: Issues
Development of a conceptual framework for people in assessing disease impact. Pharmacoeconomics,
with dementia and their carers. International Journal of 14: 499–530.
Geriatric Psychiatry, 20: 889–895. Ware, J.E. and Sherbourne, C.D. (1992). The MOS 36-item
Snow, A., Dani, R., Souchek, J. et al. (2005). Comorbid short-form health survey (SF-36). Medical Care, 30:
psychosocial symptoms and quality of life in patients 473–483.
with dementia. American Journal of Geriatric Weiner, M.F., Martin-Cook, K., Svetlik, D.A. et al. (2000).
Psychiatry, 13: 393–401. The quality of life in late-stage dementia (QUALID)
Spector, A., Thorgrimsen, L., Woods, B. et al. (2003). scale. Journal of the American Medical Directors
Efficacy of an evidence-based cognitive stimulation Association, 1: 114–116.
therapy programme for people with dementia. British Whitehouse, P. (2006). Quality of life: The bridge from the
Journal of Psychiatry, 183: 248–254. cholinergic basal forebrain to cognitive science and
Sultzer, D.L., Davis, S.M., Tariot, P.N. et al. (2008). bioethics. Journal of Alzheimer’s Disease, 9: 447–453.
Clinical symptom responses to atypical antipsychotic WHO QoL Group (1995). WHO Quality of Life-100.
­medications in Alzheimer’s disease: Phase 1 outcomes Geneva: World Health Organization Division of Mental
from the CATIE-AD effectiveness trial. American Health.
Journal of Psychiatry, 165: 844–854. Woods, B. (2006). Improved quality of life and cognitive
Takeda, A., Loveman, E., Clegg, A. et al. (2006). A stimulation therapy in dementia. Aging and Mental
­systematic review of the clinical effectiveness of Health, 10: 219–226.
­donepezil, rivastigmine and galantamine on ­cognition, Zimmerman, S., Sloane, P.D., Williams, C.S. et al. (2005).
quality of life and adverse events in Alzheimer’s disease. Dementia care and quality of life in assisted living and
International Journal of Geriatric Psychiatry, 21: 17–28. nursing homes. The Gerontologist, 45: 133–146.
33
Ethical issues

JULIAN C. HUGHES AND DANIEL STRECH

from the care home, carers may feel that it is better to go


33.1 INTRODUCTION along with this falsehood rather than to contradict the lady
and cause repeated upset.
According to ancient tradition ethics aims at the good life.
It is about how we live our lives well, about doing the right 33.2.1 CONSEQUENTIALISM
thing rather than the wrong thing; it’s about good and bad.
Applied at the level of individuals, decisions about the per- Consequentialists believe that it is the consequences of an
son should be good decisions or the right decisions for that action that determine whether it is right or wrong. In the
individual at that time. The two words ‘ethics’ and ‘morals’ case of therapeutic lying, therefore, they would argue that
are, to all intents and purposes, used synonymously despite if the consequence of the lie means that everyone is hap-
semantic differences. In this chapter, we shall sketch issues pier, then telling the lie would be the right thing to do. It
in dementia care that raise issues of right or wrong, good might be, however, that while the immediate effects of the
or bad. It should be noted, too, that ethics and morals are lie are good, in that the lady is not distressed by the news
not just relevant to personal decisions, they relate to public of her husband’s death, in the long run it may be that she
policy too: ethics is the foundation of politics. comes to distrust people. Her experience will be that the
The chapter will start by rehearsing some of the main reassurance she receives concerning her husband never
ethical theories used to think about ethical dilemmas. We materializes into his appearance. Another possible nega-
then move on to consider the spectrum of ethical issues that tive effect of the lie is that carers might start to feel that, if
arise in clinical dementia care. In the conclusion, we shall they can lie in this case, then they can lie in other cases.
speculate on how ethical decisions are made in practice. Hence, there might be a general lessening of trustworthi-
ness. Society at large, hearing that healthcare and social
care workers frequently tell lies to people with dementia,
might experience a general loss of trust in professionals.
33.2 ETHICAL THEORIES Of course, all of this may not come to pass and it may be
that there are specific instances when the consequences of
The giants of ethical theory are consequentialism, deon- a therapeutic lie seem to be wholly good.
tology and virtue ethics. In the field of medical ethics,
the notion of principlism has also played a dominant role. 33.2.2 DEONTOLOGY
Finally, there is a host of other ethical approaches, which we
shall mention briefly. All of these theories and approaches A deontologist is someone who stresses the importance of
are discussed more fully in Hughes and Baldwin (2006). To duty. Deontology is often linked to the name of Immanuel
highlight the differences between these theories we shall Kant (1724–1804). His idea was that there are certain
use the notion of ‘therapeutic lying’ (Culley et al., 2013). things which rational people should always regard as
Therapeutic lying is the idea that sometimes it can be right imperative. He believed that it would only be rational to
to say to the person with dementia something that is untrue make a rule that should apply to everyone and not just
if this will make life better in some way for the individual. to certain people. He also talked about the importance
For example, if a widow constantly says that she thinks her of not regarding people solely as a means to an end, but
deceased husband will be picking her up later in the day rather as ends in themselves. So, we should not use people

339
340 Dementia

with dementia for research to which they cannot con- 33.2.5 OTHER APPROACHES
sent, which will bring them no benefit. There are some
things, therefore, which Kant would simply have regarded There is a variety of other approaches to thinking and
as duties. One such duty was to tell the truth. His incli- dealing with ethical dilemmas and issues. Feminism, for
nation would be always to repudiate the need for thera- instance, via feminist ethics, has led to the notion of care
peutic lies. He would say that once you allow that lying ethics in which the importance of relationships is empha-
is acceptable then you have undermined a whole lot of sized. Narrative ethics stresses the importance of the per-
social institutions, which rely upon the truth being told. son’s story: both our understanding of and participation in
Contracts between people would be an example of such a it. Hermeneutic ethics stresses the importance of interpre-
social institution, including the long-established contract tation in a given context. Communicative ethics suggests
that doctors keep confidences. that the right decisions will be made if the communication
has been effective and proper. Other approaches stress the
33.2.3 VIRTUE ETHICS importance of rights, community, liberty, liberal individu-
alism and so forth. This is not the place to discuss all such
According to virtue ethics, you will know the good thing approaches in detail (Ashcroft et al., 2007), but they have a
to do if you ask the virtuous person. A person is virtu- number of things in common. They all variously stress the
ous if he or she has the inner dispositions (i.e. the virtues) importance of our interconnectedness, the importance of
to flourish or do well humanly. The virtue words simply social and cultural contexts, the ways in which we depend
describe what it is to do well as a human being. This relies upon each other as human beings engaged in practices in
on the idea that we would all accept that the good person the world (Hughes, 2011).
is the one who is wise, courageous, humble, compassion-
ate and charitable, just, patient, honest and so on. Virtue
ethics changes the subject because it makes us think in 33.3 THE SPECTRUM OF ETHICAL
these different terms. On the face of it, therapeutic lying
ISSUES IN DEMENTIA
will be bad because it involves dishonesty. But, on the
other hand, it may also involve a good deal of compas-
sion. It will require practical wisdom or sensibility in Strech et al. (2013) conducted a systematic review of the
order to decide how the virtues are to be balanced in mak- English and German literature and used qualitative textual
ing these decisions; but the discussion is no longer about analysis to categorize the ethical issues that emerged in clin-
rigid duties or calculations about consequences, instead ical care. They found 56 specific ethical issues, which they
the conversation is about the honest, brave, compassion- then divided into seven major categories. In this section, we
ate and just thing to do. shall outline this spectrum of ethical issues.

33.2.4 PRINCIPLISM 33.3.1 DIAGNOSIS AND MEDICAL


INDICATION FOR TREATMENT
Beauchamp and Childress (2001), on the basis of the more
established ethical theories, picked out what have become First, diagnosis might be made too early or too late. If people
known as the four principles of medical ethics. These are are seeking a diagnosis and it is made too late they will be
respect for autonomy, beneficence, non-maleficence and denied services from which they might benefit. On the other
justice. Decisions will have to be made concerning which hand, it will not always be right to make a diagnosis too
of these principles should be given most weight in any early. With the appearance of mild cognitive impairment
particular circumstance. One immediate concern to do (MCI) and the drive to make earlier diagnoses, in particu-
with therapeutic lying, however, is that it does not seem to lar, to make the diagnosis before dementia starts, there are
respect the person’s autonomy. ‘Autonomy’ means self-rule potential harms to people from inappropriate labelling,
and a lie undermines the person’s ability to self-rule; in fact which would of course seem less concerning if there were a
it undermines the person’s agency and standing as a per- real prospect of doing good. In order to make the diagnosis
son. On the other hand, beneficence, or doing good, might at the appropriate time it will often be very important to
suggest that it is appropriate to lie. Similarly, we may avoid take into account the views and experiences of the family,
harm (i.e. abide by the principle of non-maleficence) if we who can otherwise be ignored during the diagnostic process
tell the therapeutic lie. The notion of justice is normally (Nuffield Council on Bioethics, 2009).
used in connection with resource allocation. It could be Second, there is the risk that signs of illness will be dis-
argued that therapeutic lying, if it calms the person down regarded, which not only means that advance planning is
who might otherwise be agitated, is a much cheaper way, not possible, but again will often leave the relative without
and indeed a safer way to manage the situation than the support. Nevertheless, the psychological burdens associ-
use of drugs. So arguments could be made in favour and ated with receiving a diagnosis have to be kept in mind.
against therapeutic lying using the four principles depend- Third, having made the diagnosis, proper consideration
ing on the circumstances. needs to be given to treatment decisions. There should be no
Ethical issues 341

overestimation of the effect of current drug treatment and If there has been some advance planning, then the role of
both the benefits and harms of any form of treatment need substitute decision makers, often the family, will be easier.
to be weighed up. Once again, the views of relatives must Otherwise, the families might feel decisions to be burden-
be kept in mind. Fourth, throughout all of this process, the some. A possible bad ethical consequence of not making an
patient must be properly regarded as a person, with due early enough diagnosis is that the person is unable to make
weight given to their preferences and to their wishes. That reasonable plans. But advance care plans, particularly where
is, there must be proper respect for autonomy. these are specific and where they refuse treatment, need to
be considered and weighed very carefully from an ethical
33.3.2 DECISION-MAKING CAPACITY point of view. If, for instance, someone has said that they
do not wish to receive antibiotics if they were to be in the
People must be competent to make decisions; that is, they severer stages of dementia, it can be questioned how fully
must have decision-making capacity (see Chapter 30). thought through such a decision might have been: did the
Different jurisdictions will have different legal frameworks person have a realistic appreciation of the effects of such a
for assessing competence or capacity. But there are ethi- decision? ‘Living Wills’ or advance directives will need to
cal issues around the assessment of capacity. For instance, be interpreted correctly and then the decision as to whether
if someone lacks capacity it is important that they should or not they should be followed will require a good deal of
be supported in their decision making and not allowed to ethical consideration.
make decisions that will be harmful. Complex decisions
about capacity will often be value laden. For instance, if a 33.3.5 CONTEXT AND SOCIAL ASPECTS
person with dementia is admitted to a medical or surgical
ward and questions are raised about whether or not they We have already said that relatives and carers must be given
will be able to return home, a finding of incapacity makes it due consideration not least because of the stresses and
more likely that they will end up in some sort of long-term strains that they have to bear. The person with dementia,
care institution (Poole et al., 2014). in any case, should be seen as part of this broader social
context. When people with dementia are living together in
33.3.3 INFORMATION AND DISCLOSURE institutions, the risks associated with harm to them and
to others need careful assessment and, at the same time,
This brings us back to the question of lying. We need to the professionals involved in looking after them will need
respect a person’s autonomy, but there are some things, per- support. At the societal level, the issue of the allocation of
haps, that a person does not wish to know, so weighing up limited resources is also important. This might impinge
how much they should be told will be ethically important. on diagnosis, where certain forms of neuroimaging can be
Information can be given in a helpful or unhelpful manner: expensive, but can also have an impact on pharmacological
how things are done is also an ethical issue. Once again, it and non-pharmacological treatments.
will often be important to involve relatives. There will also
be cultural issues that need to be taken into account where, 33.3.6 THE PROCESS OF CARE
for instance, families are much more closely involved in the
older person’s care and decisions about them. Throughout the trajectory of the dementia it is important
that people are assessed to make sure that everything is being
33.3.4 DECISION MAKING AND CONSENT done to maximize the benefits and minimize the harms of
any interventions. On the whole, it seems important to sup-
It is a matter of ethical importance that the person should be port people with dementia to allow them to live as indepen-
helped to make decisions. For instance, the United Nations dently as possible and to be active in making decisions about
Convention on the Rights of Persons with Disabilities states their care, as well as to keep them in a dignified state. The
in Section 12 that people with disabilities, which would process of care will often involve talking to relatives, but this
include dementia, should be on the same legal footing as should not be to the exclusion of the person with dementia
non-disabled people when it comes to assessments of capac- him or herself. There will sometimes be conflicts in terms of
ity and they should be supported in their decision making the interests or values of the person with dementia and his or
(United Nations, 2008). But good decision making means her family or close friends. Professional and non-professional
that the right circumstances must obtain. There must be carers will require continuing education to allow them to ful-
good relationships, with enough time and with the right fil their roles. This is also likely to decrease the risk of abuse
attitude to allow the person to make a good decision. The and neglect. All of these are clearly ethical issues.
UN Convention would, in any case, suggest that decisions
should not be made for the person, but instead the person 33.3.7 SPECIFIC SITUATIONS
him or herself should be supported to make the decision.
Information from relatives is important, so there must be There are, in addition, a number of specific ethical
good communication and the information must be handled situations that arise during the course of dementia
appropriately. (see Foster et al., 2014). First, driving can be a very emotive
342 Dementia

issue. On the whole, people should be allowed to drive as clinical) judgement will be required concerning whether
long as possible, but the safety of others cannot be compro- this is so.
mised. Once again, how the person is told that they should Finally, there are a number of issues to do with the end
not drive and whether or not this involves specific assess- of life that arise in dementia care (see Chapter 34). These are
ments are ethical issues of note. often about withholding or withdrawing treatment, e.g. in
Second, sexual relationships in the context of dementia connection with the use of antibiotics in severe dementia or
can cause considerable ethical concern. This is particu- the use of artificial nutrition and hydration. A general ethi-
larly the case in care homes. Care home staff sometimes cal doctrine, which is often useful in thinking about issues
have to deal with sexual disinhibition. In addition, the such as tube feeding and withholding or withdrawing
person with dementia may forget that he or she has ever treatments, is that of ordinary and extraordinary means.
been married and attachments may form with other resi- In brief, this suggests that we are only morally obliged to
dents. On occasions, these attachments can become quite provide ordinary care. This means that extraordinary care
intimate and sexual. It is clearly important to think about can be given under particular circumstances, but this is not
safety and often the issue that emerges is to do with one a moral obligation. ‘Extraordinary’ is defined in terms of
or other partner’s capacity to consent to the sexual behav- whether the treatment is likely to be effective and whether
iour. While this is important, it is also equally important or not it is likely to be burdensome to the person and/or
that intimacy should be seen as part of the human condi- his or her family. If a treatment, such as cardiopulmonary
tion. It is a manifestation of friendship and it would be a resuscitation, is unlikely to be effective and is likely to be
good thing to encourage friendships within the commu- burdensome, then there is no moral obligation to under-
nity of a care home. take it.
Third, the issue of genetic testing frequently raises its
head in connection with dementia. The standard advice is
still that genetic testing should not be offered on a routine 33.3.8 SUICIDALITY
basis. However, there may be particular indications when it
is more reasonable for genetic testing to take place. A very There is increasing pressure in some countries to consider
strong history of early onset dementia, for instance, might euthanasia or physician-assisted suicide. This is sometimes
make genetic testing seem imperative, as would a particular put forward as a way to respect the person’s autonomy. But
condition such as Huntington’s disease. broader issues to do with the effects of permissive laws must
Fourth, the debates about the use of global positioning also be considered. It seems better, in any case, to emphasize
systems and other forms of electronic monitoring, or indeed the possibility that people with dementia can live well rather
the use of robots, remains a burning issue. Arguments than offer the option of death as a way, perhaps, to avoid the
against electronic monitoring are often couched in terms difficulties of providing good quality care for people with
dementia.
of the restrictions of liberty that these systems allow. Some
Another ethical issue that arises in connection with
people have argued, however, that the bigger consideration
suicide in dementia is just the worry that the diagnosis
is the risk that technology may decrease the chances of and
itself will induce suicidal wishes. The literature around
necessity for real human contact.
the possibility of rational suicide once again stresses the
Fifth, technological improvements, such as more accu-
importance of assessments of capacity. There is the fur-
rate brain imaging and tests of the cerebrospinal fluid (CSF),
ther concern that some of the motivation towards suicide
mean that issues arise not only about resource allocation
will be the stigma that surrounds dementia. Hence, there
and earlier diagnosis but also about the exact meaning of
is an ethical requirement that we should look at ways in
any particular finding for the individual.
which we might improve the well-being of people with
Sixth, in some countries physical restraints are still used.
dementia.
Sometimes these are not seen as restraints, where people are
put in chairs from which they cannot arise. But any form of
restraint will raise ethical questions.
Seventh, covert medication, where medication is put into
drinks or food, often seems to be necessary, but is neverthe-
33.4 CONCLUSIONS
less a restriction of the person’s ability to make autonomous
decisions about their lives and clearly raises important ethi- Having sketched the spectrum of ethical issues that arise
cal issues. in the context of dementia based on a review of the litera-
Eighth, there has been a huge debate about the use of anti- ture, there is still a question about how in practice ethical
psychotic drugs to control agitated or aggressive behaviour decisions are made. We have already seen that the notion
in people with dementia. It is largely agreed that the more of therapeutic lying can be judged in different directions
appropriate approach should be to think of psychosocial using different ethical theories. The same will apply to
interventions that will be helpful in behaviours that others some of the other ethical approaches discussed above.
find challenging. But on occasions antipsychotic medica- But if we take any of the particular ethical issues that we
tion seems the best option, although again an ethical (and have raised in this chapter, it will always be the case that
Ethical issues 343

judgements must look at concrete circumstances. The REFERENCES


importance of context, of the relationships within that
context, as well as a proper understanding of the nar- Ashcroft, R.E., Dawson, A., Draper, H. and McMillan,
rative surrounding the individual, will always be essen- J.R. (eds.) (2007). Principles of Health Care Ethics,
tial to any particular moral or ethical judgement. These Second Edition. Chichester, United Kingdom: John
judgements will require immersion in the details of the Wiley & Sons.
particular case that must then be followed by the applica- Beauchamp, T.L. and Childress, J.F. (2001). Principles
tion of what seemed to be the appropriate moral theories of Biomedical Ethics, Fifth Edition. Oxford, United
or approaches. It should be possible to give justifications Kingdom: Oxford University Press.
for the use of any particular moral theory, to show why it Culley, H., Barber, R., Hope, A. and James, I. (2013).
seems to be more or less relevant. This process is known Therapeutic lying in dementia care. Nursing Standard,
as casuistry. It is the process of looking at things case by 28: 35–39.
case. It was recommended as part of the ethical frame- Foster, C., Herring, J. and Doron, I. (eds.) (2014). The Law
work in Dementia: Ethical Issues (Nuffield Council on and Ethics of Dementia. Oxford, United Kingdom: Hart
Bioethics, 2009). Casuistry has also been discussed in an Publishers.
earlier edition of this volume (Louw and Hughes, 2005). Hughes, J.C. (2011). Thinking Through Dementia. Oxford,
The emphasis on context, however, which means that dif- United Kingdom: Oxford University Press.
ferent theories or approaches may seem more or less per- Hughes, J.C. and Baldwin, C. (2006). Ethical Issues in
tinent to different cases, suggests that what is regarded as Dementia Care: Making Difficult Decisions. London,
right or wrong demands some sort of higher justification. United Kingdom and Philadelphia, PA: Jessica
There are different views about this, but one approach to jus- Kingsley.
tification is to consider the notion of coherence. (An alter- Louw, S.J. and Hughes, J.C. (2005). Ethical issues. In
native is to look for the foundations of a particular ethical A. Burns, J. O’Brien and D. Ames (eds.), Dementia,
theory to find some basic principle, which will then govern Third edition. London, United Kingdom: Arnold Health
and justify everything that follows. But this is practically, Sciences, pp. 230–234.
as well as theoretically, difficult.) Coherentism suggests Nuffield Council on Bioethics (2009). Dementia: Ethical
that our beliefs and the judgements we form on the basis of Issues. London, United Kingdom: Nuffield Council on
our beliefs should cohere. That is, we must have a coherent Bioethics.
approach to the ways in which we deal with individual ethi- Poole, M., Bond, J., Emmett, C. et al. (2014). Going home?
cal problems. An ethnographic study of assessment of capacity and
One final suggestion, therefore, is that when any particu- best interests in people with dementia being dis-
lar decision does not cohere with our other beliefs, this is charged from hospital. BMC Geriatrics, 14: 56.
when our moral sense comes into play. Moral sense or moral Strech, D., Mertz, M., Knüppel, H. et al. (2013). The
intuition are not particularly fashionable ideas because full spectrum of ethical issues in dementia care:
they raise the problems recognized hundreds of years ago Systematic qualitative review. The British Journal of
in connection with conscience being the basis of moral Psychiatry, 202: 400–406.
theory. Nevertheless, a through and through conscientious United Nations (2008). UN Convention on the Rights of
approach to the ethical dilemmas and problems that arise Persons with Disabilities. Available at http://www.
for people with dementia and for their carers would not be un.org/disabilities/convention/conventionfull.shtml.
a bad place to start. Accessed 30 November 2014.
34
End-of-life and palliative care in dementia

JULIAN C. HUGHES AND JENNY T. VAN DER STEEN

34.1 INTRODUCTION 34.2 THE APPLICABILITY OF THE


PALLIATIVE CARE MODEL
A philosophical question that arises in connection with
dementia is when should end-of-life or palliative care Most of the domains and recommendations of the Delphi
begin? In the popular mind, palliative care is associated study were quickly and unanimously accepted by the
with dying. So palliative care begins when the person experts consulted. But the notion that palliative care is
is dying, but when is that? These are both practical and applicable to dementia was one area where consensus was
conceptual problems. They are even more pointed when only moderate rather than full. It is easy to understand
it comes to dementia. For dementia is a condition that is why. Does it not seem inappropriate, when a person is still
variable in both its nature and progression. We can stipu- fairly fit, even if he or she has forgetfulness and other mild
late that end-of-life care in dementia refers to the last 6 problems, to say that the person requires palliative or end-
or 12 months of life. But why should this be the case? The of-life care?
person with dementia may be quite active 5 months before First, almost all dementias are progressive; second, they
he or she dies. We could, alternatively, stipulate that end- are life-shortening. Our view is that facing the reality is
of-life care refers only to the last few hours or days. But likely to improve care. It can do this in three ways. First, the
this would then ignore all those decisions – about artificial perspective of palliative care may make treatment decisions
nutrition and hydration, the use of antibiotics or cardio- more appropriate: what can be cured should be cured, but
pulmonary resuscitation – which are about end-of-life and sometimes the aim should be simply to maintain the person’s
are often much better made well before the last few hours functional level as opposed to cure. Second, once healthcare
or days. It makes much more sense, therefore, to extend professionals have it in mind that the person has a terminal
the concept of end-of-life backwards; in which case, it is disease, when the time is right they may be more likely to
more natural to speak of palliative care. attend to important end-of-life issues, such as pain relief and
In this chapter, we wish to take a broad view of pallia- other ways to improve comfort. Finally, the palliative care
tive care. But the notion of palliative care in the context perspective is more likely to encourage appropriate advance
of dementia is not clear. For this reason we undertook a care planning.
Delphi study involving 64 experts from 23 countries to In any case, the palliative care approach is always going
define a set of domains (11) and recommendations (57), to be applicable to chronic and progressive diseases and
which together define optimal palliative care at least in all professionals should be aware of its main tenets. The
older people with dementia (van der Steen et al., 2014). holism of palliative care is no different from the holism of
In the remainder of the chapter, we shall discuss pallia- good quality dementia care and both aim at overall quality
tive care and end-of-life decisions using the 11 domains of life. Both approaches require attention to the biological,
derived from the study. psychological, social and spiritual aspects of care, which

344
End-of-life and palliative care in dementia 345

includes involving the family and close carers in decisions. It may be that eventually the person with dementia cannot
Specialist palliative care may only be required if symptom join in these discussions, but every effort should be made
control is complex. The alternative notion of ‘supportive to help them to participate. Indeed, the United Nations
care’ is broader still and may answer some of the concerns Convention on the Rights of Persons with Disabilities specifi-
expressed about ‘palliative care’ (Hughes et al., 2010). cally says that people should be supported in their decision
making (United Nations, 2008). They should certainly not
be disadvantaged on account of their disabilities. Finally,
34.3 PERSON-CENTRED CARE, advance care plans must be stored in a way that allows ready
COMMUNICATION AND SHARED access when they are required.
The aim with all such measures is to avoid inappropri-
DECISION MAKING
ate interventions. What makes them ‘inappropriate’ is that
they would not be the investigations or treatments that the
In the field of dementia we are very used to thinking about person himself or herself would have wished for. In other
the notion of person-centred care (Kitwood, 1997). But words, advance planning is part of person-centred care.
person-centred care is also central to palliative care. To
understand the full impact of person-centred care, it is
necessary to have a broad view of what it is to be a per-
son (Hughes, 2011). Thus, we should regard the person as 34.5 CONTINUITY OF CARE
situated in a context that involves other people, especially
close family and other carers. Good communication with People with dementia and their carers are often keen to have
all involved will be essential, but it becomes more diffi- continuity of care, which brings numerous advantages. It
cult over time; so efforts must be made to keep the person makes communication much easier and much less frustrat-
involved. His or her currently expressed preferences must ing for families if they are not having repeatedly to start
be given due attention, but so too must the person’s pre- afresh. In particular, if the person with dementia moves into
viously expressed preferences (advance decision-making long-term care, it can be annoying and upsetting if all the
is discussed in Section 34.4). This sort of weighing-up of previous relationships with health and social carers simply
different aspects of the person’s life will be very impor- stop. Yet, it may not always be possible for exactly the same
tant when it comes to making decisions. Decision making staff to be involved over the full course of the person’s demen-
should, when feasible, be shared and the person (and/or tia. If this is the case, then there should at least be attempts
the ­family) involved as much as possible. to make any transfers from team to team as smooth as pos-
sible. Particularly important as the end-of-life nears, moving
the person between teams may well be detrimental and is less
than supportive for the family too.
34.4 SETTING CARE GOALS AND
How continuity can be maintained is a challenge. If it
ADVANCE PLANNING is not possible for a single person to co-ordinate or over-
see care throughout the trajectory of the dementia, a sense
If early on in the disease the person is able to set out explic- of continuity may be maintained through careful transfer.
itly what they would like care to aim at, this can help treat- Some have suggested the possibility that teams or networks
ment decisions later. The benefits of advance care planning of care might be virtual in that they need not exist as single
are numerous. They allow the person to continue to man- entities in a particular place (Hughes et al., 2010). However
age his or her affairs while still able to do so. An advance continuity of care is achieved, the purpose is to provide a
care plan also ensures that, insofar as possible, the per- relationship that will be sustaining for the person and his or
son’s wishes will be met in the future, which should bring her close family and carers.
some peace of mind and decrease the burden on loved ones
and on the family who may otherwise have conflicting
views. Conversations about how and the extent to which
34.6 PROGNOSTICATION AND
life should be prolonged are difficult and sensitive. So one
advantage of advance care planning is that these topics
TIMELY RECOGNITION OF DYING
arise and are given some consideration at a point where
the person is able to participate. It remains true that the It will always be difficult to know when and how to raise
evidence that advance care planning is effective is fairly the subject of death and dying. Talking about less traumatic
sparse, but the process of advance care planning might in aspects of advance care planning, such as financial planning,
itself be useful, even if specific items in the advance care might start relevant conversations. It would have to be made
plan are not or cannot be met. clear that prognostication in dementia is a challenge and that
Advance care plans will have to be revisited with both the death cannot be predicted with any accuracy. There are tools
patient and with the family on a regular basis and especially that can help to predict mortality, however, once again with-
when there are significant changes to the person’s health. out accuracy. Loss of the ability to pursue activities of daily
346 Dementia

living, pneumonia, problems with food and fluid intake are near to death is pointless and may lead to complications such
all found (more or less) to predict death. The lack of predict- as fluid overload, which stresses the heart and can cause pul-
ability emphasizes that honest discussions are required early monary oedema. Such issues need to be dealt with in a sensi-
on in order that end-of-life decisions can be as true as possible tive manner to reassure family and close friends.
to what the person would have wanted. Tube feeding is similarly an issue. A number of observa-
tional studies have shown that tube feeding does not affect
survival in dementia when it is compared to hand feeding.
There are, however, possible complications to tube feeding. It
34.7 AVOIDING OVERLY AGGRESSIVE,
also seems as if feeding people by hand, even when there is the
BURDENSOME OR FUTILE
risk of aspiration and consequent pneumonia, can increase
TREATMENTS much needed social interaction. Once again, communication
with all concerned is absolutely vital if decisions are being
It is often said, for good reasons, that acute hospitals are made to stop feeding. Still, nutrition earlier in dementia
unsuitable places for people with dementia. This is particu- remains important because avoiding significant weight loss
larly the case as the dementia becomes more advanced. In will help with, for instance, skin integrity later in the disease.
general, survival is shorter for patients with dementia com- The use of antibiotics is complicated in dementia (van der
pared with those without but who have strokes, hip fractures Steen et al., 2011). They are, of course, necessary in the symp-
and pneumonias. Decisions need to be made keeping in mind tomatic treatment of conditions such as urinary tract infec-
the goals of care and the stage of the person’s dementia. The tions. But some urinary tract infections are chronic and, if
person’s confusion may well worsen once transferred to hos- they are asymptomatic, do not always require treatment.
pital and the risk of delirium is relatively high. Meanwhile, There is conflicting evidence about the use of antibiotics to
staff in general hospitals are not always able to deal with the relieve symptoms in pneumonia. They can be used with cura-
specific problems of people with dementia. tive intent, if this is an appropriate treatment goal, but they
Not only is hospital admission burdensome and some- are also sometimes used in a palliative way; for instance, to
times futile for people with dementia, but polypharmacy, decrease the stickiness of secretions, even where there is no
that is where people are on multiple drugs, can also be bur- real hope of cure. The tendency for older people with dementia
densome because of the side effects of drugs as well as their to have recurrent episodes of infection is commonly observed.
interactions. Many people with dementia will also have It is a matter of delicate clinical judgement, albeit prognostic
co-morbidities, such as high blood pressure, heart disease, scores can be helpful, as to whether any particular infection is
diabetes and osteoporosis, which means that they will be on the one that will cause the demise of the patient rather than
a variety of drugs. This becomes more difficult for them if simply make them unwell for an extended period of time.
they start to develop swallowing problems. For all of these
reasons, therefore, the goals of care should be clear.
In the early stages of dementia it is entirely reasonable,
depending on the person’s general physical state, to think
34.8 OPTIMAL TREATMENT AND
in terms of prolonging life. But when the person is nearing
PROVIDING COMFORT
the end-of-life, the use of drugs such as statins to lower cho-
lesterol levels becomes much more questionable. The same We know that people with dementia, especially in the
is true of drugs that are aimed at maintaining the person’s severer stages, have symptoms that are not dissimilar from
current functioning once patients are severely impaired. those of people dying from cancer. Many will suffer from
Drugs that are intended to keep the person comfortable pain, shortness of breath and agitation or restlessness. One
should continue. Even then, the drugs will need to be moni- of the reasons for emphasizing the need for palliative care
tored. For instance, laxatives to prevent constipation can in people with dementia is because many of these symptoms
cause diarrhoea, and drugs for pain may sedate the patient are underdiagnosed and undertreated. Agitation, which is
or give them constipation, so should only be used when counted as one of the behavioural and psychological symp-
pain is present (which is not, however, to condone under-­ toms of dementia (BPSD), may have a variety of causes, one
treatment, as we discuss below). of which is pain, but it could also be caused by anxiety or
The issue of artificial nutrition and hydration was another frustration. Therefore, broad and rigorous assessments are
area where the consensus of the experts in the Delphi study required. Depression, too, is a form of BPSD, which can
was only modest rather than full. Worries about dehydra- itself cause irritable or agitated behaviour. It is reasonable,
tion are, of course, very real in older people with dementia. therefore, to consider trials of analgesics or antidepres-
Dehydration can affect survival in pneumonia, but it also sants when the behaviour of people with dementia changes.
affects the rate of healing of wounds such as ulcers. Once Distinguishing between sources of discomfort or distress
someone is actually dying, however, the requirement for in severe dementia is of paramount importance if the cor-
hydration diminishes and eventually disappears. Avoiding rect management is to be pursued. It will often be helpful
dryness of the mouth is still a priority for the sake of comfort, to speak with those who know the person very well, either
but using subcutaneous rehydration or intravenous hydration family or those providing day-to-day care.
End-of-life and palliative care in dementia 347

There is now a variety of tools to assess putative pain the help of faith leaders will often be of benefit not only to
in people with dementia. On the whole, they are not well the person with dementia, but also to his or her family and
validated. A general worry with pain scales is that they do close carers. It has often been observed that rituals and sym-
not solely pick up pain, but actually pick up distress given bols of great familiarity have helped to bring some comfort
that there is no particular symptom or sign that is pathog- to people with dementia who have held religious beliefs and
nomonic of pain (Jordan et al., 2012). It is true, nonetheless, they might, too, prove helpful to close friends and family.
that these scales may be useful, once a clinical diagnosis of
pain has been made, as a means to monitor the effective-
ness of treatment. If used for screening, such scales must be
combined with an adequate physical examination and pain 34.10 FAMILY CARE AND INVOLVEMENT
history. The gold standard, therefore, remains a thorough
clinical assessment, which will involve taking a history The holistic approach of palliative care always involves the
from the person if this is possible, their family and carers, as family and those close to the person concerned. Once again,
well as reviewing notes and carrying out observations and a this is a way of recognizing the situated nature of the per-
proper examination. son who is always embedded in a family as well as in a cul-
The first line of treatment for BPSD, however, should ture and particular social context (Hughes, 2011). Because
almost always be non-pharmacological treatments and of these intimate bonds, families will often experience
psychosocial interventions. Simply sitting with a person to caregiver burden. It is important to note that the support
give them comfort may help to alleviate symptoms. The evi- required by families continues throughout the trajectory
dence in favour of many of the psychosocial interventions of dementia. Addressing the specific needs of families dur-
has been weak, but is showing signs of strengthening, with ing these stages of dementia requires awareness, but also
some encouraging findings emerging. resources. Families differ, of course, but some will wish to
In the terminal stages in particular, the person with stay very involved even when the person with dementia is
dementia needs expert nursing care with a lot of attention moved into long-term care. The family will often act as the
being paid to positioning in order to avoid pressure sores proxy decision maker for which they will require support.
and ulcers, along with moistening of the lips. When the per- Some of the carer burden may actually be ethical burden in
son is incontinent, regular washing and changing will be that close family carers may feel as if they are being required
important to protect the perineal area. A variety of environ- to make quite difficult ethical decisions. Education and sup-
mental measures, which are also psycho-social in nature, port, therefore, are vital.
including Snoezelen, music, warmth and pleasant aromas It should not, however, be thought that all carers find
may also help to keep the person calm. caring to be a burden, since some describe positive aspects
At the very end of life more specialist palliative care may in terms of the insight and understanding that they acquire
be required. Often, if the person is dying solely from demen- through the journey of caring for someone they have loved.
tia, death may be quite peaceful without the need for medical Nevertheless, as the person fades, families will often suf-
intervention other than good nursing care. But sometimes fer symptoms of bereavement even before the person has
people with dementia are dying from other conditions, so died and families should continue to receive support.
that relief of their symptoms is more complex and requires Complicated grief reactions are more likely in those carers
specialist intervention. Syringe drivers, for instance, are not who have been heavily involved in care, or have high levels
always readily available in people’s homes or in care homes, of depressive symptoms prior to the death of the person for
but specialists in palliative care may be able to provide both whom they are caring, or where they have been caring for
the technology and know-how to enable the person to die somebody with more severe cognitive impairment.
comfortably where they have lived rather than to require a
move to a hospital or hospice.
34.11 EDUCATION OF THE
HEALTHCARE TEAM
34.9 PSYCHOSOCIAL AND SPIRITUAL
SUPPORT If palliative care is to be provided effectively, it will require
a team effort. This means that training across all those
Emotional support is going to be of great importance involved in palliative care for people with dementia will
throughout the course of dementia, from the mildest stages be required on an ongoing basis. Palliative care specialists
to the most severe. To be holistic, this must include emotional will need to learn more about dementia and dementia care
support for the family and for the carers of the person with specialists will need to know more about palliative care.
dementia. Proper holistic support will include attention to Meanwhile, those who work in institutional settings will
spiritual matters. There should be an assessment of the per- also require the skills to recognize when people are deterio-
son’s religious affiliations and involvement in order to seek rating and when they might be developing symptoms that
out the best means of providing spiritual support. Enlisting require palliative interventions.
348 Dementia

the end of life, when the person is finally dying, dementia


34.12 SOCIETAL AND ETHICAL ISSUES care and palliative care must in one way or another come
together.
Both palliative and dementia care specialists tend to be The inspiration behind palliative care is, of course, the
attuned to ethical issues, which arise in the context of holis- work of Dame Cicely Saunders who established the first
tic care (see Chapter 33). modern hospice, St Christopher’s in London, which opened
One broader societal issue is that palliative care should in 1967. In the realm of dementia care we should be able
be available to people with dementia wherever they live, to say to people with dementia, as Dame Cicely Saunders
whether in their community, in residential care or in an said: ‘You matter because you are you. You matter to the
acute hospital. Support should also be available for fami- last moment of your life, and we will do all we can to help
lies in all of these settings. The provision of palliative care you not only to die peacefully, but also to live until you die’
throughout the course of dementia and wherever the per- (Saunders, 2006, p.137).
son is residing will be facilitated when there is good col-
laboration between dementia health and care services and REFERENCES
specialist palliative care services. Close working between
teams will be beneficial for all concerned. But the commit- Hughes, J.C. (2011). Thinking through Dementia. Oxford:
ment to this must start early with specific training for both Oxford University Press.
physicians and for nurses at undergraduate and postgradu- Hughes, J.C., Lloyd-Williams, M. and Sachs, G.A. (eds.)
ate level to include palliative care for patients with illnesses (2010). Supportive Care for the Person with Dementia.
other than cancer. Given the challenging nature of this work Oxford: Oxford University Press.
and the increase in the prevalence of dementia, there should Jordan, A., Regnard, C., O’Brien, J.T. and Hughes,
be systemic economic incentives to encourage excellence in J.C. (2012). Pain and distress in advanced demen-
palliative and end-of-life care for people with dementia. tia: Choosing the right tools for the job. Palliative
The issues that professionals and families face in connec- Medicine, 26: 873–878.
tion with palliative care for people with dementia also need Kitwood, T. (1997). Dementia Reconsidered: The Person
to be given a higher profile in a way that will encourage the Comes First. Buckingham: Open University Press.
general public to be more supportive of individuals and fami- Nuffield Council on Bioethics (2009). Dementia: Ethical
lies who live with dementia. This is in keeping with the notion Issues. London: Nuffield Council on Bioethics.
of solidarity, which was emphasized in the report Dementia: Saunders, C. (2006). Cicely Saunders. Selected Writings
Ethical Issues (Nuffield Council on Bioethics, 2009). 1958–2004. Oxford: Oxford University Press.
United Nations (2008). UN Convention on the Rights of
Persons with Disabilities. Accessed 30 November
2014. Available at http://www.un.org/disabilities/­
34.13 CONCLUSION convention/conventionfull.shtml.
van der Steen, J.T. (2011). Prolonged life and increased
Palliative care for people with dementia is not optional. It symptoms vs prolonged dying and increased comfort
is essential. The needs of people with dementia, especially after antibiotic treatment in patients with dementia
in the moderate to severe stages of the disease, are about and pneumonia. Archives of Internal Medicine, 171:
as complex as any that face health and social care profes- 93–94.
sionals. The issues of pain and distress in severe dementia, van der Steen, J.T., Radbruch, L., Hertogh, C.M.P.M. et al.,
where the person is unable to convey information about the on behalf of the European Association for Palliative
potential causes of his or her symptoms or signs, are them- Care (EAPC) (2014). White paper defining optimal
selves complex and require specialist help at times. This palliative care in older people with dementia: A Delphi
help may come from dementia specialists, but may also study and recommendations from the European
come from palliative care where the concerns are about Association for Palliative Care. Palliative Medicine,
the possibility of pain and its management. Especially at 28: 197–209.
35
Advanced dementia and care at the end of life

KIRSTEN MOORE AND ELIZABETH SAMPSON

hold up head independently (Reisberg, 1988). The Global


35.1 INTRODUCTION Deterioration Scale (GDS) uses a similar approach to stages
of disease progression but incorporates both cognitive and
Dementia is now more commonly understood to be a functional abilities (Reisberg et al., 1982). The Bedford
terminal condition and, as such, has been identified as one Alzheimer Nursing Scale – Severity Subscale (BANS-S)
where a palliative care approach may be beneficial (Connor assesses severity in dementia of the Alzheimer type by incor-
and Bermedo, 2014; van der Steen et al., 2014). A growing porating cognition, activities of daily living and appearance
number of deaths are attributable to dementia and it is now of pathological symptoms (including sleep wake cycle dis-
the fourth highest cause of death in high income coun- turbance and contractures) (Volicer et al., 1994).
tries (World Health Organization, 2014). However, it is still These are mainly research tools, however, and in day-to-day
under-recognized as a cause of death on death certificates care the advanced stages of dementia are defined by depen-
(Wachterman et al., 2008). In addition, a review of dementia dence in all care, limited speech, extreme apathy, incontinence
strategies from 14 countries found that 8 did not refer to and difficulties with chewing and swallowing and increased
palliative care (Nakanishi et al., 2015). While recognition occurrence of infections (Forstl and Kurz, 1999). Management
of the need for a palliative approach is growing, there is still of other co-morbidities may become difficult as the person
limited research evidence to this approach in dementia care. may not be able to agree to various procedures or medications.
This chapter begins with an overview of the com- The reduced ability to communicate during advanced demen-
mon symptoms that occur during the advanced stages of tia creates challenges to identifying physical and psychosocial
dementia followed by a discussion of the difficulties associ- needs, such as pain, hunger, boredom or loneliness.
ated with identifying end of life in dementia and the chal- Table 35.1 shows the common symptoms for people with
lenges of planning for end-of-life care. The third section dementia towards the end of life. Vandervoort et al. (2013)
will discuss comfort care and holistic care and how the considered symptoms associated with end of life according
goals of care change during the advanced stage of disease. to dementia severity. In their sample of nursing home resi-
The final section highlights the importance of working dents who died, 54% had very severe/advanced dementia,
with family members as those who have knowledge and 28% had severe dementia and 18% had mild or moderate
insights into the person with dementia but also as those dementia. Occurrence of pneumonia and febrile episodes
who may be experiencing a prolonged period of pre-death in the last month did not differ between stages of dementia,
grief as they witness the multiple losses of the family mem- although choking, swallowing difficulties, incontinence and
ber they have known before the onset of dementia. dehydration during the last week were higher for those with
more advanced dementia.

35.2 WHAT IS ADVANCED DEMENTIA?


35.3 IDENTIFYING AND PLANNING
There are numerous validated questionnaires that have
FOR THE END OF LIFE
been developed to assess the severity of dementia. The
Functional Assessment Staging (FAST) focuses on function The experience of end of life at different stages of dementia
and requires a proxy to rate the level of disability on a scale may be similar (Vandervoort et al., 2013) and only 41% of
ranging from no difficulties through to loss of ability to long-term care residents who die with dementia have reached
349
350 Dementia

Table 35.1 Common end-of-life symptoms and needs in dementia


Study and method Common end-of-life symptoms and needs (%)
Longitudinal prospective study of 323 U.S. nursing home Last 3 months:
residents with advanced dementia. 55% died during the
• Eating problems (90)
18-month period of the study (Mitchell et al., 2009)
• Pneumonia (37)
• Febrile episodes (32)
• Dyspnoea, pain, pressure ulcers and aspiration all
increased towards the end of life
Retrospective study of 198 Belgium nursing home residents Last month:
with dementia at any stage (Vandervoort et al., 2013)
• Most common symptoms: pain, fear, anxiety
• Pneumonia (32)
• Febrile episodes (43)
• Eating problems (66)
Last week:
• Urinary incontinence (89)
• Faecal incontinence (86)
• Dehydration (39)
• Decubitus (27)

Systematic literature review (Potter et al., 2013) • Requires assistance for daily tasks, self-care and
continence
• Eating and feeding difficulties
• Reduced mobility and posture
• Contractures
• Persistent agitation and withdrawal
• Recurrent infections
• Inability to recognize familiar people and things

the advanced stages of disease (van der Steen et al., 2013b). One of the goals of comfort care is to avoid burdensome
During the advanced stages, however, quality of life is gener- interventions that may increase distress and pain yet may not
ally very low. At this point, the goals of care tend to shift from create any substantial increase in quality of life or life expec-
active treatments to a palliative and comfort approach (van tancy. Burdensome interventions, however, are still common
der Steen et al., 2014). As dementia may progress very slowly, in advanced stages of disease. Mitchell et al. (2009) identified
it can be difficult to identify a point in time when someone that 41% of nursing home residents with severe dementia had
shifts from moderate-to-severe dementia. This may be pre- a burdensome intervention in the last 3 months of life. These
cipitated by a sentinel event such as a fall or it may be a slow most commonly included parenteral therapy (29%), being
gradual process over months or years. While it may be helpful hospitalized (12%) and being tube fed (7%).
to inform families of when end of life is nearing, prognostic Comfort care aims to minimize pain, agitation and dis-
tools have only shown modest results in predicting end of life tress with an overall goal of maximizing quality of life. In
(Mitchell et al., 2010). advanced dementia and end-of-life care a holistic, interdis-
One of the difficulties of estimating survival is that ciplinary approach with individualized care plans provide a
there may be multiple causes of death, and events that sound basis for assessing needs and planning and providing
can cause final death may also respond to treatment. care (Kovach et al., 1996; Hall et al., 2011). An integrated
For example, while people with dementia are more than approach involving a care coordinator who provides a sin-
twice as likely to die with pneumonia than those with- gle point of contact for families and ensures that plans are
out dementia (Foley et al., 2014), they may also respond followed and kept relevant also appears to be critical for
to treatment so that their health temporarily stabilizes. improving end-of-life care (Elliott et al., 2014). A person-
However, as the underlying causes of the pneumonia, centred approach to end-of-life care encourages meeting
such as dysphagia, are likely to remain, the risk of devel- the goals and wishes of the person with dementia; however,
oping pneumonia again is high. The broader concept of identifying these goals during the advanced stages can be
frailty is relevant here as the cumulative effect of declines difficult and reliance on proxy decision makers or prefer-
in multiple body systems may increase the likelihood of ences documented by the person when they still had capac-
death (Fried et al., 2004). ity may be required.
Advanced dementia and care at the end of life 351

settings (Robinson et al., 2013). Health professionals and


35.4 ADVANCE CARE PLANNING patients tend to be hesitant to initiate end-of-life discussions
during consultations (Almack et al., 2012). Professionals
Advance care planning (ACP) involves informal discus- also queried the usefulness of ACPs given patient’s prefer-
sions and may include formal documentation of decisions ences were not always able to be fulfilled, such as wishing to
about future and end-of-life care while the person still has remain at home (Robinson et al., 2013).
capacity. Making plans for the future may involve legally Few studies have obtained the views from people with
appointing someone to act as a proxy decision maker for early stages of dementia about future care preferences. Some
when the person with dementia no longer has capacity. studies have looked at hypothetical scenarios to investigate
Although the legal terms and roles differ across juris- how older people might make decisions about their future
dictions, the role is often called an Enduring or Lasting healthcare. Gjerdingen et al. (1999), found that most older
Power of Attorney. During the early stages of dementia, people would not want burdensome interventions such as
people may still have capacity to record decisions about CPR, tube feeding, hospitalization or antibiotics during
their future care, such as whether or not and under what advanced dementia. Low et al. (2003) interviewed nursing
circumstances they would want certain interventions such home residents without dementia about interventions they
as cardiopulmonary resuscitation (CPR), artificial feeding would want if they had advanced dementia and recurrent
or hydration and mechanical ventilation. ACP offers an pneumonia. The majority wanted antibiotic treatment and
opportunity for people with the early stages of dementia to hospitalization but only a quarter would agree to artificial
express their values and preferences to the health profes- feeding. A study involving people with mild dementia who
sionals and family members who are likely to be making had capacity to participate in a discussion on ACP found
future decisions on their behalf. that they had difficulty envisaging their future selves and
While ACPs appear a promising way of ensuring the the possibility of being more dependent on others (Dening
wishes of the person with dementia are followed, there are et al., 2013). Even during the early stages, people with
many challenges to implementing ACP and evidence of dementia may struggle to think about and plan for possible
effectiveness is modest. A systematic review of ACP inter- future situations.
ventions only identified four studies, all of which were Often ACPs include consideration of CPR. CPR in
conducted in nursing home settings where the majority of people with advanced dementia is unlikely to be success-
residents lacked capacity to participate in ACP discussions ful (Cooper et al., 2006), is an undignified way to die and
(Robinson et al., 2012). The review concluded that there was is undertaken with a high risk of multiple rib and ster-
limited evidence of benefits for residents, although there num fractures (Lederer et al., 2004). Studies investigat-
appeared a reduction in inappropriate hospital admissions ing hypothetical scenarios of whether people would want
(Robinson et al., 2012). CPR or mechanical ventilation if they had dementia show
Families report numerous obstacles to discussing mixed responses (Westenhaver et al., 2010), although
future care with their relative during the early stages of video education and experience of having a relative with
dementia such as not recognizing the importance of ACP dementia may increase the likelihood of people choosing
until it was too late, actively avoiding the discussion, feel- comfort care over life prolonging treatment (Volandes
ing that the personality of the person with dementia was a et al., 2007).
barrier to the discussion or that the person with dementia
was in denial of their diagnosis (Hirschman et al., 2008).
Hirschman et al. (2008) also found that family carers
regretted not having had these discussions earlier and not
35.5 PLACE OF END-OF-LIFE CARE
having had more specific discussions about topics such as
medications and tube feeding. They also found that law- While most people with dementia tend to die in a residen-
yers, accountants and health professionals played a key tial care setting (see Table 35.2), there is very little known
role in initiating discussions about future plans and care about the preferences of people with dementia and their
(Hirschman et al., 2008). Religious beliefs, education, feel- family about preferred place of death (Badrakalimuthu and
ings of guilt, carer burden and negative assumptions about Barclay, 2014). International comparisons also show large
quality of life can influence family carers’ decisions about differences across countries in location of death and this can
end of life and may prevent a family member being able to be partly explained by availability of long-term care beds
make best interest decisions regarding their relative’s care (Reyniers et al., 2014) but may also reflect varying cultural
(Dening et al., 2011). practices and preferences. The high proportion of nursing
Professionals involved in working with people in pal- home deaths in the Netherlands may reflect the medical
liative and dementia care also describe numerous obstacles specialty of elderly care physicians (Koopmans et al., 2010)
to implementing advance care plans including when and working within the nursing home setting and providing a
who was responsible for instigating discussions, planning high level of medical support.
for all possible scenarios, concerns about the terminology Transfer to hospital from care homes during the final
used and the applicability of various documents in different stages of life is often considered undesirable with minimal
352 Dementia

Table 35.2 Location of dementia-related deaths


Location of death (%)
Nursing home/­
Country long term care Home Hospital Other Reference
Belgium 2008 66.7 11.2 21.6 0.5 Reyniers et al. (2014)
Canada 2008 59.4 3.4 32.3 4.9 Reyniers et al. (2014)
Czech Republic 2008 61.5 10.6 27.5 0.4 Reyniers et al. (2014)
England 2001 57.9 4.4 37.5 0.3 Sleeman et al. (2014)
England 2006 48.8 NR 45.4 NR Sleeman et al. (2014)
England 2010 52.6 6.7 40.1 0.6 Sleeman et al. (2014)
France 2008 34.0 27.2 35.9 2.9 Reyniers et al. (2014)
Germany 2008 26.9 42.4 26.2 4.5 Escobar Pinzon et al. (2013)
Hungary 2008 NR NR 62.3 37.7 (Reyniers et al. (2014)
Italy 2008 19.5 42.2 32.1 6.2 Reyniers et al. (2014)
Mexico 2008 NR 69.3 26.2 4.5 Reyniers et al. (2014)
New Zealand 2008 76.6 4.5 14.3 4.7 Reyniers et al. (2014)
The Netherlands 2008 93.1 3.8 1.6 1.5 Reyniers et al. (2014)
Scotland 2003 60.8 5.0 33.9 0.4 Houttekier et al. (2010)
South Korea 2008 5.5 20.5 73.6 0.4 Reyniers et al. (2014)
Spain 2010 20.1 46.1 33.6 0.2 Reyniers et al. (2014)
United States 2007 62.6 15.3 13.2 8.9 Reyniers et al. (2014)
Wales 2008 48.9 4.2 43.1 3.8 Reyniers et al. (2014)
Abbreviation: NR, not reported.

benefits in terms of life expectancy or quality of death.


People with dementia have an increased risk of adverse 35.6 COMFORT CARE
events in hospital (Watkin et al., 2012) and the ability to pro-
vide person-centred and comfort care in the hospital setting Meeting the needs of people with dementia at the end of
is challenging. Transfers to hospital may also be more costly life requires consideration of their physical, psychological,
to health and care systems (Lane et al., 1998). social, spiritual, environmental, supportive and individual
Also included in Table 35.2 are dementia-related deaths needs (Perrar et al., 2015). Psychosocial needs are impor-
occurring in England between 2001 and 2010, which demon- tant and may not be adequately addressed, particularly in
strates that from 2006 to 2010, the proportion of dementia- settings based on a medical model where demands on staff
related deaths in care homes increased and hospital deaths lead to a focus on physical tasks over positive social engage-
decreased (Sleeman et al., 2014). This perhaps reflects recent ment. Kitwood (1997) argued that the medicalization of
concerted efforts to provide end-of-life care in care homes dementia has led to an overemphasis on the losses associ-
and avoid transfers to hospital at end of life. ated with dementia with a failure to acknowledge the needs
Hospice care is associated with better emotional associated with maintaining personhood including the
support, higher levels of respectful treatment and satis- need for attachment, comfort, inclusion, identity and occu-
faction with care when compared with nursing home and pation. Holistic person-centred approaches are now widely
hospital settings (Teno et al., 2004; Shega et al., 2008). espoused and have been shown to be effective in reducing
However, the specialized nature of these services and agitation in nursing home settings (Chenoweth et al., 2009;
their limited availability is unlikely to meet the grow- Livingston et al., 2014).
ing number of people living and dying with dementia. People with advanced dementia may not be able to ver-
A more realistic approach is to improve end-of-life care bally communicate needs and signs of distress, and with-
skills within nursing home and long-term care settings drawal and agitation can indicate unmet need. Trials for
to reduce the need for transfers to hospital for symp- reducing distress and behavioural and psychological symp-
tom management. Bringing hospice expertise into the toms to dementia (BPSD) often have mixed results and tend
care home setting may be a way of achieving this goal to highlight the limitation of research approaches that rely
(Stevenson and Bramson, 2009). Barriers to end-of-life on a single intervention to address an issue. Approaches that
care in residential settings also need to be addressed, are multifaceted and incorporate individual and holistic
such as variable engagement from medical practitioners, assessment are more likely to be effective in reducing symp-
access to out of hours support and availability of training toms. For example, the Serial Trial Intervention has been
(Seymour et al., 2011). shown to effectively increase nurses’ level of assessment of
Advanced dementia and care at the end of life 353

symptoms and persistence in intervening combined with endoscopic gastrostomy (PEG) and nasogastric tubes for
reduced distress among residents (Kovach et al., 2006a). The providing nutrition during advanced dementia is still
intervention consists of behaviour change identification fol- common in some countries (Bentur et al., 2014; Wada et al.,
lowed by serial assessment and intervention (Kovach et al., 2014); however, there is a lack of evidence for its efficacy
2006b). The approach has five steps and if any step is ineffec- in improving life expectancy or reducing pneumonia risk
tive in improving the symptom the next step is trialed. The (Sampson et al., 2009; Goldberg and Altman, 2014). Studies
steps include the following: investigating artificial feeding have also failed to consider
issues of quality of life and comfort (Sampson et al., 2009).
1. Physical assessment and treatment Artificial feeding also raises ethical issues related to euthana-
2. Assessment of affect (environmental stressors, balance sia and prolonging suffering. Less invasive approaches may be
of stimulation and rest and at least 10–20 minutes of preferable, such as postural adjustments and use of modified
positive social engagement twice daily) and addressing textured diets and thickened fluids. While these approaches
any gaps identified are commonly used in nursing homes and hospitals, the
3. Trialling non-pharmacological approaches overall benefit of modified diets is questionable given their
4. Introducing or increasing analgesics reduced acceptability and potential risk of malnutrition and
5. Consultation with experts in mental health and ageing dehydration (Sura et al., 2012). Referral to a dietitian for those
and possibly the use of psychotropic medication who are nutritionally at risk or a speech therapist for those
with swallowing difficulties is recommended.
Fewer studies have examined strategies for reducing A multidisciplinary approach involving physiotherapists
distress for people living with dementia at home, although and occupational therapists for problems with mobility
there is some evidence that family carers of people who have and contractures, and nurse specialists for pressure ulcer
high levels of BPSD tend to have limited understanding care, can be useful where routine management such as
and repertoire of strategies to reduce these symptoms regular movement and pressure relieving devices are inef-
(Moore et al., 2013). fective. Breathing difficulties during the last days of life are
Kovach et al.’s (2006a) study highlighted the importance another symptom that may be distressing for families (Wee
of addressing pain with 46% of residents in the interven- et al., 2006), yet there is little consensus about treatment.
tion group, compared with only 3% of the control group Researchers who recently developed practice guidelines for
residents being prescribed an analgesic. Pain management managing pneumonia among nursing home residents with
in advanced dementia is particularly challenging given the dementia found that there was a lack of research evidence
subjective nature of pain and importance of self-report. and so relied on a Delphi process to gain consensus among
A number of measures have been developed that rely on experts regarding preferred treatment options (van der
observing possible signs of pain such as facial grimaces and Maaden et al., 2014). The process identified that while palli-
negative vocalizations. Psychometrically, the most prom- ative care in dementia required a unique approach, existing
ising tools include the Abbey Pain Scale, DOLOPLUS2, palliative care guidelines were still relevant for treatment of
Pain Assessment in Advanced Dementia and the Pain symptoms. However, the experts did not agree on a number
Assessment Checklist for Seniors with Limited Ability to of interventions such as the use of oxygen for dyspnea and
Communicate (Hadjistavropoulos et al., 2014). The extent to anti-cholinergics for rattling breath with the latter treat-
which these measures are identifying physical pain or emo- ment being considered a strategy for relieving the distress
tional distress is difficult to ascertain (Jordan et al., 2012) of relatives more than for the well-being of the dying patient
and therefore identifying the source and potential remedy (van der Maaden et al., 2014).
can be difficult and may require a trial and error approach.
Namaste care is another approach underpinned by a
person-centred approach and which engages people through
sensory input, comfort and pleasure. It aims to enhance
35.7 WORKING WITH FAMILY
social interaction and shared activity as well as support
family through the progression of dementia in a positive Family carers across the globe have a vital role as they pro-
context by seeking to improve quality of life. It has been vide the bulk of care to people with dementia (Alzheimer’s
demonstrated to reduce neuropsychiatric symptoms among Disease International, 2009) and in the advanced stages
nursing home residents with advanced dementia without they are usually relied on as proxy decision makers regard-
requiring an increase in care staff (Stacpoole et al., 2014). ing end-of-life care and treatment. Family members can
The authors of this study also acknowledged that the suc- provide vital information to assist service providers and
cess of the programme relied on good support from medical health professionals in the provision of care. While they
staff to ensure effective clinical care and pain management may not always know what the person with dementia would
(Stacpoole et al., 2014). have wanted in terms of treatment, they are best placed to
Maintaining nutrition and hydration in advanced demen- inform a holistic approach to care and to have insight into
tia is challenging as people may lose interest in eating and the goals, values and preferences of the person with demen-
develop difficulties with swallowing. The use of percutaneous tia both during and prior to developing dementia.
354 Dementia

Involving family members in discussions about end- just after diagnosis (Chang et al., 2010). Health professionals
of-life care is also important. For example, a U.S. study of reported that having a booklet was useful for aiding discus-
nursing home residents with advanced dementia found sions with family and that these booklets should be widely
that when family members believed their relatives had less available and do not always need to be provided by a health
than 6 months to live and understood likely clinical com- professional (Chang et al., 2010; van der Steen et al., 2011).
plications, the person with dementia was significantly less The timing and sensitive delivery of information is impor-
likely to receive a burdensome intervention during their tant as some carers may find it distressing to hear about
last 3 months of life (Mitchell et al., 2009). Another study advanced symptoms of dementia, particularly during the
in Dutch long-term care facilities found that while only early stages when many symptoms may not be evident and
43% of family members reported dementia as a disease you the focus is on ‘living well’ with dementia.
can die from (prior to the resident’s death), agreement with The needs of family carers are also critical as they are at
this statement was associated with higher resident comfort high risk of experiencing burden and depression (Pinquart
at end of life as assessed by the physician (van der Steen and Sorensen, 2003; Etters et al., 2008). While much research
et al., 2013b). In this study, however, family members’ level has focused on this, the importance of grief should not be
of understanding of dementia prognosis and complications underestimated. Pre-death grief in dementia highlights the
did not impact on the resident’s comfort at end of life. importance of grief throughout various losses associated
Both of these studies found that family carers were rarely with dementia including communication, personhood and
counselled on how long the person with dementia was likely capacity to recognize family members. Pre-death grief dif-
to live (18%–21%) or clinical complications associated with fers from anticipatory grief that has often been described in
advanced dementia (33%–39%) (Mitchell et al., 2009; van der the context of cancer where the person who has cancer can
Steen et al., 2013b). These figures appear exceptionally low communicate with family to resolve old conflicts and help
when the participants with dementia were living in long-term them adjust to the impending loss (Lindauer and Harvath,
care facilities and most had advanced dementia. As described 2014). Discussions can have a therapeutic effect by assisting
above, there are many obstacles to end-of-life discussions and family members to emotionally prepare, but the actual loss
if family carers are unaware that dementia is a terminal dis- of the person has not yet occurred and can only be antici-
ease, it is understandable they would not initiate these discus- pated. In dementia, family carers will experience losses
sions. The onus lies on health professionals to raise end-of-life while the person with dementia is alive and so the grief
discussions particularly since studies have shown that discus- process may be expanded over many years in a protracted
sions about end-of-life care with family members increases period of loss.
satisfaction with care (Reinhardt et al., 2014) and end-of-life Another important loss can be when the person with
care (Engel et al., 2006) and can help carers in making treat- dementia moves into residential care. This transition can
ment decisions (Caplan et al., 2006). cause significant grief and guilt for family carers (Afram
Communicating with family carers about advanced et al., 2014). It can indicate the end of a long period of
dementia requires patience and sensitivity. Family car- cohabitation and for spousal carers this can be a large defin-
ers may not be prepared to commit to documented care ing element of their marriage (Moore and Dow, 2014). It
decisions, possibly as they attach more importance to is unclear to what extent pre-death grief may help family
advance directives than health professionals (Rurup et al., carers adjust or complicate grieving when the person with
2006). However, despite reluctance to formalize plans, dementia dies. A study by Schulz et al. (2003) found that
they do appear to appreciate and benefit from the discus- 72% of family carers described their relative’s death as a
sion (Sampson et al., 2011). Caplan et al. (2006) ‘found that relief and while depressive symptoms increased towards
most families said that previously no one had explained to death, they dropped in the 15 weeks post-death. This was
them what the natural history of dementia was or that their in contrast to the slight increase in depressive symptoms
relative in a [nursing home] was actually dying. They were reported by carers after placing their relative in residential
relieved to have this information, which allowed them to care. Only 21% of carers in this study accessed bereavement
think more clearly about what treatment would be helpful services after their relative died. These findings suggest that
and what would not’ (p. 584). counselling and bereavement support for family carers may
Booklets on end-of-life care and advanced dementia be needed while the person with dementia is alive.
have been shown to be helpful for family carers and health
professionals and could be used to promote end-of-life
discussions. Booklets have been developed and evaluated
in Australia (Chang et al., 2010) and Canada (Arcand and
35.8 CONCLUSIONS
Caron, 2005) and the Canadian booklet has been modified
for use in the Netherlands, Italy and Japan (van der Steen Dementia is a terminal condition requiring a pallia-
et al., 2013a). A study of the Australian booklet found that tive approach, particularly during the advanced stages
two-thirds of carers preferred receiving the booklet at a where the goals of care may shift towards providing com-
specific stage of dementia and of these, 72% indicated that fort and allowing a natural death. The advanced stage of
they would have liked the booklet at onset of symptoms or dementia is characterized by high levels of dependence
Advanced dementia and care at the end of life 355

and low quality of life. Palliative care requires a holis- Bentur, N., Sternberg, S., Shuldiner, J. and Dwolatzky, T.
tic and coordinated approach that involves the person (2014). Feeding tubes for older people with advanced
with dementia and their family members as far as pos- dementia living in the community in Israel. American
sible. Communicating with the person becomes difficult Journal of Alzheimers Disease and Other Dementia,
as dementia advances and therefore addressing their 30 (2): 165–172.
needs becomes challenging. Agitation and distress can Caplan, G.A., Meller, A., Squires, B. et al. (2006). Advance
be signs of unmet needs such as physical pain or lack of care planning and hospital in the nursing home. Age
social engagement. Education of both the person with Ageing, 35: 581–585.
dementia and their family appears important for improv- Chang, E., Easterbrook, S., Hancock, K. et al. (2010).
ing outcomes such as satisfaction with care and reducing Evaluation of an information booklet for caregivers
burdensome interventions; however, the timing of infor- of people with dementia: An Australian perspective.
mation provision is difficult to assess and is often delayed Nursing and Health Science, 12: 45–51.
and avoided. While people with dementia may be able to Chenoweth, L., King, M.T., Jeon, Y.H. et al. (2009). Caring
have input into decisions about future care in the early for Aged Dementia Care Resident Study (CADRES)
stages of dementia, there is evidence to suggest that they of person-centred care, dementia-care mapping, and
have difficulty imagining themselves in the future when usual care in dementia: A cluster-randomised trial.
they may be more dependent on others. Family carers can Lancet Neurology, 8: 317–325.
also be influenced by their own experiences of burden Connor, S. and Bermedo, M. (eds.) (2014). Global Atlas of
and pre-death grief and may not always be in a position Palliative Care at the End of Life. Geneva, Switzerland:
to make decisions that reflect the wishes of the person Worldwide Palliative Care Alliance, World Health
with dementia. More research is needed to better under- Organization.
stand management strategies for numerous symptoms Cooper, S., Janghorbani, M. and Cooper, G. (2006). A
associated with advanced dementia, however, there are decade of in-hospital resuscitation: Outcomes and
promising findings for multifaceted and person-centred prediction of survival? Resuscitation, 68: 231–237.
interventions, which reduce pain and agitation. Person- Dening, K.H., Jones, L. and Sampson, E.L. (2011). Advance
centred approaches also require healthcare profession- care planning for people with dementia: A review.
als to develop positive relationships with family carers International Psychogeriatrics, 23: 1535–1551.
to support joint decision making and to develop a better Dening, K.H., Jones, L. and Sampson, E.L. (2013).
understanding of the needs and preferences of the person Preferences for end-of-life care: A nominal group
with dementia. study of people with dementia and their family carers.
Palliative Medicne, 27: 409–417.
Elliott, M., Harrington, J., Moore, K. et al. (2014). A proto-
REFERENCES col for an exploratory phase I mixed-methods study of
enhanced integrated care for care home residents with
Afram, B., Verbeek, H., Bleijlevens, M.H. and Hamers, J.P. advanced dementia: The Compassion Intervention.
(2014). Needs of informal caregivers during transition BMJ Open, 4: e005661.
from home towards institutional care in dementia: A Engel, S.E., Kiely, D.K. and Mitchell, S.L. (2006).
systematic review of qualitative studies. International Satisfaction with end-of-life care for nursing home
Psychogeriatrics, 27 (6): 891–902. residents with advanced dementia. Journal of
Almack, K., Cox, K., Moghaddam, N. et al. (2012). After American Geriatric Society, 54: 1567–1572.
you: Conversations between patients and healthcare Escobar Pinzon, L.C., Claus, M., Perrar, K.M. et al. (2013).
professionals in planning for end of life care. BMC Dying with dementia: Symptom burden, quality
Palliative Care, 11 (1): 15. of care, and place of death. Deutsches Ärzteblatt
Alzheimer’s Disease International (2009). World Alzheimer International, 110: 195–202.
Report 2009. In M. Prince and J. Jackson (eds.), Etters, L., Goodall, D. and Harrison, B.E. (2008). Caregiver
London, United Kingdom: Alzheimer’s Disease burden among dementia patient caregivers: A review
International. of the literature. Journal of the American Academy of
Arcand, M. and Caron, C. (2005). Comfort Care at the Nurse Practitioners, 20: 423–428.
End of Life for Persons with Alzheimer’s Disease or Foley, N.C., Affoo, R.H. and Martin, R.E. (2014). A system-
Other Degenerative Diseases of the Brain: A Guide atic review and meta-analysis examining pneumonia-
for Caregivers. Canada: Centre de santé et de ser- associated mortality in dementia. Dementia and
vices sociaux-Institut universitaire de gériatrie de Geriatric Cognitive Disorders, 39: 52–67.
Sherbrooke. Forstl, H. and Kurz, A. (1999). Clinical features of
Badrakalimuthu, V. and Barclay, S. (2014). Do people with Alzheimer’s disease. European Archives of Psychiatry
dementia die at their preferred location of death? A and Clinical Neuroscience, 249: 288–290.
systematic literature review and narrative synthesis. Fried, L.P., Ferrucci, L., Darer, J. et al. (2004). Untangling
Age Ageing, 43: 13–19. the concepts of disability, frailty, and comorbidity:
356 Dementia

Implications for improved targeting and care. The Lederer, W., Mair, D., Rabl, W. and Baubin, M. (2004).
Journal of Gerontology Series A Biological Sciences Frequency of rib and sternum fractures associated
Medical Sciences, 59: 255–263. with out-of-hospital cardiopulmonary resuscita-
Gjerdingen, D.K., Neff, J.A., Wang, M. and Chaloner, K. tion is underestimated by conventional chest X-ray.
(1999). Older persons’ opinions about life-sustaining Resuscitation, 60: 157–162.
procedures in the face of dementia. Archives of Family Lindauer, A. and Harvath, T.A. (2014). Pre-death grief in
Medicine, 8: 421–425. the context of dementia caregiving: A concept analy-
Goldberg, L.S. and Altman, K.W. (2014). The role of gas- sis. Journal of Advanced Nursing, 70: 2196–2207.
trostomy tube placement in advanced dementia with Livingston, G., Kelly, L., Lewis-Holmes, E. et al. (2014).
dysphagia: A critical review. Clinical Interventions in Non-pharmacological interventions for agitation
Aging, 9: 1733–1739. in dementia: Systematic review of randomised
Hadjistavropoulos, T., Herr, K., Prkachin, K.M. et al. (2014). controlled trials. The British Journal of Psychiatry,
Pain assessment in elderly adults with dementia. 205: 436–442.
Lancet Neurology, 13: 1216–1227. Low, J.A., Chan, D.K., Hung, W.T. and Chye, R. (2003).
Hall, S., Kolliakou, A., Petkova, H. et al. (2011). Treatment of recurrent aspiration pneumonia in end-
Interventions for improving palliative care for older stage dementia: Preferences and choices of a group
people living in nursing care homes. Cochrane of elderly nursing home residents. Internal Medicine
Database Systematic Reviews, (3): CD007132. Journal, 33: 345–349.
Hirschman, K.B., Kapo, J.M. and Karlawish, J.H. (2008). Mitchell, S.L., Miller, S.C., Teno, J.M. et al. (2010).
Identifying the factors that facilitate or hinder advance Prediction of 6-month survival of nursing home resi-
planning by persons with dementia. Alzheimer Disease dents with advanced dementia using ADEPT vs hos-
and Associated Disorders, 22: 293–298. pice eligibility guidelines. The Journal of the American
Houttekier, D., Cohen, J., Bilsen, J. et al. (2010). Place of Medical Association, 304: 1929–1935.
death of older persons with dementia. A study in five Mitchell, S.L., Teno, J.M., Kiely, D.K. et al. (2009). The
European countries. Journal of the American Geriatrics clinical course of advanced dementia. New England
Society, 58: 751–756. Journal of Medicine, 361: 1529–1538.
Jordan, A., Regnard, C., O’Brien, J.T. and Hughes, Moore, K. and Dow, B. (2014). Carers continuing to
J.C. (2012). Pain and distress in advanced demen- care after residential care placement. International
tia: Choosing the right tools for the job. Palliative Psychogeriatrics, 27 (6): 877–880.
Medicine, 26: 873–878. Moore, K., Ozanne, E., Ames, D. and Dow, B. (2013).
Kitwood, T. (1997). Dementia Reconsidered: The Person How do family carers respond to behavioral and
Comes First. Buckingham, United Kingdom: Adult psychological symptoms of dementia? International
Lives: A Life Course Perspective. Psychogeriatrics, 25: 743–753.
Koopmans, R.T., Lavrijsen, J.C., Hoek, J.F. et al. (2010). Nakanishi, M., Nakashima, T., Shindo, Y. et al. (2015).
Dutch elderly care physician: A new generation An evaluation of palliative care contents in national
of nursing home physician specialists. Journal of dementia strategies in reference to the European
American Geriatric Society, 58: 1807–1809. Association for Palliative Care white paper.
Kovach, C., Kovach, S.A. and Wilson, P.E. (1996). The International Psychogeriatrics, 27 (9): 1551–1561.
effects of hospice interventions on behaviors, Perrar, K.M., Schmidt, H., Eisenmann, Y. et al. (2015).
discomfort, and physical complications of end Needs of people with severe dementia at the end-
stage dementia nursing home residents. American of-life: A systematic review. Journal of Alzheimers
Journal of Alzheimer’s Disease and Other Dementias, Disease, 43: 397–413.
11: 7–15. Pinquart, M. and Sorensen, S. (2003). Differences between
Kovach, C.R., Logan, B.R., Noonan, P.E. (2006a). Effects of caregivers and noncaregivers in psychological health
the serial trial intervention on discomfort and behavior and physical health: A meta-analysis. Psychology and
of nursing home residents with dementia. American Aging, 18: 250–267.
Journal of Alzheimers Disease and Other Dementia, Potter, J.M., Fernando, R. and Humpel, N. (2013).
21: 147–155. Development and evaluation of the REACH (recog-
Kovach, C.R., Noonan, P.E., Schlidt, A.M. et al. (2006b). nise end of life and care holistically) out in dementia
The serial trial intervention: An innovative approach to toolkit. Australasian Journal on Ageing, 32: 241–246.
meeting needs of individuals with dementia. Journal of Reinhardt, J.P., Chichin, E., Posner, L. and Kassabian, S.
Gerontological Nursing, 32: 18–25; quiz 26-17. (2014). Vital conversations with family in the nurs-
Lane, M.J., Davis, D.R., Cornman, C.B. et al. (1998). ing home: Preparation for end-stage dementia care.
Location of death as an indicator of end-of-life costs Journal of Social Work in End of Life and Palliative
for the person with dementia. American Journal Care, 10 (2): 112–126.
of Alzheimer’s Disease and Other Dementias, Reisberg, B. (1988). Functional assessment staging (FAST).
13: 208–210. Psychopharmacology Bulletin, 24: 653–659.
Advanced dementia and care at the end of life 357

Reisberg, B., Ferris, S.H., de Leon, M.J. and Crook, T. Stevenson, D.G. and Bramson, J.S. (2009). Hospice care
(1982). The Global Deterioration Scale for assessment in the nursing home setting: A review of the litera-
of primary degenerative dementia. American Journal ture. Journal of Pain and Symptom Management,
of Psychiatry, 139: 1136–1139. 38: 440–451.
Reyniers, T., Deliens, L., Pasman, H.R. et al. (2014). Sura, L., Madhavan, A., Carnaby, G. and Crary, M.A.
International variation in place of death of older (2012). Dysphagia in the elderly: Management and
people who died from dementia in 14 European and nutritional considerations. Clinical Interventions in
non-European countries. Journal of the American Aging, 7: 287–298.
Medical Directors Association, 16 (2): 165–171. Teno, J.M., Clarridge, B.R., Casey, V. et al. (2004). Family
Robinson, L., Dickinson, C., Bamford, C. et al. (2013). perspectives on end-of-life care at the last place of
A qualitative study: Professionals’ experiences of care. The Journal of the American Medical Association,
advance care planning in dementia and palliative care, 291: 88–93.
‘a good idea in theory but …’. Palliative Medicine, van der Maaden, T., Steen, J.T., Vet, H.C.W. et al. (2014).
27: 401–408. Development of a practice guideline for optimal symp-
Robinson, L., Dickinson C., Rousseau, N. et al. (2012). A tom relief for patients with pneumonia and dementia
systematic review of the effectiveness of advance in nursing homes using a Delphi study. International
care planning interventions for people with cognitive Journal of Geriatric Psychiatry, 30 (5): 487–496.
impairment and dementia. Age Ageing, 41: 263–269. van der Steen, J.T., Hertogh, C.M., de Graas, T. et al.
Rurup, M.L., Onwuteaka-Philipsen, B.D., Pasman, H.R. et (2013a). Translation and cross-cultural adaptation of a
al. (2006). Attitudes of physicians, nurses and rela- family booklet on comfort care in dementia: Sensitive
tives towards end-of-life decisions concerning nursing topics revised before implementation. Journal of
home patients with dementia. Patient Education and Medicine Ethics, 39 (2): 104–109.
Counseling, 61 (3): 372–380. van der Steen, J.T., Onwuteaka-Philipsen, B.D., Knol, D.L.
Sampson, E.L., Candy, B. and Jones, L. (2009). Enteral et al. (2013b). Caregivers’ understanding of dementia
tube feeding for older people with advanced demen- predicts patients’ comfort at death: A prospective
tia. Cochrane Database Systematic Reviews, 2 (2): observational study. BMC Medicine, 11: 105.
CD007209. van der Steen, J.T, Radbruch, L. Hertogh, C.M. et al.
Sampson, E.L., Jones, L., Thuné-Boyle, I.C.V. et al. (2011). (2014). White paper defining optimal palliative care
Palliative assessment and advance care planning in older people with dementia: A Delphi study and
in severe dementia: An exploratory randomized recommendations from the European Association for
controlled trial of a complex intervention. Palliative Palliative Care. Palliative Medicine, 28: 197–209.
Medicine, 25: 197–209. van der Steen, J. T., Toscani, F., de Graas T. et al. (2011).
Schulz, R., Mendelsohn, A.B., Haley, W.E. et al. (2003). Physicians’ and nurses’ perceived usefulness and
End-of-life care and the effects of bereavement on acceptability of a family information booklet about
family caregivers of persons with dementia. New comfort care in advanced dementia. Journal of
England Journal of Medicine, 349: 1936–1942. Palliative Medicine, 14: 614–622.
Seymour, J.E., Kumar, A. and Froggatt, K. (2011). Do Vandervoort, A., Van den Block, L., van der Steen, J.T. et
nursing homes for older people have the support they al. (2013). Nursing home residents dying with demen-
need to provide end-of-life care? A mixed methods tia in Flanders, Belgium: A nationwide postmortem
enquiry in England. Palliative Medicine, 25: 125–138. study on clinical characteristics and quality of dying.
Shega, J.W., Hougham, G.W., Stocking, C.B. et al. (2008). Journal of the American Medical Directors Association,
Patients dying with dementia: Experience at the end 14: 485–492.
of life and impact of hospice care. Journal of Pain and Volandes, A.E., Lehmann, L.S., Cook, E.F. et al. (2007).
Symptom Management, 35: 499–507. Using video images of dementia in advance care plan-
Sleeman, K.E., Ho, Y.K., Verne, J. et al. (2014). Reversal of ning. Archives of Internal Medicine, 167: 828–833.
English trend towards hospital death in dementia: A Volicer, L., Hurley, A.C., Lathi, D.C. and Kowall, N.W.
population-based study of place of death and associ- (1994). Measurement of severity in advanced
ated individual and regional factors, 2001–2010. BMC Alzheimer’s disease. Journal of Gerontology, 49:
Neurology, 14: 59. M223–M226.
Stacpoole, M., Hockley, J., Thompsell, A. et al. (2014). The Wachterman, M., Kiely, D.K. and Mitchell, S.L. (2008).
Namaste Care programme can reduce behavioural Reporting dementia on the death certificates of nurs-
symptoms in care home residents with advanced ing home residents dying with end-stage dementia.
dementia. International Journal of Geriatric Psychiatry. The Journal of the American Medical Association, 300:
30 (7): 702–709. 2608–2610.
358 Dementia

Wada, T., Imai, H., Fukutomi, E. et al. (2014). Preferred Wee, B.L., Coleman, P.G., Hillier, R. and Holgate, S.H. (2006).
feeding methods for dysphagia due to end-stage The sound of death rattle I: Are relatives distressed by
dementia in community-dwelling elderly people in hearing this sound? Palliative Medicine, 20: 171–175.
Japan. Journal of the American Geriatric Society, Westenhaver, T.F., Krassa, T.J., Bonner, G.J. and Wilkie,
62: 1810–1811. D.J. (2010). Advance care plans for CPR or mechani-
Watkin, L., Blanchard, M.R., Tookman, A. and Sampson, cal ventilation in patients with dementia. The Nurse
E.L. (2012). Prospective cohort study of adverse Practitioner, 35: 38–42.
events in older people admitted to the acute gen- World Health Organization (2014). The Top 10 Causes of
eral hospital: Risk factors and the impact of demen- Death. Fact sheet No.310. Available at http://www.
tia. International Journal of Geriatric Psychiatry, who.int/mediacentre/factsheets/fs310/en/. Accessed 2
27: 76–82. June 2016.
36
Alzheimer’s associations and societies

HENRY BRODATY, NICOLE PESA AND GLENN REES

Research activity is the third factor, aimed at improving


36.1 INTRODUCTION diagnosis and assessment, discovering preventative factors
and treatments for dementia and determining the impact
Alzheimer’s disease (AD) associations are voluntary orga- on carers and ways to reduce their burden and stress lev-
nizations that advocate on behalf of people with dementia els. The importance of preventative factors and consequent
(PWD) and their carers. The growth in these associations need for prevention studies was highlighted by the World
has been brought about by a quantum leap of public aware- Alzheimer’s Report (Alzheimer’s Disease International,
ness about AD and related disorders, particularly in devel- 2014) and research activity has been gaining momentum
oped countries. The rise in public awareness has occurred as in this area (Gillette-Guyonnet et al., 2009; Richard et al.,
a result of a number of converging factors. 2009; Kivipelto et al., 2013; Ngandu et al., 2014).
The ‘greying’ of the world’s population or ageing is the The final factor is the breakdown of traditional family
major risk factor for dementia. Average life expectancy in structures with the changing roles of women and adult chil-
developed countries is currently greater than 75 years for dren and changing values that have reduced the moral obli-
males and 80 years for women. Approximately 8% of the gation to care for older members of the community. This
world’s population is aged 65 years and older, and in the has increased pressure on governments and healthcare sys-
United Kingdom people aged 65 years and over comprise tems to support the needs of people with dementia and their
17% of the population (United Nations, 2013). There is esti- carers.
mated to be over 44 million PWD worldwide, and the num- Being a carer of a person with dementia is very stressful,
ber of people affected is expected to surpass 136 million by and it is known that carers experience higher levels of psy-
2050 (Alzheimer’s Disease International, 2014). Moreover, chological distress and lower levels of self-efficacy and sub-
there is a common misconception that dementia only occurs jective well-being than both non-carers and carers of people
in developed nations. The majority of PWD (approximately with other types of diseases and disabilities (Pinquart and
30 million or 60% of the world’s population) live in lower Sorensen, 2003). Carers seek each other out for support and
income countries with this number set to triple to nearly 90 company, and as a result Alzheimer’s associations around
million or 70% of the world’s population by 2050. the world have grown. In developed countries such as the
Second, there has been increasing public awareness of United Kingdom, United States and Australia, Alzheimer’s
dementia and AD and resultant interest and fear of the dis- associations have been a powerful voice for promoting car-
ease. A recent poll in the United Kingdom shows that sig- ers’ and care recipients’ needs.
nificantly more people over 50 are more scared of getting The role played by Alzheimer’s organizations worldwide
dementia than they are of getting cancer. The poll (www. has changed dramatically over the past 10 years. On both
saga.co.uk) showed that while 1 in 10 people surveyed were the international and the national levels, these organiza-
frightened of getting cancer, almost 70% were fearful of tions have transformed to become the agents of change in
developing dementia. This has been fuelled by publicized advocating for national plans to make dementia a health
cases of prominent people who have developed demen- priority. Their common objectives are to improve the qual-
tia (such as Ronald Reagan, Margaret Thatcher, Charles ity of life of PWD through awareness, reducing the stigma
Bronson and Charlton Heston) and medical breakthroughs, that results from a diagnosis of dementia, improving access
including discovery of contributing factors and trials of new to dementia services, promoting community understand-
treatments. ing that the risk of dementia may be reduced and increasing

359
360 Dementia

investment in research to better identify those at risk and distribute. ADI provides its member countries with materi-
delay progression of the disease. als they can adapt to their culture and language.
ADI’s main aims are to build and strengthen member
associations by facilitating the sharing of expertise and
resources and encourage the formation of associations in
36.2 ALZHEIMER’S DISEASE countries where they do not exist. To address these aims,
INTERNATIONAL ADI developed the 3-day annual Alzheimer University
training program in 1998, a series of workshops that aim to
National organizations are brought together under Alzheimer’s strengthen associations by focusing on issues fundamental
Disease International (ADI), a global federation of 86 to running an organization (e.g. organizational structure).
Alzheimer’s associations (as of 2016) (see Figure 36.1). ADI For new and emerging associations, workshops are designed
focuses on a unique combination of global solutions to raise to help them develop the skills to establish an association,
awareness of dementia around the world and local knowledge including identifying aims, fundraising, recruiting volun-
to empower, support and care for PWD locally in each c­ ountry. teers, running support groups, raising awareness, influenc-
Founded in 1984, ADI is governed by a ­council of ­representatives ing public policy, governance and providing and distributing
from each member association that meets annually and elects information (Alzheimer’s Disease International, 2005). ADI
a board and an executive team, led by the chairman. Funding financially supports participants to attend.
comes from membership fees, conference fees, ­donations and ADI disseminates information and its annual World
investments. Ibero-Latin America and Asia-Pacific regional Alzheimer Reports have been hugely influential g­ uiding both
groupings hold their own annual c­onferences and produce health research and policy. They have published reports on the
publications relevant to their zone and ADI has established global prevalence (Alzheimer’s Disease International, 2009)
an Asia-Pacific regional office in Singapore. Europe has an and economic impact (Alzheimer’s Disease International,
independent organization, Alzheimer Europe (http://www. 2010) of dementia, the benefits of early diagnosis and
alzheimer-europe.org/), which collaborates closely with ADI. intervention (Alzheimer’s Disease International, 2011),
­
ADI has close associations with the World Health ­overcoming stigma (Alzheimer’s Disease International, 2012)
Organization (WHO), the International Psychogeriatric and long-term care (Alzheimer’s Disease International, 2013).
Association, World Psychiatric Association and the World The most recent report examined the evidence for risk reduc-
Federation of Neurology. The pharmaceutical ­ industry tion and prevention (Alzheimer’s Disease International,
seeks ADI’s advice about dementia treatment and ­consumer 2014). ADI issues a regular newsletter, fact sheets and
needs. ADI and WHO collaborated to produce a caregiver ­publications on a number of issues relevant to PWD and their
publication, the WHO provides speakers for ADI confer- carers and Alzheimer’s associations. Publications are avail-
ences and the organizations work together to achieve fund- able on the website www.alz.co.uk, and most are ­available in
ing for projects. WHO supported the launch of ADI’s World English and Spanish. Links to websites that provide dementia
Alzheimer’s Day in 1994. Held annually on 21 September, information in other languages are also provided.
associations around the world mark World Alzheimer’s Globally, ADI campaigns for policy change from gov-
Day by holding ‘memory walks’, open days, lectures, train- ernments and the WHO and for recognition of dementia
ing courses and entertainment designed to raise awareness as a public health priority. In March 2015, the First WHO
about dementia. ADI supplies its ­members with a bulletin Ministerial Conference on Dementia was held involv-
that has a special focus each year and materials to which ing 85 countries. There is the prospect of a Resolution at
they can add their own information, download, print and the World Health Assembly on Dementia in May 2017.
Twenty-five national dementia plans are in place (2016).
An important part of ADI advocacy is the partnership
with Dementia Alliance International, an organization
of people with dementia. An annual international confer-
ence brings together PWD, their families, clinicians, care
professionals and researchers to share and exchange infor-
mation, resources and knowledge. From its 1985 inception
in Brussels, the conference has developed into a significant
international stage to raise awareness about AD and show-
case the growing unified effort and support for progress in
dementia policy and research.
Locally, ADI supports national Alzheimer’s associations
Figure 36.1 Map of the world with those countries
to effectively advocate for PWD and their caregivers by
belonging to Alzheimer’s Disease International shown
in dark grey. (From Alzheimer’s Disease International, equipping them with the skills and know-how to provide
World Alzheimer Report 2009: The Global Prevalence of high quality services.
Dementia, Alzheimer’s Disease International, London, The Twinning Programme launched in 2005 is another
United Kingdom, 2009.) ADI initiative that embraces peer-to-peer sharing of
Alzheimer’s associations and societies 361

knowledge, skills and experience between Alzheimer’s Associations’ (or similar) to Alzheimer’s associations even
associations from different countries. The vision for the though other dementias are covered.
programme is that the partnerships created will strengthen Core functions that Alzheimer’s associations across the
both associations by tackling issues together. world aspire to deliver include the following:
ADI facilitates a research network, the 10/66 Dementia
Research Group that carries out population-based research ●● Raising public awareness to promote an understanding
on dementia and its risk factors in lower- and middle- of dementia and reduce the stigma associated with the
income countries, including China, India, Russia, Nigeria, condition
South Africa, Mexico and other countries, especially in ●● Providing support services, education and information
Latin America (www.alz.co.uk/1066). The group, estab- educating carers and health professionals
lished in 1998, has published over 100 scientific papers in ●● Circulating publications and resources
peer-reviewed journals. ●● Advocating to governments for improved health
The 10/66 name derives from the fact that less than 10% systems that recognize the need for timely diagnosis,
of all population-based research into dementia is directed improved care in hospitals, access to palliative care
towards the 66% of all PWD who live in developing countries ●● Advocating for more investment into research
and was formed to redress the balance. Funding comes partly
from ADI and mostly from grants; it has endorsement from The capacity of individual Alzheimer’s associations to
WHO. 10/66 has published results of studies from 28 centres undertake these roles varies enormously between developed
in India, China, southeast Asia, Latin America, Nigeria and and developing countries. Some associations in many devel-
Russia and randomized controlled trials (RCTs) of caregiver oping countries have no association or if they do have only
interventions in Russia, India, Venezuela, Peru, Dominican a few volunteers to provide information and assistance. At
Republic and China, Mexico, Chile and Argentina. the other extreme are organizations in developed countries
Interventions were designed to provide basic education with sophisticated awareness activities, leading edge ser-
about dementia and specific training on managing prob- vices and capacity to invest in dementia research.
lem behaviours. Three simple modules were delivered over The evolution of Alzheimer’s associations across the world
5 half-hour sessions, targeted at the main carer and also has followed a similar pattern from an initial focus on family
involving other family members (Dias et al., 2008; Gavrilova carers to being inclusive of PWD of all ages; from a priority
et al., 2008; Guerra et al., 2010). for awareness and information to the provision of services and
Epidemiological studies involve surveys of residents training and education; from consumer resources to sophis-
aged 65 years and older with sample sizes of at least 2,000 ticated publications on dementia; from media to developing
in each Latin American country, with an ultimate total policy and working with governments on dementia national
sample of 25,000 (10/66 Dementia Research Group, 2008). plans. This evolution has gathered pace in recent years and has
The 10/66 group developed and validated a one-phase edu- put great strain on the resources of the associations.
cation- and culture-fair dementia diagnostic algorithm to Many countries now have national plans with the
be used in older populations with little formal education aim of making dementia a health priority including
(Prince et al., 2003, 2008) and to obtain information about Australia, South Korea, France, United Kingdom, Norway,
care arrangements for PWD (Prince, 2004), the impact United States, Netherlands, Finland, Denmark, Belgium,
of caring on their carers and common behavioural prob- Luxembourg, Switzerland, Israel, Taiwan, Japan, Mexico,
lems (10/66 Dementia Research Group, 2004) and associa- Costa Rica and Cuba. The elements of these plans vary but
tions with subjective memory complaints (10/66 Dementia often include awareness and information, timely diagnosis,
Research Group, 2009). Qualitative research studies have access to ­community and residential services, improved care
demonstrated a lack of awareness in these countries that in ­hospitals and training and education.
dementia is an organic brain disease and a belief that These plans are important not only in achieving need for
symptoms were a normal part of aging (Shaji et al., 2003). action on dementia but promoting partnerships between
Member associations actively support medical research Alzheimer’s organizations, health and aged care sectors and
in their countries and ADI’s Medical and Scientific the research community.
Advisory Panel provides expert advice and acts as interna- Important to the operation of many Alzheimer’s organi-
tional ambassadors for ADI. zations are consumer networks to empower PWD and their
carers to advocate for themselves. Advocacy and empower-
ment through person-centred and consumer-directed care
is a necessary shift in care culture, and the recognition that
36.3 NATIONAL ORGANIZATIONS PWD should be consulted and have choices about the care
they receive is a requisite for good dementia care. The philos-
The main aim of all Alzheimer’s associations is to improve ophy of person-centred care is based on the idea that instead
the quality of life for PWD and their families (http://www. of providing the same care to every person, care should
alz.co.uk/adi/). Many of these associations changed their be tailored to the needs and preferences of the individual.
names from ‘Alzheimer Disease and Related Disorders Consumer-directed care takes this concept further and
362 Dementia

recognizes that the consumer, to the extent they are capable, professionals. Often people who manage and care for PWD
should make choices about the types of care services they both at home and in healthcare settings are untrained
access and the delivery of those services. These care philoso- or undertrained and rely on programmes developed by
phies are important across all care services including respite Alzheimer’s associations. Even though education alone
care, a major care service in Western countries. While con- has limited benefits (Brodaty, 1994; Brodaty et al., 1994;
sumer choice is gaining momentum globally, there are still Marriott et al., 2000), it is an integral part of any interven-
concerns about issues in care such as physical and medical tion and combined with counselling has been found to
restraint. Together, ADI and individual national Alzheimer’s improve carer depression, burden, social support and self-
associations reinforce an international dedication to improv- efficacy (see Chapter 14 and particularly: [Belle et al., 2006;
ing the lives of people living with dementia, finding a cure for Mittelman et al., 2006]).
the disease and continuing the fight against dementia. In 2012/2013, the UK Alzheimer’s Society piloted an
Associations vary in their governance and ­ service intergenerational exchange initiative in an effort to foster
­provision. Associations may be centralized with a national dementia-friendly generations and communities. Schools
body and branches through the country (e.g. United Kingdom) and colleges participated in the programme, which aimed
or may have a federated structure, with a­ utonomous organi- to educate children and young people, remove stigma and
zation in each state or province, which join together to form provide opportunities for interaction with PWD. An evalu-
a national body (e.g. Canada, Australia). While membership ation of 13 of the 22 participating schools found key ben-
is open to anyone with an interest in dementia, associations efits including increased awareness of dementia and how to
mainly cater for c­ arers and PWD. Many people working in assist PWD to live well, reduced stigma and fear, recogni-
the dementia area also join as do those who wish to lend their tion of the importance of dementia education in schools,
support. Most ­associations strive to have a broad cross section and knowledge of carer support (Atkinson and Bray, 2013).
of the ‘dementia community’ on their boards – consumers The impact of the programme spread beyond that of demen-
i.e. carers and (increasingly) PWD, service providers, ­doctors tia awareness, spilling into areas such as personal develop-
and people with influence who are interested from the g­ eneral ment and extended beyond the school community and have
community. It is generally considered desirable that consum- since been compiled into a Dementia Resource Suite. Over
ers constitute at least half of the board. 140 schools delivered a dementia awareness program in
Standard services provided by Alzheimer’s associations 2013/2014 and the initiative is set to grow.
are support groups, free telephone help lines, telephone The UK Alzheimer’s Society runs a number of other
counselling, information via the web, education, training, training programmes designed for PWD, informal and
public awareness, media campaigns and public advocacy. professional carers that cover topics including introduction
Additionally, associations may provide in-person counsel- to dementia care, ideas for activities for PWD, nutrition,
ling, drop-in centres, speaker panels, opportunities for vol- improving communication, facilitating the move to residen-
unteers and funding for research. tial care and dealing with grief and other emotions, as well
as train-the-trainer programmes. They are run at various
locations and can be tailored for individual workplaces and
36.4 INFORMATION run in-house (such as nursing homes). The society also pub-
lishes the training materials (Alzheimer’s Society, 2009b).
Alzheimer’s Australia developed national Dementia and
Provision of information is one of the most important aims of
Memory Community that provides information services
Alzheimer’s associations. They assist PWD, families, carers,
about dementia and preventative strategies, carer educa-
health professionals, governments and members of the public
tion courses, conferences for families and professionals
to obtain information about the disease, how to manage legal
and awareness raising activities. In 2012, the Australian
issues and problem behaviours and how to access community
Department of Health and Ageing funded the National
and health services. Sources of information include websites,
Younger Onset Dementia Key Worker Program. The pro-
factsheets, access to research publications, newsletters, infor-
gramme provides individually tailored education and sup-
mation sessions, support group meetings and telephone ‘hot-
port to both individuals and service providers by providing
lines’. To be able to offer these services, associations require
them with a direct point of contact through a key worker. The
an office, access to appropriate technology and staff or volun-
Living with Memory Loss programme for carers and PWD
teers. Collaboration between physicians and associations can
involves dementia education, information about treatment
improve the quality and amount of information provided to
options and services, strategies and emotional support, con-
PWD and their carers (Fortinsky et al., 2002).
ducted over 7 weekly 2-hour sessions. In an uncontrolled
evaluation of the programme of 339 carers and 231 PWD
participating in 41 groups, significant improvements in
36.5 TRAINING general mental health scores, more positive perceptions of
caregiving, reduced depression scores and decreased stress
Alzheimer’s associations coordinate and run training and from problem behaviours were evident for carers either
education programmes for PWD, their carers and health immediately or 3 months following the programme and for
Alzheimer’s associations and societies 363

depression scores for PWD at the end of the programme geriatric care and orientation to dementia care for staff at aged
and at follow-ups (Bird et al., 2005). care facilities. The Lanka Alzheimer’s Foundation in Sri Lanka
The U.S. Alzheimer’s Association offers a number of runs periodic workshops and in-house training programmes
professional training programmes and workshops for for- for formal and informal carers on a ­one-to-one basis (Lanka
mal and informal carers such as ‘Foundations of Dementia Alzheimer’s Foundation, 2009). In 2013, Alzheimer’s Pakistan
Care’ and ‘Activity Based Dementia Care’. Available Rawalpindi Chapter introduced a 2-day course on caregiving
­programmes vary by state. In Cleveland, OH, primary care containing information on ­general care, respite care, current
physicians collaborated with the Cleveland chapter of the treatments and genetic aetiology. Participants received a cer-
association to provide individualized ­education to family tificate upon completion.
caregivers and capable care recipients r­egarding d ­ ementia
care and available services in the community. In an
­evaluation study involving 44 participants who completed
the full ­intervention, the programme led to enhanced self- 36.6 SERVICES
efficacy for ­managing dementia ­symptoms and ­accessing
community ­
­ services and high levels of ­ satisfaction Services used by carers include home care, day care, in-home
(Fortinsky et al., 2002). The Alzheimer’s ­association has (sitting), residential respite and permanent care. Carers
also developed online ­professional ­training programmes have different needs and finding the right solution can help
and e-workshops. These include essentiALZ, an individual reduce carer stress and depression and delay or make insti-
training ­programme offered to both family and ­professional tutionalization of the care recipient less likely (Callahan
carers that provide ­certification in quality dementia care. et al., 2006; Mittelman et al., 2006). Meeting the service
There are two ­certification options: Dementia Advanced needs of carers is difficult because of limited resources.
Care and Dementia-Related Behaviour. The first c­overs Despite the demands on dementia carers and their high
basic dementia and person-centred care, ­communication, levels of stress and burden, even when services are available,
diet, recognizing pain and minimization of falls. The s­ econd their use is low (Brodaty et al., 2005). Impediments to using
covers approaches and key responses to ­dementia-related services were perceived lack of need, resistance to accept-
behaviours. Both courses involve prerequisite ­completion ing help, not having enquired, lack of knowledge, lack of
of 8–10 hours of various CARES® programmes offered by availability, previous bad experiences, ineligibility and cost.
the ­association and recertification is required every 2 years. Many carers who denied needing services displayed other
Another ­programme offered under the CARES® umbrella is indications of need, such as low levels of life satisfaction and
CARES: A Dementia Caregiving Approach for Direct Care satisfaction with their role and high levels of resentment
Workers and Dementia Care Training for Team Leaders. Ten and overload (Brodaty et al., 2005). Alzheimer’s associa-
1-hour modules, using an online interactive tool, have been tions have an important role in providing, enhancing and
designed for nurses, social workers and other ­professionals facilitating use of services for PWD and their families. They
who work with PWD. It covers issues relating to caring can also inform governments how to deliver and monitor
for, communicating with and engaging PWD in mean- services with reasonable standards of care at economic cost.
ingful activity. Dementia Care Training for Team Leaders For example, the Alzheimer Society of Finland examines
­provides guidance for those who supervise people who care these issues through a survey of the social and healthcare
for dementia patients, such as nursing home administra- sectors every 5 years in their national memory barometer.
tors and unit coordinators but not basic care issues. These The survey elicits information on the standard of services
resources can be purchased through the U.S. Alzheimer’s provided to PWD and their families (Sirpa et al., 2011).
Association website (Alzheimer’s Association, 2009b).
Many other associations run programmes such as the
Singapore’s Alzheimer Disease Association’s (http://www.
alz.org.sg/) training programme for family caregivers. The
36.7 RESEARCH
16-hour programme contains four core modules including
understanding person-centred care, behaviour of PWD, com- Research in AD and other dementias is critical, but glob-
munication, community resources and self-care for caregivers. ally is underfunded in relation to disease burden or in many
Alzheimer’s associations in low- and middle-income coun- countries funding is completely lacking. In Australia, for
tries are increasingly developing training programmes to raise every seven research projects funded in cancer, only one is
awareness and educate carers and healthcare workers about funded in dementia (Anstey and Kiely, 2012). In the United
dementia. The Alzheimer’s and Related Disorders Society of Kingdom, the government invests eight times more in can-
India launched a Comprehensive Dementia Care Program in cer than dementia research (Alzheimer’s Society, 2009a). The
Cochin in 2004. The 10-month training programme includes World Alzheimer Report (Alzheimer’s Disease International,
nursing, community health and welfare and the challenges 2009) ­ highlighted that worldwide research funding is
of geriatric nursing in dementia (Alzheimer’s and Related skewed to disease with high levels of mortality such as can-
Disorders Society of India, 2009). They have expanded their cer and heart disease while those with high levels of disease
training programmes to include 6- and 12-month courses in burden are under-resourced. Alzheimer’s associations act
364 Dementia

as advocacy groups to pressure funding bodies and govern- World Dementia Envoy. The United Kingdom and European
ments to make funding available for research. Additionally, Union have substantially increased their research funding.
they facilitate researchers’ access to PWD and their families
and give advice on research projects, particularly about the
carer p ­ erspective. The UK Alzheimer’s Society has a large
­public ­involvement in dementia research, ­investing over £6 36.9 SUPPORT
million ­annually, funding over 100 projects at ­universities
and other ­institutions and w ­ orking in ­partnership with the Emotional and practical supports are offered through
­scientific community and people affected by dementia. The carer groups or individually through in-person or tele-
Quality Research in Dementia network of 80 ­community phone counselling. Support groups are the backbone of
­volunteers works with s­ cientists to set the research agenda Alzheimer’s associations (and may be offered by residential
and award grants to ensure the best projects, which facilities and other organizations).
are likely to have the ­ biggest impact, receive funding Support groups, also called self-help groups, are gather-
(Alzheimer’s Society, 2015). The Alzheimer’s association ings of people who come together with a common problem
in the United States facilitates dementia research by offer- or goal to provide support to members, share their expe-
ing grants, p ­ ublishing several peer-reviewed journals, the riences, problems, ideas and strategies, learn and gather
flagship being the monthly Alzheimer’s & Dementia: The information about a topic and encourage each other to
Journal of the Alzheimer’s Association, hosting the annual take care of their own health and well-being (Alzheimer’s
Alzheimer’s Association International Conference and con- Disease International, 2000). They are available for most
vening a Research Roundtable, a consortium of Alzheimer’s chronic conditions and clinicians should be aware and able
association staff, advisors, scientists, academics and gov- to provide relevant information about them.
ernment a­ gencies. The roundtable facilitates the develop- The advantages of support groups are as follows:
ment and ­implementation of new dementia treatments and
sponsors the association grants (Alzheimer’s Association, ●● Support, both emotional and practical
2009c). Researchers help Alzheimer’s associations by advis- ●● Education
ing, contributing to publications, submitting research ideas ●● Universalization – a sense of belonging and not being
for funding consideration and delivering presentations. alone
●● Identity and a sense of purpose

However, they are not appropriate for everyone – some


people may feel uncomfortable sharing their feelings in a
36.8 PUBLIC AWARENESS group setting. Support groups are not clinical and are dis-
tinct from group psychotherapy sessions, as members are
Alzheimer’s associations help raise awareness about demen- not viewed as patients. They are usually led by a profes-
tia, which increases public interest and coverage in the sional such as a psychologist and/or someone who is a carer.
media and fosters recognition of dementia as a priority area Professional leaders of groups have relevant qualifications
of concern. This is achieved through publications, courses, and training, whereas peer group leaders tend to rely more
training materials, funding and contributing to research on experience. Regardless, a group leader needs appropriate
and conferences and posting information on websites. Some skills, including public speaking, encouraging participa-
associations are involved in lobbying politicians, form- tion, listening without judging, ensuring respect of group
ing links with professional associations, or engaging high- rules and identifying individuals requiring more support
profile personalities who hugely raise public awareness, (Alzheimer’s Disease International, 2000). Particularly for
help destigmatize dementia and increase research funding. peer-led groups, it may be appropriate to have two leaders
For example, in the United States, former President Ronald so that if a member requires individual support during a
Regan publicly announced his diagnosis of AD, in New session, or if the leader becomes unavailable e.g. if caring
Zealand a well-recognized former football captain received responsibilities need attention, the group does not become
much media attention and in Australia, a famous media leaderless (Alzheimer’s Disease International, 2000).
personality became president of Alzheimer’s Australia and Alzheimer support groups are usually provided for car-
then Australian of the year. In response to a movement run ers and increasingly for people with (early stage) dementia,
by Alzheimer Europe and its member organizations, on 16 or for both PWD and carers. The timing, structure and con-
December 2008 the Council of the European Union recog- tent vary between groups that often meet monthly for 1–2
nized that AD constitutes a ‘priority for action’ (Council of hours, sometimes highly structured with set discussion top-
the European Union, 2008; Alzheimer Europe, 2009); and ics, videos and guest speakers or unstructured to allow for
in 2011, the United Nations Summit on Non-Communicable more informal interaction. Some groups are located next to
Diseases (NCD) declared AD a major disease area (United day centres that have activities for patients while carers attend
Nations, 2011). In 2013, the G8 made dementia a priority and the support group. Anecdotally, some carers benefit greatly
subsequently appointed a World Dementia Council and a from support groups and attend regularly for months or years
Alzheimer’s associations and societies 365

(Ebenstein, 2004). Others find they are unsuitable and others Chu et al., 2011), higher subjective well-being (Pinquart
may attend irregularly, during a particularly stressful time and Sorensen, 2006), better self-reported general health
or only when invited. Carers who never attend may feel that (Javadpour et al., 2008), higher morale (Gonyea, 1990),
they have sufficient support (e.g. social, counselling) or may enhanced competence and empowerment (Chan and
be unaware of the existence of groups or how to access them. O’Connor, 2008) and increased use of formal support ser-
Emerging technology has allowed support to be pro- vices (Ebenstein, 2004).
vided in ways other than face-to-face, such as by telephone The efficacy of support groups is equivocal. In Pinquart
or online. This is important for reaching isolated carers par- and Sorenson’s (2006) systematic review, only one of the
ticularly in rural areas (O’Connell et al., 2014), and to allow support group interventions (number of support interven-
for carers to access support at any time and from any loca- tions reviewed not specified) resulted in a significant posi-
tion. Interventions include conference calling among family tive effect on one outcome measure, subjective well-being.
members, telephone support systems with automated mes- Parker et al.’s (2008) systematic review of carer interven-
sages, stress monitoring and advice, respite calls for care tions included three support group interventions. A support
recipients, online discussion groups, electronic reminder group led by a nurse over 12 weeks, which included psycho-
services, computer-based forums and question and answer logical support, education and problem solving reported
sessions (Internet and non-Internet-based networks), email, significant reductions in distress and improvements in
electronic encyclopaedias and libraries, online virtual envi- quality of life for participants (Fung and Chein, 2002). In
ronments using avatars and computer-based decision sup- the other two studies support group participation had no
port modules (McClure and Sanders, 2008; Powell et al., significant effect on participants’ depression or self-esteem
2008; Martindale-Adams et al., 2013; O’Connor et al., 2014). scores (Pillemer and Suitor, 2002) or on neuropsychiatric
Like carers, PWD may need support and people to talk to symptoms of the participants’ care recipients (Senanarong
who are going through the same experiences, especially in et al., 2004). Telephone and Internet-based support inter-
the early stages of the illness. The presumption that PWD are ventions were found to enhance carers’ and care recipients’
unable to benefit from support groups due to lack of aware- quality of life and independent living skills (Eisdorfer et al.,
ness of their disease and its implications is erroneous. A small 2003; Topo, 2009). Support groups are most likely to have
number of studies have examined the effectiveness of support positive effects if they include a counselling component and
groups for PWD and a recent systematic review found that individualized strategies rather than only providing edu-
they may reduce depression and improve quality of life and cation and general training (Selwood et al., 2007). Groups
self-esteem (Leung et al., 2014). These groups require careful led by professionals tend to produce greater improvement
planning and should be led by trained facilitators (Alzheimer’s in participants’ psychological functioning, while peer-led
Disease International, 2003; Beattie et al., 2007). Examples groups increase informal support networks and feelings
include the online ‘virtual’ group The Dementia Alliance of self-efficacy (Toseland et al., 1989). Carer variables are
International (www.dementiaallianceinternational.org) and also important. Longitudinal evidence suggests that sup-
the ‘Memory club’ a support group for people with early- port group attendance is predicted by caregiver perceptions
stage dementia and their carers (Zarit et al., 2004). Following of similarity between themselves and group members and
the Netherlands, Alzheimer’s associations in the United the amount of social support offered (Steffen and Mangum,
Kingdom, Ireland and Australia have developed ‘Alzheimer 2012). Participation is likely to be more beneficial for carers
cafes’ for people with early-stage dementia and their families. whose social networks do not include people who under-
Their purpose is to provide social and emotional support, and stand their experiences, and carers who are more stressed,
a forum for informal advice and consultations with staff from are dissatisfied with their caregiving role, are employed or
the associations (Bailey and Moriarty, 2006; Mather, 2006). have care recipients who are more apathetic or in nursing
People with early-stage dementia have been able to be suc- homes (Cuijpers et al., 1996; Pillemer and Suitor, 2002).
cessfully integrated into these programmes and anecdotal A major barrier to carer attendance at support groups,
evidence indicates that they enjoy the sessions and find them reported by 80% of carers, is that they had not received advice
beneficial (Mather, 2006). encouraging them to attend (Molinari et al., 1994). The main
ways support groups build and maintain membership are
through general public awareness and outreach to individuals
who share the group’s concern but are not currently attend-
ing (Wituk et al., 2002). Groups tend to be more successful
36.10 THE EFFICACY OF SUPPORT
when they have specific strategies to target and recruit poten-
GROUPS tial members (Wituk et al., 2002). It is also the responsibility
of health professionals such as general practitioners (GPs) to
Participation in support groups has been found to have provide carers with information about support groups; and
a positive impact on the well-being of carers (Chien communication problems (including use of jargon, lack of
et al., 2011). Specifically, it has been found to be associ- face-to-face contact) between the carer and health professional
ated with lower levels of carer burden (Parker et al., 2008; can impede such referrals (Bruce et al., 2002; Carpentier et al.,
Chien et al., 2011) and depression (Chien et al., 2011; 2008). Recommendations by GPs about joining supportive
366 Dementia

organizations (Donath et al., 2010) improved communication determine whether similarly positive outcomes would be
between health professionals and carers, and active outreach found for other minority group participants in a culturally
by support groups may facilitate their use. diverse group.
Support groups can be made more appropriate and
attractive to a range of cultures through the following:

36.11 THE IMPACT OF CULTURAL ●● Active outreach by groups to culturally diverse


FACTORS communities
●● Group leaders addressing from the outset the reluctance
of some cultures to ‘speak out’ about negative feelings
In Western countries such as the United States, Australia about the caregiving role, normalizing feelings of guilt
and the United Kingdom, ethnic minority groups are and reframing traditional beliefs about speaking out
under-represented in the use of community services such (Chan and O’Connor, 2008)
as support groups (Henderson et al., 1993; Chan and ●● Recruiting and training ethnic support group leaders
O’Connor, 2008; Low et al., 2009). While it is unclear ●● Finding a culturally neutral site for the group to meet
why carers from minority groups are less likely to attend ●● Letting participants decide the form of the group
support groups, a number of contributing factors may be (Henderson, 1992; Henderson et al., 1993)
the diverse interpretations of dementia and the stigma
attached to the disease in some cultures, programmes may A number of studies have trialled supportive interven-
be culturally insensitive or noninclusive in their approach, tions for family carers in low- and middle-income coun-
and minority group experiences and expectations may tries. A 12-week support group for dementia caregivers
differ from the other ‘mainstream’ participants (Chan and in Taiwan was found to reduce depression levels after the
O’Connor, 2008). Cultural differences occur at multiple intervention and at 1-month follow-up (Chu et al., 2011).
levels (e.g. intrapersonal, interpersonal and environmen- A Thai intervention including support group participa-
tal) and in multiple domains (e.g. psychosocial health, life tion (Senanarong et al., 2004) helped to decrease neuro-
satisfaction, caregiving appraisals, spirituality, coping, psychiatric symptoms of care recipients and had a small
self-efficacy, physical functioning, social support, filial effect on reducing carer distress (see Chapter 14). In Iran,
responsibility, familism, views towards elders and use of 29 female carers attended 8 weekly sessions in a support
formal services and healthcare) (Napoles et al., 2010). For group conducted by a trained senior psychiatry resident
instance, ethnic minority groups may find it difficult to involving education and interactive group activities based
relate to mainstream support group practice norms such on the Alzheimer’s Association Guidelines for Carers. Post-
as questioning authority and discussing conflict and feel- intervention scores showed highly significant reductions in
ings towards caregiving (Diaz, 2002). perceived stress and general health of carers and the neu-
In Australia, dementia is five times more prevalent in ropsychiatric symptoms of their care recipients (Javadpour
aboriginal Australians than non-aboriginal Australians, et al., 2008). There was no longitudinal follow-up and addi-
yet there is a severe lack of services available for Aboriginal tional research would help to consolidate these findings.
PWD and their families including caregiver support (Smith
et al., 2011). Smith et al. (2011) identified a number of factors
that would facilitate access to care, including implemen-
tation of community-based and culturally safe services, 36.12 SPECIAL SUPPORT GROUPS
increased Aboriginal workforce within services, cultural
training for staff external to the community and increased People with younger-onset dementia or uncommon forms
education and awareness of dementia. of dementia, gay couples affected by dementia, people in
Despite these limitations, there is evidence that diverse second (or later marriages) or people who are very promi-
cultural groups can benefit from participating in sup- nent in their community and their families may find that
port groups. A U.S. study reported positive psychological mainstream support groups are unsuitable (Moore, 2002;
benefits for a culturally tailored support group for Latino Chaston et al., 2004; Arai et al., 2007). Some Alzheimer’s
caregivers, who are heavily under-represented in the associations (e.g. Alzheimer’s Australia) have developed
healthcare system (Gonyea et al., 2014). Chinese people, support groups tailored specifically for groups such as those
who have culturally defined caregiving roles based on the with younger-onset dementia, those in the early stages of
notion of filial piety and are traditionally known to be dementia and their families, multicultural carers, male car-
uncomfortable discussing personal problems, were able ers, adult children of PWD and people with frontotemporal
to openly express their feelings and difficulties in a sup- lobe dementia and Pick’s disease (Alzheimer’s Australia,
port group environment with other carers who had simi- 2005, 2009). The U.S. Alzheimer’s Association has online
lar experiences and experienced significant reductions support groups for Spanish-speaking carers and carers
in distress levels compared to controls (Fung and Chein, who have lost their care recipient (Alzheimer’s Association,
2002; Leung et al., 2014). Further research is required to 2009a).
Alzheimer’s associations and societies 367

Alzheimer’s Disease International (2000). Starting a


36.13 CONCLUSIONS Self-Help Group. London, UK: Alzheimer’s Disease
International.
Alzheimer’s associations are a major support for PWD and Alzheimer’s Disease International (2003). How to Include
their families and have shaped policy internationally and People with Dementia in the Activities of Alzheimer
nationally, and raised awareness among policy makers, cli- Associations. London, UK: Alzheimer’s Disease
nicians and the general public. They are a key part of the International.
awareness, political and societal processes to destigmatize Alzheimer’s Disease International (2005). The Alzheimer
dementia, to assure adequate services and to improve the University. Available at http://www.alz.co.uk/adi/alzuni.
quality of life of those living with dementia. The outcomes html. Accessed 15 January 2015.
of the activities of these associations can be seen in the Alzheimer’s Disease International (2009). World
implementation of national plans, and most recently, in the Alzheimer Report 2009: The Global Prevalence
commitment of the G7 Dementia Summit to develop an of Dementia. London, UK: Alzheimer’s Disease
international dementia research action plan. While recog- International.
nizing their substantial achievements, there is still difficulty Alzheimer’s Disease International (2010). World
in attracting necessary funding and establishing well-inte- Alzheimer Report 2010: The Global Economic Impact
grated care. of Dementia. London, UK: Alzheimer’s Disease
Alzheimer’s associations are an important therapeutic International.
intervention in their own right and should be part of the Alzheimer’s Disease International (2011). World Alzheimer
armamentarium of clinicians helping those who live with Report 2011: The Benefits of Early Diagnosis and
dementia. It may be helpful for clinicians to ‘prescribe’ con- Intervention. London, UK: Alzheimer’s Disease
tact with an Alzheimer’s association as part of a manage- International.
ment package. The rise and development of associations Alzheimer’s Disease International (2012). World Alzheimer
around the world is testimony to their value. Report 2012: Overcoming the Stigma of Dementia.
London, UK: Alzheimer’s Disease International.
Alzheimer’s Disease International (2013). World Alzheimer
Report 2013: Journey of Caring: An Analysis of Long-
REFERENCES term Care for Dementia. London, UK: Alzheimer’s
Disease International.
10/66 Dementia Research Group (2004). Behavioural and Alzheimer’s Disease International (2014). World
psychological symptoms of dementia in developing Alzheimer’s Report 2014: Dementia and Risk
countries. International Psychogeriatrics, 16: 1–19. Reduction: An Analysis of Protective and Modifiable
10/66 Dementia Research Group (2008). Population-Based Factors. London, UK: Alzheimer’s Disease
Epidemiological Studies (Prevalence Phase). Available International.
at http://www.alz.co.uk/1066/pop_based_ep_studies_ Alzheimer Europe (2009). Making Dementia a Public
prevalence_phase.php. Accessed 15 January 2015. Health Priority 2006. Available at www.dementia-in
10/66 Dementia Research Group (2009). Subjective -europe.eu/. Accessed 15 January 2015.
memory deficits in people with and without dementia: Alzheimer’s and Related Disorders Society of India (2009).
Findings from the 10/66 dementia research group pilot Comprehensive Dementia Care Program at Cochin.
studies in low- and middle-income countries. Journal of Available at http://www.alzheimer.org.in/. Accessed
the American Geriatrics Society, 57: ­2118–2124. 15 January 2015.
Alzheimer’s Association (2009a). Alzheimer’s Association Alzheimer’s Society (2009a). Our Research Programme.
Online Community. Available at http://www.alz. Available at http://www.alzheimers.org.uk/site/scripts/
org/professionals_and_researchers_education.asp. documents.php?categoryID=200307. Accessed 15
Accessed 15 January 2015. January 2015.
Alzheimer’s Association (2009b). Professional Training Alzheimer’s Society (2009b). Training. Available at http://
and Education Programs. Available at http://www.alz. www.alzheimers.org.uk/site/scripts/documents_info.
org/professionals_and_researchers_researchers.asp. php?categoryID=200294&documentID=347. Accessed
Accessed 15 January 2015. 15 January 2015.
Alzheimer’s Association (2009c). Research Centre. Alzheimer’s Society (2015). Join the Research Network.
Available at http://www.alzheimers.infopop.cc/eve. Available at http://www.alzheimers.org.uk/site/scripts/
Accessed 15 January 2015. documents_info.php?documentID=1109. Accessed
Alzheimer’s Australia (2005). Next Steps: Younger Onset 6 January 2015.
Dementia. Canberra, Australia: Alzheimer’s Australia. Anstey, K. and Kiely, K. (2012). Evaluation of NHMRC Data
Alzheimer’s Australia (2009). Services and Support. on the Funding of Dementia Research in Australia. A
Available at http://www.alzheimers.org.au/content. Report for Alzheimer’s Australia. Canberra, Australia:
cfm?categoryid=9. Accessed 15 January 2015. Alzheimer’s Australia.
368 Dementia

Arai, A., Matsumoto, T., Ikeda, M. and Arai, Y. (2007). Do Chan, S.M. and O’Connor, D.L. (2008). Finding a voice:
family caregivers perceive more difficulty when they The experiences of Chinese family members partici-
look after patients with early onset dementia com- pating in family support groups. Social Work with
pared to those with late onset dementia. International Groups, 31: 117–135.
Journal of Geriatric Psychiatry, 22: 1255–1261. Chaston, D., Pollard, N. and Jubb, D. (2004). Young onset
Atkinson, T. and Bray, J. (2013). Dementia Awareness and dementia: A case for real empowerment. Journal of
Intergenerational Exchange in Schools: A Pioneer Project Dementia Care, 12: 24–26.
supporting Dementia Friendly Communities: Report by Chien, L.Y., Chu, H., Guo, J.L. et al. (2011). Caregiver
the Association of Dementia Studies at the University support groups in patients with dementia: A meta-
of Worcester for Alzheimer’s Society United Kingdom. analysis. International Journal of Geriatric Psychiatry,
London, UK: Alzheimer’s Society United Kingdom. 26: 1089–1098.
Bailey, J. and Moriarty, J. (2006). Alzheimer’s Society Chu, H., Yang, C.Y., Liao, Y.H. et al. (2011). The effects of
Derby branch: A comprehensive programme of a support group on dementia caregivers’ burden and
support for people with dementia and their carers. depression. Journal of Aging and Health, 23: 228–241.
Dementia: The International Journal of Social Research Council of the European Union (2008). Council
and Practice, 5: 293–296. Conclusions on Public Health Strategies to Combat
Beattie, E., Bossen, A., Boyne, S. et al. (2007). Review Neurodegenerative Diseases Associated with Ageing
of Scientific Evidence Addressing Prevalence, and in Particular Alzheimer’s Disease. Brussels,
Documented Needs, and Interdisciplinary Research: Belgium: Council of the European Union.
Persons in Early Stage Alzheimer’s Dementia. Chicago, Cuijpers, P., Hosman, C.M.H. and Munnichs, J.M.A. (1996).
IL: National Alzheimer’s Association. Change mechanisms of support groups for caregivers
Belle, S., Burgio, L., Burns, R. et al. (2006). Enhancing the of dementia patients. International Psychogeriatrics,
quality of life of dementia caregivers from different 8: 575–587.
ethnic or racial groups. Annals of Internal Medicine, Dias, A., Dewey, M.E., D’Souza, J. et al. (2008). The
145: 727–738. effectiveness of a home care program for supporting
Bird, M., Caldwell, T., Maller, J. and Korten, A. (2005). caregivers of persons with dementia in developing
Alzheimer’s Australia Early Stage Dementia Support countries: A randomised controlled trial from Goa,
and Respite Project: Final Report on the National India. PLoS ONE, 3: 1–7.
Evaluation. Queanbeyan, Australia: Australian Diaz, T.P. (2002). Group work from an Asian Pacific Island
Government. perspective: Making connections between group
Brodaty, H. (1994). Dementia and the family. In S. Bloch, worker ethnicity and practice. Social Work with
Hafner, J., Harari, E. and Szmukler, G.I. (eds.), The Groups, 25: 43–60.
Family in Clinical Psychiatry. Oxford, UK: Oxford Donath, C., Grassel, E., Grossfeld-Schmitz, M. et al. (2010).
University Press, pp. 224–246. Effects of general practitioner training and family sup-
Brodaty, H., Roberts, K. and Peters, K. (1994). Quasi- port services on the care of home-dwelling dementia
experimental evaluation of an educational model for patients – Results of a controlled cluster-randomized
dementia caregivers. International Journal of Geriatric study. BMC Health Services Research, 10: 314.
Psychiatry, 9: 195–204. Ebenstein, H. (2004). The relationship between ­caregiver
Brodaty, H., Thomson, C., Thompson, C. and Fine, M. support groups and the market framework of caregiv-
(2005). Why caregivers of people with dementia and ing. Journal of Gerontological Social Work, 44: 127–149.
memory loss don’t use services. International Journal Eisdorfer, C., Czaja, S., Loewenstein, D. et al. (2003).
of Geriatric Psychiatry, 20: 537–546. The effect of a family therapy and technology-
Bruce, D.G., Paley, G.A., Underwood, P.J. et al. (2002). based ­intervention on caregiver depression. The
Communication problems between dementia carers Gerontologist, 43: 521–531.
and general practitioners: Effect on access to com- Fortinsky, R.H., Unson, C.G. and Garcia, R.I. (2002).
munity support services. Medical Journal of Australia, Helping family caregivers by linking primary care
177: 186–188. ­physicians with community-based dementia care
Callahan, C.M., Boustani, M.D., Unverzagt, F.W. et al. services. Dementia: The International Journal of Social
(2006). Effectiveness of collaborative care for older Research and Practice, 1: 227–240.
adults with Alzheimer disease in primary care: A Fung, W. and Chein, W. (2002). The effectiveness of a mutual
randomized controlled trial. Journal of the American support group for family caregivers of a relative with
Medical Association, 295: 2148–2157. dementia. Archives of Psychiatric Nursing, 16: 134–144.
Carpentier, N., Pomey, M.P., Contreras, R. and Olazabal, I. Gavrilova, S.I., Ferri, C.P., Mikhaylova, N. et al. (2008).
(2008). Social care interface in early-stage demen- Helping carers to care: The 10/66 Dementia Research
tia: Practitioners’ perspectives on the links between Group’s randomized control trial of a caregiver
formal and informal networks. Journal of Aging and ­intervention in Russia. International Journal of Geriatric
Health, 20: 710–738. Psychiatry, 24: 347–354.
Alzheimer’s associations and societies 369

Gillette-Guyonnet, S., Andrieu, S., Dantoine, T. et al. Mather, L. (2006). Memory Lane Café: Follow-up sup-
(2009). Commentary on “A roadmap for the ­prevention port for people with early stage dementia and their
of dementia II. Leon Thal Symposium 2008.” The families and carers. Dementia: The International
Multidomain Alzheimer Preventive Trial (MAPT): Journal of Social Research and Practice, 5: 290–293.
A new approach to the prevention of Alzheimer’s McClure, C. and Sanders, S. (2008). Familial caregivers
disease. Alzheimer’s & Dementia: The Journal of the of older adults. In Cummings, S. and Kropf, N. (eds.),
Alzheimer’s Association, 5: 114–121. Handbook of Psychosocial Interventions with Older
Gonyea, J.G. (1990). Alzheimer’s disease support group Adults: Evidence-Based Approaches. Indianapolis, IN:
participation and caregiver well-being. Clinical Haworth Press, pp. 293–320.
Gerontologist, 10: 17–34. Mittelman, M.S., Haley, W.E., Clay, O.J. and Roth, D.L.
Gonyea, J.G., Lopez, L.M. and Velasquez, E.H. (2014). The (2006). Improving caregiver well-being delays nursing
effectiveness of a culturally sensitive cognitive behav- home placement of patients with Alzheimer disease.
ioral group intervention for Latino Alzheimer’s caregiv- Neurology, 67: 1592–1599.
ers. The Gerontologist, 56 (2): 292–302. Molinari, V., Nelson, N., Shekelle, S. and Crothers, M.K.
Guerra, M., Ferri, C., Fonseca, M. et al. (2010). Helping (1994). Family support groups of the Alzheimer’s asso-
carers to care – The 10/66 Dementia Research Group’s cation: An analysis of attendees and non-attendees.
randomized control trial of a caregiver intervention in Journal of Applied Gerontology, 13: 86–98.
Peru. Revista Brasileira de Psiquiatria, 33 (1): 47–54. Moore, W. (2002). Lesbian and gay elders: Connecting
Henderson, J.N. (1992). The power of support: care providers through a telephone support group.
Alzheimer’s disease support groups for minority famil- Journal of Gay and Lesbian Social Services, 14: 23–41.
lies. Aging, 363 (364): 24–28. Napoles, A.M., Chadiha, L., Eversley, R. and Moreno-John,
Henderson, J.N., Gutierrez-Mayka, M., Garcia, J. and G. (2010). Reviews: Developing culturally sensitive
Boyd, S. (1993). A model for Alzheimer’s disease sup- dementia caregiver interventions: Are we there yet?
port group development in African-American and American Journal of Alzheimers Disease and Other
Hispanic populations. The Gerontologist, 33: 409–414. Dementias, 25: 389–406.
Javadpour, A., Ahmadzadeh, L. and Bahredar, M.J. Ngandu, T., Lehtisalo, J., Levälahti, E. et al. (2014).
(2008). An educative support group for female family Recruitment and baseline characteristics of partici-
caregivers: Impact on caregivers psychological pants in the finnish geriatric intervention study to
distress and patient’s neuropsychiatry symptoms. prevent cognitive impairment and disability (FINGER).
International Journal of Geriatric Psychiatry, International Journal of Environmental Research and
24: 469–471. Public Health, 11 (9): 9345–9360.
Kivipelto, M., Solomon, A., Ahtiluoto, S. et al. (2013). The O’Connell, M.E., Crossley, M., Cammer, A. et al. (2014).
Finnish geriatric intervention study to prevent cogni- Development and evaluation of a telehealth videocon-
tive impairment and disability (FINGER). Alzheimer’s & ferenced support group for rural spouses of individu-
Dementia: The Journal of the Alzheimer’s Association, als diagnosed with atypical early-onset dementias.
9: 657–665. Dementia (London), 13: 382–395.
Lanka Alzheimer’s Foundation (2009). Training for O’Connor, M.F., Arizmendi, B.J. and Kaszniak, A.W.
Carers. Available at http://www.alzlanka.org/index. (2014). Virtually supportive: A feasibility pilot study of
php?option=com_content&task=view&id=44&Ite an online support group for dementia caregivers in
mid=28. Accessed 15 January 2015. a 3D virtual environment. Journal of Aging Studies,
Leung, P., Orrell, M. and Orgeta, V. (2014). Social sup- 30: 87–93.
port group interventions in people with dementia and Parker, D., Mills, S. and Abbey, J. (2008). Effectiveness of
mild cognitive impairment: A systematic review of the interventions that assist caregivers to support people
literature. International Journal of Geriatric Psychiatry, with dementia living in the community: A system-
30 (1): 1–9. atic review. International Journal of Evidence-Based
Low, L.-F., Draper, B., Cheng, A. et al. (2009). Future Healthcare, 6: 137–172.
research on dementia relating to culturally and linguis- Pillemer, K. and Suitor, J. (2002). Peer support for
tically diverse communities. Australasian Journal on Alzheimer’s caregivers. Research on Aging,
Ageing, 28: 144–148. 24: 171–192.
Marriott, A., Donaldson, C., Tarrier, N. and Burns, ­A . Pinquart, M. and Sorensen, S. (2003). Differences between
(2000). Effectiveness of cognitive-behavioural ­family caregivers and noncaregivers in psychological health
intervention in reducing the burden of care in carers and physical health: A meta-analysis. Psychology and
of patients with Alzheimer’s disease. British Journal of Aging, 18: 250–267.
Psychiatry, 176: 557–62. Pinquart, M. and Sorensen, S. (2006). Helping caregiv-
Martindale-Adams, J., Nichols, L.O., Burns, R. et al. (2013). ers of persons with dementia: Which interventions
A trial of dementia caregiver telephone support. work and how large are their effects? International
Canadian Jounral of Nursing Research, 45: 30–48. Psychogeriatrics, 18: 1–19.
370 Dementia

Powell, J., Chiu, T. and Eysenbach, G. (2008). A systematic Sirpa, G., Tamminen, A., Eronen, A. et al. (2011). The
review of networked technologies supporting carers Memory Barometer 2010. Finland: Alzheimer Society
of people with dementia. Journal of Telemedicine and of Finland.
Telecare, 14: 154–156. Smith, K., Flicker, L., Shadforth, G. et al. (2011). Gotta be
Prince, M. (2004). 10/66 Dementia Research Group. Care sit down and worked out together: Views of Aboriginal
arrangements for people with dementia in developing caregivers and service providers on ways to improve
countries. International Journal of Geriatric Psychiatry, dementia care for Aboriginal Australians. Rural and
19: 170–177. Remote Health, 11: 1650.
Prince, M., Acosta, D., Chiu, H. et al. (2003). Dementia Steffen, A.M. and Mangum, K.R. (2012). Predicting atten-
diagnosis in developing countries: A cross-cultural dance at dementia family support groups. American
validation study. Lancet, 361: 909–917. Journal of Alzheimers Disease and Other Dementias,
Prince, M.J., de Rodriguez, J.L., Noriega, L. et al. 27: 633–639.
(2008). The 10/66 Dementia Research Group’s fully Topo, P. (2009). Technology studies to meet the needs of
operationalised DSM-IV dementia computerized people with dementia and their caregivers: A literature
diagnostic algorithm, compared with the 10/66 review. Journal of Applied Gerontology, 28: 5–37.
dementia algorithm and a clinician diagnosis: Toseland, R.W., Rossiter, C.M. and Labrecque, M.S.
A population validation study. BMC Public Health, (1989). The effectiveness of peer-led and profes-
8w: 219. sionally led groups to support family caregivers. The
Richard, E., Van den Heuvel, E., van Charante, E.P.M. et al. Gerontologist, 29: 465–471.
(2009). Prevention of dementia by intensive vascular United Nations (2011). Draft Resolution Submitted by the
care (PreDIVA): A cluster-randomized trial in progress. President of the General Assembly Political Declaration
Alzheimer Disease and Associated Disorders, 23 (3): of the High-Level Meeting of the General Assembly
198–204. on the Prevention and Control of Non-communicable
Selwood, A., Johnston, K., Katona, C. et al. (2007). A sys- Diseases. New York, NY: United Nations.
tematic review of the effect of psychological interven- United Nations (2013). World Population Prospects: The
tions on family caregivers of people with dementia. 2012 Revision. New York, NY: Department of Economic
Journal of Affective Disorders, 101: 75–89. and Social Affairs Population Division of the United
Senanarong, V., Jamjumras, P., Harmpahdungkit, K. et al. Nations.
(2004). A counselling intervention for caregivers: Effect Wituk, S.A., Shepherd, M.D., Warren, M. and Meissen, G.
on neuropsychiatric symptoms. International Journal of (2002). Factors contributing to the survival of self-help
Geriatric Psychiatry, 19: 781–788. groups. American Journal of Community Psychology,
Shaji, K.S., Smitha, K., Lal, K.P. and Prince, M.J. (2003). 30: 349–366.
Caregivers of people with Alzheimer’s disease: A Zarit, S.H., Femia, E.E., Watson, J. et al. (2004). Memory
qualitative study from the Indian 10/66 Dementia club: A group intervention for people with early stage
Research Network. International Journal of Geriatric dementia and their care partners. The Gerontologist,
Psychiatry, 18: 1–6. 44: 262–269.
37
Health economic aspects of dementia

ANDERS A. WIMO, LINUS JÖNSSON AND BENGT WINBLAD

categorized as direct costs (of resources used) and indirect


37.1 INTRODUCTION costs (of resources lost due to e.g. mortality, morbidity and
production losses). Direct costs are often presented in two
A highly prevalent and debilitating condition, such as demen- ways: direct medical costs (e.g. hospital care, clinic visits,
tia, presents a challenge for any healthcare and social sup- drugs etc.) and direct non-medical costs (e.g. social/home
port system. Care of people with dementia is extremely costly services, nursing home care). Informal care (see below) is
(Wimo et al., 2013a), and there is an important contribution often regarded as an indirect cost although this is not always
by informal carers (Wimo and Prince, 2010). In high-income obvious since many carers are retired and sometimes also
countries, it has been debated how healthcare and social sup- paid for parts of their care.
port systems will be able to manage the increasing number
of elderly people in general (The Organisation for Economic 37.2.2 PERSPECTIVE
Co-operation and Development [OECD], 2013), and people
with dementia in particular (Lovestone, 2002; Banerjee, 2012; An important purpose of economic evaluation is that it should
OECD, 2015). In low- and middle-income countries, the chal- serve as a tool for decision making regarding allocation of
lenge is rather to establish a care infrastructure for long-term scarce resources (Drummond et al., 2004). There are many
care (ADI, 2013). Although the global economic impact of different stakeholders involved in delivery and financing of
dementia is enormous, the scientific base in terms of health dementia care. Therefore it is essential to define the perspec-
economic studies in dementia care is comparatively small. The tive of any pharmacoeconomic analysis, i.e. from whose view-
first (and so far only) basic textbook in this field (Wimo et al., point costs and benefits are calculated. The viewpoint may be
1998a) was published in 1998 and reviewers have stressed the a county council, a municipality, the public sector in general,
need for further studies (Jönsson et al., 2000; Shukla et al., an insurance company, a family member or a patient. The
2000; Lamb and Goa, 2001; Morris, 2001; NICE, 2001; Gray, main cost components are costs of living (in its wide context,
2002; Grutzendler and Clegg et al., 2002; Lyseng-Williamson including also care in nursing homes) and informal caring
and Plosker, 2002; Wolfson et al., 2002; Birks and Harvey, (Wimo et al., 2003b; Schaller et al., 2015). There is a complex
2003; Jönsson, 2003a; Leung et al., 2003; Lyseng-Williamson interaction between those who finance the care and those
and Plosker, 2003; Olin and Schneider, 2003; SBU, 2008; who may benefit from the results of interventions. Therefore,
NICE, 2009; Jönsson and Wimo, 2009; Pouryamout et al., the detection of suboptimal incentives is an essential part of
2012; Costa et al., 2013; Knapp et al., 2013). the economic analysis. Limiting the economic analysis only
to the healthcare sector or the public sector excludes substan-
tial components. Thus, a societal perspective where all rele-
vant costs are identified and included regardless of where they
37.2 COSTING occur and of who pays is often to be preferred.

37.2.1 DIRECT AND INDIRECT COSTS 37.2.3 INFORMAL CARE


The opportunity cost is the value of a resource in its best Informal carers (mostly spouses or children) are of great
alternative use. This is the value that should theoretically importance in dementia care. An informal carer is often
be assigned to a resource within the context of a cost-of- part of a ‘dementia family’. In that sense, their situation
illness (COI) study or economic evaluation. Costs are often can be analysed in terms of burden, quality of life, coping,
371
372 Dementia

stress, social network and morbidity (Max et al., 1995; 37.2.4 GEOGRAPHIC TRANSFERABILITY
Jansson et al., 1998; Schulz and Beach, 1999; Wimo et al.,
1999b; Almberg et al., 2000; Etters et al., 2008; Raccichini The way the care is financed and organized as well as the
et al., 2009; Muangpaisan et al., 2010). relative supply of different forms of care, the general taxa-
They are also often providers of an extensive amount of tion level and the economic strength of countries are factors
unpaid informal care (Rice et al., 1993; Stommel et al., 1994; that make cost comparisons between countries difficult.
Langa et al., 2001; Moore et al., 2001; Wimo et al., 2002; Comparing currencies presents problems. Usual cur-
Nordberg et al., 2005; Knapp and Prince, 2007; Mesterton rency exchange rates reflect trade between countries rather
et al., 2010; Hurd et al., 2013; Wimo et al., 2013a, 2013b), than purchasing power. Purchase power parities (PPPs)
which constitute a great part of the societal costs. (OECD PPP, 2003), are probably better to use than exchange
Measuring caregiver time is problematic. Support in rates. Comparisons over time both within and between
personal activities of daily living (ADLs) and instrumen- countries are also linked to uncertainty. Consumer price
tal ADLs (IADLs) are well-defined activities. However, a indices are often used, but an index that reflects changes in
substantial part of care activities is linked to supervision the healthcare sector is better. The Statistical Office of the
to manage behavioural symptoms or to prevent dangerous European Communities (Eurostat) presents Harmonized
events (Wimo et al., 2002). The assessments of supervision Indices of Consumer Prices (HICPs) for different sectors,
and surveillance in terms of minutes and hours may be including healthcare.
difficult. In multinational studies, the results can be aggregated in
Two other important factors to consider are care pro- terms of resource use (and not in monetary terms) from every
ductivity and joint production. A formal and professional country. The cost calculations can then be presented in one
carer is probably more efficient and takes less time to sup- currency, based on aggregated resource use. However, if there
port ADL tasks than an informal carer. This must be con- are great differences in care organization between coun-
sidered if a replacement approach is used (see below). Joint tries, aggregation of resource utilization results may also be
production means that one activity can result in different problematic. For example, the concept of ‘nursing home’ can
outcomes, or the patient and the carer are doing things include a wide range of resources in terms of number of staff
together (e.g. shopping). and their competence, physical environment and technical
Costing informal care is also a complicated and contro- equipment, resulting in a great variation in costs. There are
versial issue (Koopmanschap, 1998; Jönsson et al., 2000; van also care concepts that are used just in one or few countries
den Berg et al., 2006). There are two frequently used meth- such as DOMUS care in the United Kingdom. (Beecham
ods, the opportunity cost approach and the replacement et al., 1993), Group Living in Sweden (Wimo et al., 1995) and
cost approach. Special Care Units (SCUs) in the United States. (Maas et al.,
Whether the carer is paid or not is not of interest for the 1998). Home care may denote social services with poorly
economic valuation. Payment has an impact on the distri- qualified staff, but also specific teams focused on demen-
bution of the economic burden, but not on the total societal tia. Even if a care organization can be divided roughly into
cost. The relevant cost is the opportunity cost and thus, the levels (such as nursing home care, intermediate care alter-
question is to identify the alternative use of the carer’s time. natives, home care), such a simplistic division questions the
If the alternative is working in the labour market, the cost validity of multinational intervention studies. Thus, another
for informal care should be valued to the production loss option could be to aggregate costs based on a classification
due to absence from work. of countries in terms of health and social care systems. Such
However, more problematic is the costing of leisure a classification could be based on how care is organized and
time and care time during retirement. There is no consen- financed, economic strength and major differences in care
sus on how this should be performed. The cost for the car- culture (such as the role of informal care).
er’s time may include the value of the time used as leisure
time (Karlsson et al., 1998). However, there is no such mar-
ket price available. Willingness-to-pay approaches – for
example the contingent valuation method (Johannesson 37.3 OUTCOME MEASURES
et al., 1992) are options to get prices and studies with these
approaches have been published in the field of demen- Cognition is considered as a mandatory primary efficacy
tia (Konig and Wettstein, 2002; Gustavsson et al., 2010; outcome in most dementia trials. For the patient, the cog-
Jutkowitz et al., 2010). nitive capacity is indeed a relevant outcome since it affects
With the replacement cost approach it is assumed that the intellectual capacity and thinking. However, other out-
informal carer should be replaced with a professional carer comes probably are more important for the informal carers
in a 1:1 ratio. The informal carer’s time and the professional such as ADL capacity, mood-depression and behavioural
carer’s time are regarded as perfect substitutes. However, we and psychological symptoms of dementia (BPSD). For both
do not know if there will actually be a complete replacement patients and carers, aspects of quality of life are significant
with a professional carer (in practice, most likely to a lower outcomes. For staff ADL capacity and BPSD, and in a wider
replacement level). context, care planning, are essential.
Health economic aspects of dementia 373

Surrogate end points are, in principle, regarded as inap- as ‘mobility’ are less problematic. In a project where proxy-
propriate to use in cost-effectiveness analysis (Johannesson rated and self-rated QoL were used simultaneously, great
et al., 1996).The Mini-Mental State Examination (MMSE) differences between the two approaches occurred (Jönsson,
(Folstein et al., 1975) is often used in dementia studies as 2003b; Jönsson et al., 2006a).
well as other measures of cognitive capacity such as the In principle, two kinds of QoL instruments can be used:
Alzheimer’s Disease Assessment Scale – cognitive subscale diagnosis-specific or generic instruments (Bowling, 1995;
(ADAS-cog) (Rosen et al., 1984). However, in our view they Spilker, 1996; Salek et al., 1998; Walker et al., 1998).
should be regarded as surrogate end points. Since the MMSE More or less dementia-specific instruments such as the
is highly correlated with costs (Ernst et al., 1997; Hux et al., dementia quality of life instrument (DQoL) (Brod et al.,
1998; Wimo et al., 1998c; Jönsson et al., 1999a), and also to 1999), Quality of Life in Alzheimer’s disease (QOL-AD)
other, more relevant outcomes, such as ADL capacity, posi- (Selai et al., 2001; Logsdon et al., 2002) and Quality of Life
tion in care organization and severity of dementia, it has Assessment Schedule (QOLAS) (Selai et al., 2000; Elstner
nevertheless frequently been used in economic evaluations et al., 2001) may be useful in trials with a cost consequence
of dementia. The MMSE is also used in most longitudinal design, but are inappropriate for complete economic
population-based studies and thus it may be useful as a link evaluations since these instruments not are preference
between clinical studies and population studies in the dis- based (Torrance et al., 1996; Siegel et al., 1997). However,
cussions of efficacy versus clinical effectiveness. However, Dementia–Quaity of life (DEMQOL) is a dementia-specific
when both MMSE and ADL are included in the same data instrument (Smith et al., 2007), where there also is a pref-
sets, ADL has a stronger correlation to resource use and erence-based version, Dementia–Quaity of life–Utilities
costs than cognition in terms of MMSE (Gustavsson et al., (DEMQOL-U) (Mulhern et al., 2013).
2011; Lindholm et al., 2013). Delay in progression in terms There is a great number of generic QoL scales, such as
of severity presents a broader outcome than cognition and the Sickness Impact Profile (SIP) (Bergner et al., 1981), the
severity scales such as the Clinical Dementia Rating (CDR) Short Form series (SF) (for example, SF-36 and the utility
(Berg, 1988) and the Global Deterioration Scale (GDS) instrument SF-6D) (Ware and Sherbourne, 1992; Walters
(Reisberg et al., 1988) or multidimensional instruments and Brazier, 2003), the QLA-scale (Blau, 1977), the Health
such as the Gottfries–Bråne–Steen scale (GBS) (Brane Utilities Index (HUI) (Torrance et al., 1996; Neumann
et al., 2001) are thus suggested as more relevant for use than et al., 2000), the EuroQoL/EQ-5D (Coucill et al., 2001),
cognition. 15-D (Sintonen, 2001) and the Quality of Well-Being Scale
Postponing or preventing nursing home entry has also (QWBS) (Kerner et al., 1998).
been suggested. A study by Knopman et al. (1996) indicated HUI, EQ-5D, QWBS, SF-6D and the caregiver quality of
that nursing home care can be postponed about 1 year with life index (CQLI) (Mohide et al., 1988) can be used to cal-
tacrine treatment. In a Swedish study (Wallin et al., 2004) of culate quality-adjusted life years (QALYs) (Torrance, 1997).
tacrine, the findings were similar. Sano et al. (1997) found QALYs are used in cost–utility analysis (CUA) and reflect
that Vitamin E/selegiline treatment postponed nursing both quantity and quality of life (Torrance, 1996; Torrance et
home placement. A study on donepezil indicated an even al., 1996) and give opportunities for comparisons with other
longer postponement of institutionalization (Geldmacher diseases. Utilities are expressed as a figure between 0 (death)
et al., 2003). However, the situation is more complex regard- and 1 (perfect health). Disease-related utilities have also
ing carers. It may be an advantage that spouses can go on been developed, e.g. in Alzheimer’s disease (AD) by Ekman
living together, but a prolonged period at home may be more et al. (2007) and DEMQOL-U as mentioned above (Mulhern
stressful for the informal carer (Max, 1996). This poten- et al., 2013). QALYs are not uncontroversial (Tsuchiya et al.,
tial risk was, however, not confirmed in the two published 2003). Chronic incurable progressive disorders may be mis-
randomized controlled trials (RCTs) where the amount of favoured when compared with surgical treatments, such as
informal care was measured (Wimo et al., 2003a, 2003b). cataract surgery or hip replacement surgery.
Quality of life (QoL) of both patients and carers is a rel- There are also other approaches, such as disability-
evant outcome. Health-related QoL includes several dimen- adjusted life years (DALYs) (Allotey et al., 2003) and healthy-
sions, such as ADLs, social interaction, perception, pain, years equivalents (HYE) (Dolan, 2000). However, DALYs
anxiety and economic status. One potential problem in QoL (often used by the World Health Organization [WHO]) focus
assessments is the patient’s difficulty to value his/her own on productivity and disability rather than QoL, and HYEs
health condition (Stewart and Brod, 1996). Thus, proxies require a great number of health scenarios (Torrance, 1996).
(mostly carers) are often used. Proxies are not necessarily a Burden scales, such as the Burden Interview (BI) (Zarit
problem. The U.S. panel for cost-effectiveness recommends et al., 1980) can to some extent be used as indicators of carer
that the general public should value health states and in that QoL, but the interplay between patient status, carer QoL
sense, an indirect measurement is not disqualified (Siegel and burden is complex (Deeken et al., 2003).
et al., 1997). However, if the proxy is a family member, the The choice of outcome measure in economic evaluation
answers may reflect, in part, the situation and interests of in dementia is not an obvious one. As different outcome
the proxy, which must be considered. Some attributes may measures give different pieces of information, several out-
be difficult for proxies, such as ‘pain’, whereas others, such come measures can be used.
374 Dementia

37.4.2 STUDY DESIGNS
37.4 STUDY DESIGN AND DATA
COLLECTION There are two basic types of health economic studies (Box
37.2): descriptive studies and normative studies (economic
37.4.1 MEASURING RESOURCE USE evaluations). Here we refer more or less to the classification
suggested by Drummond et al. (2004).
The costing process usually consists of two phases; first,
resource utilization is expressed in terms of physical units
37.4.2.1 Descriptive studies
(such as nursing home days) and second, the resource uti-
lization figures are calculated into costs by the use of unit Cost description (CD) describes only the costs (no outcome)
costs for each resource. Resource Utilization in Dementia of a single treatment/care, without making any comparison
(RUD) (Wimo et al., 1998b) is an example of a framework between alternative treatments. Thus, CD cannot give any
assessing the use of formal and informal resources, aiming support in priority discussions, but presents a framework
at calculating costs from a societal perspective (Box 37.1). for how costs are calculated.
RUD has been used in several studies (Wimo et al., 1999a; COI studies are also descriptive. The COI is equal to what
Wimo et al., 2000; Wimo et al., 2003a; Wimo et al., 2003b). these resources would have been used for if there had been
A comprehensive battery can be time-demanding and no case of illness (the opportunity cost). Two approaches can
thus, a short version, the RUD Lite (Wimo and Winblad, be used: an incidence approach or a prevalence approach.
2003b) was developed. It was possible to limit the number With the incidence approach the costs for new cases are
of variables in the RUD considerably without losing the estimated where both the annual costs and future (dis-
societal perspective in the analysis; RUD Lite covers about counted) costs are included. In the prevalence approach the
95% of the costs resulting from the complete RUD (Wimo costs for all cases during, e.g. a year, are estimated – both for
and Winblad, 2003b). One part of the RUD is the caregiver those who have dementia already and for new cases occur-
time subscale. The Client Service Receipt Inventory (CSRI) ring during the year in question (Lindgren, 1981; Rice et al.,
(Beecham and Knapp, 2001) and the Resource Use Inventory 1985). Only exceptionally have both approaches shown the
(RUI) (Sano et al., 2006) are other instruments aiming at same results. The choice of approach depends on the pur-
a broad assessment of resource use while the Caregiver pose of the study; if the aim is to obtain information for
Activities Time Survey (CATS) (Clipp and Moore, 1995) analysis of the economic consequences of interventions, the
and the Caregiver Activity Survey (CAS) (Davis et al., 1997) incidence approach is preferable. If the idea is to estimate
focus on caregiver time. the economic burden during a defined year, the prevalence
Since the organization of dementia care varies between approach is the best option.
countries, any resource use battery must be adapted to the Another issue is whether to use a top-down or a bottom-
specific situation in a country. up approach. In the ‘top-down’ approach, the total national
cost for a specific resource is distributed on different diseases.
Different types of registry data are often used in ‘top-down’
analyses. The ‘bottom-up’ method starts from a defined sub-
BOX 37.1: Components of a resource group with e.g. dementia, and registers all COI related to it,
utilization battery, Resource Utilization followed by an extrapolation to the total dementia popula-
in Dementia tion. In any COI study, it is problematic to compensate for

Patient Carer
Accomodation/ Informal care time
long-term care (for patient) BOX 37.2: Different kinds of health
(Work status) Work status economic studies
Respite care (Respite care)
Hospital care Hospital care Descriptive studies
Out clinic visits Out clinic visits CD Cost description
Social service Social service COI Cost of illness
Home nursing care Home nursing care Evaluation studies
Day care Day care CA Cost analysis (incomplete evaluation)
Drug use Drug use CMA Cost–minimization analysis
Source: Wimo, A. et al., Evaluation of the resource utilization CEA Cost-effectiveness analysis
and caregiver time in anti-dementia drug trials – A CBA Cost–benefit analysis
quantitative battery, In Wimo, A. et al. (eds.), The
CUA Cost–utility analysis
Health Economis of Dementia, London, United
Kingdom, John Wiley & Sons, 1998b. CCA Cost–consequence analysis
Health economic aspects of dementia 375

comorbidity and to identify the costs related to a single condi- In a cost–minimization analysis (CMA) the effects of
tion (such as dementia), ‘net costs’, may be difficult to estimate. different treatments are assumed to be equivalent and the
In a Swedish COI study (Wimo et al., 2014b) the proportion of analysis is focused on to identify the cheapest therapy.
net costs (costs assumed to only due to dementia) and gross The value of CMA is questioned, since the assumption
costs (costs including comorbidities) was about 75%. of similar treatment effects is problematic (Briggs and
It is also important to specify the included cost categories O’Brien, 2001).
(Is e.g. cost of informal care included?). It is obvious that In a cost-effectiveness analysis (CEA), the effect is
dementia care is costly, but the great range in cost figures in expressed as a non-monetary quantified unit. It may be the
Table 37.1 illustrates that the costing approaches may vary costs for ‘a saved year of living’ or in dementia ‘the cost per
in studies (Wimo, 2009). nursing home admission averted’. A cost–benefit analysis
In an estimate of the worldwide societal costs of demen- (CBA) expresses all costs and outcomes in monetary units.
tia, resulting in a total cost of $604 billion (US) in 2010, we Few attempts have been made to apply CBA to dementia.
included direct costs ($353 billion [US]) and costs of infor- It has, however, been argued that CBA approaches such as
mal care ($252 billion [US]) (Wimo et al., 2013a) (Table ‘willingness-to-pay’ could be of value (Wimo et al., 2002;
37.2), but the variability in the sensitivity analysis (Table Gustavsson et al., 2010). In a CUA, the effect is expressed as
37.3) illustrates the importance of transparency, otherwise utilities, such as QALYs (Torrance, 1997).
it is difficult to know whether presented cost differences In a cost–consequence analysis (CCA), cost and out-
depend on costing methods or true differences in costs. comes are listed and presented separately. The value of CCA
Issues that should be considered are for example, which cost is debatable (Winblad et al., 1997), since results may be
categories are included, whether the sample is population- difficult to interpret and the selection of outcomes may be
based or convenience samples. biased. However, in a situation where it is difficult to iden-
tify the most relevant and single outcome measure, it may
be of more interest to capture as much information as fea-
37.4.2.2 Normative studies sible (Mauskopf et al., 1998).
(economic evaluations)
37.4.3 ECONOMIC MODELS
Economic evaluations are aimed at being a support in deci-
sion making. In a cost analysis (CA), only the costs of dif- An intervention in dementia may influence the lifelong
ferent therapies are compared (not outcomes). Thus, a CA is course of the disease. Therefore, an economic evaluation
an incomplete economic evaluation. A complete economic should try to take into account impacts on costs and out-
evaluation includes the incremental costs ([C] and out- comes during the remaining whole lifespan. Most clinical
comes [E]) and comparisons between different treatment studies last for 6–18 months. Due to practical and ethical
alternatives. It is often expressed as the incremental cost- issues, single studies covering the whole disease period
effectiveness ratio (ICER): (C/(E, although net benefit con- will probably never be undertaken. One option to deter-
cepts also can be used. mine long-term effects is to extend ongoing studies. In

Table 37.1 Cost-of-illness studies expressed as $US (inflated to 2014)


Cost per person with
dementia Cost categories
Country/region $US 2014 included Source
World 20,549 D, IC Wimo et al. (2013a)
EU27 28,452 D, IC Wimo et al. (2008)
Europe 24,048 D, IC Gustavsson et al. (2013)
United States 45,413 D, IC Hurd et al. (2013)
Canada 31,952 D, IC Alzheimer Society (2010)
United Kingdom 51,236 D, IC Prince et al. (2014)
France 30,317 D, IC Rigaud et al. (2003)
Germany 75,572 D, IC community Schwarzkopf et al. (2011)
Ireland 56,089 D, IC Connolly et al. (2014)
Nordic 18,385 D, IC Jönsson et al. (2006b)
Italy 69,195 D, IC Cavallo and Fattore (1997)
Spain 40,304 D Atance Martinez et al. (2004)
Sweden 58,804 D, IC Wimo et al. (2014b)
Abbreviations: D, direct costs; IC, informal care.
376 Dementia

Table 37.2 Costs per person with dementia ($US) and aggregated costs (bns US$) by World Bank income
classification in 2010
Aggregated costs (US$ bn)
Number of
Per capita people with Total Informal care Direct Direct social
costs ($US) dementia costs (all ADLs) medical costs costs
Low income 868 5,036,979 4.37 2.52 1.23 0.62
Lower middle income 3,109 9,395,204 29.21 18.90 6.74 3.57
Upper middle income 6,827 4,759,025    32.49 13.70 10.44 8.35
High income 32,865 16,367,508 537.91 216.77 78.00 243.14
All 16,986 35,558,717 603.99 251.89 96.41 255.69
Source: Wimo, A. et al., Alzheimer’s Dementia, 9, 1–11, 2013a.
Abbreviation: ADL, activities of daily living.

Table 37.3 Sensitivity analysis of the global costs Wimo et al., 2014a). The basic idea is that, results from a
(bns US$) of dementia short core period are extrapolated to a longer period. Inputs
from several sources, e.g. efficacy, costing and progression,
Costs are used. Markov models are frequently used for this pur-
Base case 603.99 pose (Sonnenberg and Leventhal, 1998). Other approaches,
Basic ADLs only for 469.60 such as decision tree models (Weinstein and Fineberg,
informal care 1980), survival models (Fenn and Gray, 1999) and discrete
All ADL + supervision 783.29 events simulation (Caro, 2005) have been used. In a review,
for informal care 13 types of models regarding Alzheimer’s disease (AD)
Regression model 601.63 were identified (Cohen and Neumann, 2008). Discounting
of costs (and perhaps outcomes) is recommended. The dis-
Spouse caregiving 548.29
count rate is often 3%–5% (Siegel et al., 1997), but it can be
valued at 50% of
varied in a sensitivity analysis.
average wage
The use of models is often under discussion. The use
Spouse caregiving 520.44
of cognition as the vehicle for costs is questioned (Green,
valued at 25% of
2007). Long-term effects, such as exhaustion of carers and
average wage
staff, which do not occur in the core period and symptoms,
Replacement cost 616.71 such as BPSD, which do not have a linear progressive course
(average wage for a (Ferris and Mittelman, 1996) are difficult to model. A model
social care is also linked, implicitly or explicitly, to a specific organiza-
professional) tion of care. The assumptions in any model must be criti-
PPPs instead of 647.60 cally analysed.
exchange rates
Source: Wimo, A., et al., Alzheimer’s Dementia., 9, 1–11, 2013a.
Abbreviation: PPPs, purchase power parities.

37.4.4 EVALUATION OF
RCTs it is difficult to maintain the original design for many PHARMACEUTICALS
years, but long-term follow-up studies have been used for
cholinesterase inhibitors (ChEIs) (Knopman et al., 1996; Even if it would be advantageous to design studies purely
Geldmacher et al., 2003; Wallin et al., 2004; Gustavsson for health economic purposes, pharmacoeconomics will
et al., 2012). However, there are several drawbacks, such probably be part of comprehensive phase III/IV studies
as selection bias, patients lost to follow-up and problems (Hill and McGettigan, 1998). It is, however, important
defining controls. Another interesting option is to analyse to refine the methods used (Briggs and O’Brien, 2001).
registry data and to merge databases, e.g. record linkage of It has also been debated whether pharmacoeconomic
national care registers with quality-of-care registers and aspects are best included in phase III or phase IV studies
population-based studies to analyse long-term effects in (Schneider, 1998).
real life with multilevel statistical methods. However, vari- Several pharmacoeconomic evaluations have been pub-
ous modelling approaches are the most common ways of lished, mainly on the ChEIs (some on memantine) with var-
trying to analyse the long-term effects (Buxton et al., 1997; ious approaches. The early tacrine studies focused on costs
Health economic aspects of dementia 377

and they are also based on the same efficacy study (Knapp outcomes that demonstrate efficacy on both a cognitive and
et al., 1994). Even if treatment seems to be cost saving, the a functional or global assessment scale (FDA, 2013).
range is large in these studies, from about 1%–17%, indicat- Alzheimer Europe (AE) have repeatedly updated informa-
ing different methodological approaches but also problems. tion about access and reimbursement and still there is a vari-
Few studies are empirical (Table 37.4) and most evaluations ability in reimbursement in Europe regarding the ChEIs and
are based on models (Table 37.5). The empirical studies are memantine (Alzheimer Europe, 2006, Alzheimer Europe,
short term studies with weak statistical power for the cost 2012) and the situation is even more pronounced from a world-
results. While most previous pharmacoeconomic models wide viewpoint (Suh et al., 2009). The U.K. controversy regard-
have focused of ChEIs and memantine, new but so far not ing the recommendations from the National Institute for
proven disease-modifying treatment (DMT) options have Health and Care Excellence (NICE) for drug treatment in AD
been of interest for modelling. As seen in Table 37.5, sev- highlights how delicate this issue is (NICE, 2009). The critical
eral modelling methods have been used. In contrast to the question is the reimbursement decisions (Drummond, 2003).
empirical studies, most models indicate cost-effectiveness, For example, in Sweden, where the Dental and Pharmaceutical
either in terms of absolute dominance (<0) or versus a will- Benefits Agency, TLV, a central government agency, decides
ingness-to-pay for a gained QALY in the ICER (which often whether a pharmaceutical product or dental care procedure
is US$50,000 or more). should be reimbursed, the ChEIs are reimbursed like other
Drug trials are often criticized regarding representative- drugs. The experiences from Australia and Canada with drug
ness. Schneider (Schneider et al., 1997) showed that study authorities demanding health economic evaluations as part of
populations in dementia drug trials only reflect about 7% of the reimbursement decision process illustrate that this issue
a general dementia population. The progression rate of cog- is indeed complicated (Hill and McGettigan, 1998). The for-
nition is slower in placebo groups in most clinical studies mer Canadian Health Technology Assessment Guidelines
than in population-based studies. Another issue is whether for Pharmacoeconomics (CCOHTA) (now Canadian Agency
study populations reflect the dementia care organization to for Drugs and Technologies in Health [CADTH]) focused on
which the results are generalized. meta-analysis and epidemiological data and emphasized the
New drugs need to meet requirements by several dif- use of CBA and CUA (CCOHTA, 1997).
ferent regulatory bodies before being made available to
patients. While the marketing authorization process in the
European Union is harmonized and centralized through 37.4.5 EVALUATION OF CARE
the European Medicines Agency (EMA) (guidelines cur- PROGRAMMES
rently under revision) (EMA, 2014), decisions about pric-
ing and reimbursement are made on the country, regional The term ‘non-pharmacological treatment’ is commonly
and local levels. In the United States, specific guidelines for used to describe care interventions such as day care, carer
the treatment of the early stage AD have been presented by support, living arrangements and case management. We
the Food and Drug Administration (FDA), which focus on think this term is inappropriate since in most cases it

Table 37.4 Randomized controlled trials with empirical pharmacoeconomic data (costs inflated to 2014)
Results Sensitivity
Drug Country Duration US$ 2005a analysis Source
Donepezil Nordic 1 year 1426 (ns) 654–1549 Wimo et al. (2003a)
Donepezil Canada, France, 6 months 365 (ns) 85–582 Feldman et al.
Australia (2004)
Donepezil United Kingdom 60 weeks −1137 (ns) Yes, but results Courtney et al.
not presented (2004)
Memantine United States 6 months 9364 (p < 0.05) Yes, but results Wimo et al. (2003b)
not presented
(per month)
228 (r), −11 (o),
−235 (q)
Risperidone (r), United States 9 months Overall p = 0.02 Different WTP Rosenheck et al.
olanzapine (o) and (placebo lower) levels (2007)
quetiapine (q)

Notes: ns, non-significant; WTP, willingness-to-pay.


a A positive value indicates net savings with treatment.
378 Dementia
Table 37.5 Cost-effectiveness models (costs are inflated as $US 2014)
Model Range in sensitivity
length Model ICER or analysis or similar of
Drug (Years) type Country Outcome Cost diff.a similarb ICER Source
Donepezil 5 Markov United Kingdom Severity −3,152 12,671 2,094–21,991 Stewart et al. (1998)
Donepezil 5 Markov Canada Severity 1,067 <0 <0–10,435 O’Brien et al. (1999)
Donepezil 5 Markov Sweden Severity 507 <0 <0–4,026 Jönsson et al. (1999b)
Donepezil 2 Markov United States QALYs 108 <0 <0–624,295 Neumann et al. (1999)
Donepezil 2 Markov Japan QALYs 303 <0 <0–52,913 Ikeda et al. (2002)
Rivastigmine 2 Survival Canada QALYs 522 <0 Threshold analysis Hauber et al. (2000)
different WTPs
Galantamine 10 AHEAD Canada QALYs 927 <0 <0–50,791 Getsios et al. (2001)
Galantamine 10.5 AHEAD Sweden NNT 3,823 <0 NA Garfield et al. (2002)
FTCc
Galantamine 10.5 AHEAD United States NNT FTC 4,988 <0 <0–18,539 Migliaccio-Walle
et al. (2003)
Galantamine 10 AHEAD The Netherlands QALYs 2,206 <0 <0–6,412 Caro et al. (2002)
Galantamine 10 AHEAD United Kingdom QALYs −972 17,547 11,727–35,184 Ward et al. (2003)
Memantine 2 Markov United Kingdom QALYs 3,882 <0 <0–291,670 Jones et al. (2004)
Memantine 5 Markov Finland Depend 2,061 <0 <0 in 94% of Monte François et al. (2004)
Carlo simulations
Memantine 5 Markov Canada QALYs 16,126 <0 <0 all scenarios Jönsson (2005)
Donepezil 10 DESd United Kingdom QALYs 9,597 <0 <0 in all scenarios Getsios et al. (2010)
Donepezil 2 Markov Japan QALYs −3,320 −5,428 −3,320 to −11,253 Kasuya and Meguro (2010)

DMTe 10 DES UK QALYs 77,520 <0 <0 in all scenarios Guo et al. (2014)
DMT 20 Markov Sweden QALYs −35,898 −43,998 −793 to −314,027 Skoldunger et al. (2013)

Abbreviations: Cost diff., cost difference; ICER, incremental cost-effectiveness ratio; QALYs, quality-adjusted life years; AHEAD, Assessment of Health Economics in Alzheimer’s
disease; NA, not available.
a A positive value indicates net savings with treatment with ChEIs.

b A positive value indicates a cost per gained QALY/avoided deterioration in severity; <0 cost savings and a positive outcome.

c Number needed to treat to avoid full-time care.

d Discrete events simulation.

e Disease-modifying treatment.
Health economic aspects of dementia 379

reflects basic care for people with dementia and therefore 37.5.2 MILD COGNITIVE IMPAIRMENT
we use the term ‘programmes’. We also regard multidomain (MCI)
packages including lifestyle changes and basic drug treat-
ment as programmes. The economic literature in this field is With the hopes for DMT and prevention of dementia, there
sparse and heterogenous, but some studies have been pub- is now an interest to include interventions that start in MCI
lished (Table 37.6), both with empirical data and with mod- (or similar concepts) in economic evaluations. Such evalu-
elling approaches. The problem with the empirical studies, ations also include cost-effectiveness discussions regarding
besides their heterogeneity, is that study samples are often diagnostic methods, such as biomarkers. Health economic
small, resulting in weak statistical power and wide confi- evaluations of MCI can be divided into short-term and
dence intervals. Bootstrapping methods can partly com- long-term effects (Wimo and Winblad, 2003a). In the short
pensate for that. term, an intervention can perhaps result in a reduction of
informal care in terms of IADL. Thus it is questionable
whether the costs of an intervention will be offset from a
37.4.6 HANDLING UNCERTAINTY short-term viewpoint. In the long run, the question will be
whether survival is affected and whether there will be any
In any economic analysis, it is important to include a
transitions of symptoms and resource use during the course
discussion of uncertainty (Briggs and O’Brien, 2001)
of the dementia. To design studies to answer these questions
and to test the robustness of the analysis in a sensitiv-
is not easy. A synthetic approach with RCTs covering seg-
ity analysis. Roughly, there are two major approaches for
ments of the dementia course, modelling studies and open
sensitivity analyses: deterministic and probabilistic sen-
follow-up studies probably will be needed, resulting in some
sitivity analysis. With the deterministic approach (such
kind of ‘best guess’ judgement. Modelling seems to be the
as one-way or two-way/multiple-way sensitivity analysis),
most appropriate method and some of the modelling stud-
one or perhaps two inputs are varied at each occasion.
ies in Tables 37.6 and 37.7 starts in MCI. One model with
In a probabilistic sensitivity analysis, several inputs’ sta-
a hypothesized DMT starting in MCI showed no cost sav-
tistical variability (such as standard deviations) is var-
ings but with a standard willingness-to-pay level, the DMT
ied randomly and simultaneously with several iterations.
could be regarded as cost-effective (Skoldunger et al., 2013).
Examples of inputs that are varied in a sensitivity analy-
sis are unit costs for different items and discount rates.
In dementia, one of the most important cost drivers is 37.5.3 SEVERE DEMENTIA
the cost of informal care. Both the quantities (What kind
of informal care activities are included?) and the costing Several years in the course of dementia are spent in the stage
approach may be of interest to vary. The range in costs of of severe dementia (Winblad et al., 1999). In a model of a
informal care can vary a lot depending on how it is quan- Swedish cohort, the course of dementia with an estimated
tified and price fixed. survival up to 9 years was simulated (Wimo et al., 1998c).
As shown in Table 37.2, it is obvious that dementia care About 75% of the total costs occurred in severe dementia,
is very costly globally, but the great range in cost figures indicating that the maximal span for benefits during the
from the sensitivity analysis in Table 37.3 illustrates that total course in terms of cost reduction is only 25%, if efficacy
depending on assumptions, the resulting costs may vary is shown only for mild-to-moderate dementia. The results
considerably. indicate that severe dementia must be the focus for improved
care and research. Many of the instruments we use for out-
come research show floor effects in severe dementia and
they are not sufficiently sensitive to detect clinically relevant
changes (Winblad et al., 1999). There are studies that have
37.5 EVALUATION BY DISEASE shown positive effects in severe dementia for memantine
SEVERITY (Reisberg et al., 2003), donepezil (Winblad et al., 2006) and
to some extent galantamine (Burns et al., 2009), but health
37.5.1 COSTS AND DISEASE SEVERITY economic studies for severe dementia are rare (one example
is a study on memantine [Wimo et al., 2003b]).
There is a strong relationship between costs of dementia care
and the severity of dementia. In a population-based Swedish
COI study with a bottom-up approach (Wimo et al., 2009),
where costs were expressed in terms of the CDR, the cost
37.6 CONCLUSIONS
ratio between severe dementia and no decline was 6.5 (Table
37.7). There is also a strong association between costs and The health economics of dementia is still in a premature state,
functional decline (Quentin et al., 2009), and ADL capac- even if the number of published studies is increasing. There is
ity is also a stronger predictor of costs than cognition a great need for methodological improvement. In view of the
(Gustavsson et al., 2011; Lindholm et al., 2013). increasing number of people with dementia and existing as
380 Dementia
Table 37.6 Economic evaluations of programmes (costs are inflated as $US 2014)
Sensitivity
ICER in base analysis or
Programme Design Country Duration Outcome Results option similar Source
Day Care Prospective Sweden 6 months WY −4,018 11,941/WY NA Wimo et al. (1990)
non-RCT
Day Care Prospective Sweden 1 year QALYs 5,865 Neutral NA Wimo et al. (1994)
non-RCT
Caregiver Prospective RCT Canada 6 months QALYs 1,021 30,799/QALY NA Drummond et al. (1991)
support
Group Living Markov model Sweden 8 years QALYs 12,763 Dominance <0 in all Wimo et al. (1995)
scenarios
Caregiver CBA model Switzerland ? Monetary Dominance <0 in all Nocera et al. (2002)
support scenarios
Caregiver Markov model Finland 5 years QALYs 3,834 Dominance 71% of Monte Martikainen et al. (2004)
support Carlo
simulations
cost saving
and QALY
gaining
Case RCT + The Netherlands 6 months ‘Successful 294 5,167/unit 95% vs. WTP of Melis et al. (2008)
management bootstrapping treatment’ 37,800
Case RCT + The Netherlands 3 months ‘Successful 2,691 Dominance(94% 99% vs. WTP of Graff et al. (2008)
management bootstrapping treatment’ probability) 2,200 per
‘successful
treatment’
Case RCT + The Netherlands 1 year QALYs 100 2,129/QALY 72% vs. WTP of Wolfs et al. (2007)
management bootstrapping 50,000
Caregiver Prospective Sweden 5 years QALYs 90/month NA NA Dahlrup et al. (2013)
support non-RCT
Caregiver RCT + The Netherlands 1 years QALYs −6,326 240,164 <0 to 2,001,862 Joling et al. (2013)
support bootstrapping
Diagnostic + DES model United States Lifetime QALYs 12,029 Dominance <0 in all Guo et al. (2012)
donepezil scenarios
Multidomain Markov model Sweden/Finland 20 years QALYs 3,300 Dominance <0 to 64,105 Zhang et al. (2011)
package

Notes: CBA, cost–benefit analysis; RCT, randomized controlled trials; WY, well years.
Health economic aspects of dementia 381

Table 37.7 Societal costs of dementia in Sweden in relation Beecham, J. and Knapp, M. (2001). Costing psychiat-
to Clinical Dementia Rating (CDR) (costs are inflated as ric interventions. In Thornicroft, G. (ed.), Measuring
$US 2014) Mental Health Needs. London, United Kingdom:
Gaskell, pp. 200–224.
Cognitive
Berg, L. (1988). Clinical Dementia Rating (CDR).
CDR decline Gross costs Net costs
Psychopharmacology Bulletin, 24: 637–639.
0 None 8,000 – Bergner, M., Bobbitt, R.A., Carter, W.B. and Gilson, B.S.
0.5 Light 22,000 – (1981). The Sickness Impact Profile: Development and
1 Mild 31,400 23,400 final revision of a health status measure. Medical Care,
2 Moderate 43,000 35,000 19: 787–805.
3 Severe 51,900 43,900 Birks, J.S. and Harvey, R. (2003). Donepezil for dementia
due to Alzheimer’s disease (Cochrane review). Oxford,
Wimo, A. et al., Läkartidningen, 106, 1277–1282,
Source:  United Kingdom: Software update, Cochrane Library.
2009.
Blau, T.H. (1977). Quality of life, social indicators and crite-
ria of change. Professional Psychology, 8: 464–473.
Bowling, A. (1995). Measuring Health. A Review of Quality
well as new drugs in the pipeline, there is also great interest in of Life Measurement Scales. Milton Keynes, England:
pharmacoeconomics among the drug authorities in different Open University Press.
countries and in the pharmaceutical companies engaged in Brane, G., Gottfries, C.G. and Winblad, B. (2001). The
drug development. The sparse economic literature on differ- Gottfries-Brane-Steen scale: Validity, reliability and
ent care programmes and prevention is also a great challenge. application in anti-dementia drug trials. Dementia and
Geriatric Cognitive Disorders, 12: 1–14.
Briggs, A.H. and O’Brien, B.J. (2001). The death of cost-
REFERENCES minimization analysis? Health Economics, 10: 179–84.
Brod, M., Stewart, A.L., Sands, L. and Walton, P. (1999).
ADI (2013). World Alzheimer Report 2013. Journey of Conceptualization and measurement of quality of life
Caring. An Analysis of Long Term Care for Dementia. in dementia: The dementia quality of life instrument
London, United Kingdom: Alzheimer’s Disease (DQoL). Gerontologist, 39: 25–35.
International. Burns, A., Bernabei, R., Bullock, R. et al. (2009). Safety and
Allotey, P., Reidpath, D., Kouame, A. and Cummins, R. efficacy of galantamine (Reminyl) in severe Alzheimer’s
(2003). The DALY, context and the determinants of disease (the SERAD study): A randomised, placebo-
the severity of disease: An exploratory comparison of controlled, double-blind trial. Lancet Neurology, 8:
paraplegia in Australia and Cameroon. Social Science 39–47.
and Medicine, 57: 949–958. Buxton, M.J., Drummond, M.F., Van Hout, B.A. et al.
Almberg, B., Grafstrom, M., Krichbaum, K. and Winblad, (1997). Modelling in economic evaluation: An unavoid-
B. (2000). The interplay of institution and family able fact of life. Health Economics, 6: 217–227.
caregiving: Relations between patient hassles, nursing Caro, J.J. (2005). Pharmacoeconomic analyses using
home hassles and caregivers’ burnout. International discrete event simulation. Pharmacoeconomics, 23:
Journal of Geriatric Psychiatry, 15: 931–939. 323–332.
Alzheimer Europe (2006). Dementia in Europe Yearbook Caro, J.J., Salas, M., Ward, A. et al. (2002). Economic
2006. Luxembourg, Europe: Alzheimer Europe. analysis of galantamine, a cholinesterase inhibitor,
Alzheimer Europe (2012). Dementia in Europe Yearbook in the treatment of patients with mild to moderate
2012 – National Dementia Strategies (Diagnosis, Alzheimer’s disease in the Netherlands. Dementia and
Treatment and Research). Luxembourg, Europe: Geriatric Cognitive Disorders, 14: 84–89.
Alzheimer Europe. Cavallo, M.C. and Fattore, G. (1997). The economic
Alzheimer Society (2010). Rising Tide: The Impact and social burden of Alzheimer disease on families
of Dementia on Canadian Society. Toronto, ON: in the Lombardy region of Italy. Alzheimer Disease
Alzheimer Society. Associated Disorders, 11: 184–190.
Atance Martinez, J.C., Yusta Izquierdo, A. and Grupeli CCOHTA (1997). Guidelines for Economic Evaluation of
Gardel, B.E. (2004). [Costs study in Alzheimer’s dis- Pharmaceuticals: Canada, Second Edition. Ottawa,
ease]. Revista Clinica Espanola, 204: 64–69. Canada: The Canadian Coordinating Office for Health
Banerjee, S. (2012). The macroeconomics of dementia Technology Assessment.
– Will the world economy get Alzheimer’s disease? Clegg, A., Bryant, J., Nicholson, T. et al. (2002). Clinical
Archives of Medical Research, 43: 705–709. and cost-effectiveness of donepezil, rivastigmine,
Beecham, J., Cambridge, P., Hallam, A. and Knapp, M. and galantamine for Alzheimer’s disease. A sys-
(1993). The cost of domus care. International Journal of tematic review. International Journal of Technology
Geriatric Psychiatry, 8: 827–831. Assessment in Health Care, 18: 497–507.
382 Dementia

Clipp, E.C. and Moore, M.J. (1995). Caregiver time Elstner, K., Selai, C.E., Trimble, M.R. and Robertson, M.M.
use: An outcome measure in clinical trial research (2001). Quality of life (QOL) of patients with Gilles de
on Alzheimer’s disease. Clinical Pharmacology and la Tourette’s syndrome. Acta Psychiatrica Scandinavica,
Therapeutics, 58: 228–236. 103: 52–59.
Cohen, J.T. and Neumann, P.J. (2008). Decision analytic EMA (2014). Discussion Paper on the Clinical Investigation
models for Alzheimer’s disease: State of the art and of Medicines for the Treatment of Alzheimer’s Disease
future directions. Alzheimer’s & Dementia: The Journal and Other Dementias. London, United Kingdom: EMA
of the Alzheimer’s Association, 4: 212–222. (European Medicines Agency).
Connolly, S., Gillespie, P., O’shea, E. et al. (2014). Ernst, R.L., Hay, J.W., Fenn, C. et al. (1997). Cognitive
Estimating the economic and social costs of dementia function and the costs of Alzheimer disease. An
in Ireland. Dementia (London), 13: 5–22. exploratory study. Archives of Neurology, 54: 687–693.
Costa, N., Ferlicoq, L., Derumeaux-Burel, H. et al. (2013). Etters, L., Goodall, D. and Harrison, B.E. (2008). Caregiver
Comparison of informal care time and costs in differ- burden among dementia patient caregivers: A review
ent age-related dementias: A review. Biomed Research of the literature. Journal of the American Academy of
International, 2013: 852368. Nurse Practitioners, 20: 423–428.
Coucill, W., Bryan, S., Bentham, P. et al. (2001). EQ-5D in FDA (2013). Guidance for Industry. Alzheimer’s Disease:
patients with dementia: An investigation of inter-rater Developing Drugs for the Treatment of Early
agreement. Medical Care, 39: 760–771. Stage Disease. Silver Spring, MD: Food and Drug
Courtney, C., Farrell, D., Gray, R. (2004). Long-term Administration (FDA).
donepezil treatment in 565 patients with Alzheimer’s Feldman, H., Gauthier, S., Hecker, J. et al. (2004).
disease (AD2000): Randomised double-blind trial. Economic evaluation of donepezil in moderate to
Lancet, 363: 2105–2115. severe Alzheimer disease. Neurology, 63: 644–650.
Dahlrup, B., Nordell, E., Steen Carlsson, K. and Elmstahl, Fenn, P. and Gray, A. (1999). Estimating long-term cost
S. (2013). Health economic analysis on a psychosocial savings from treatment of Alzheimer’s disease. A mod-
intervention for family caregivers of persons with elling approach. Pharmacoeconomics, 16: 165–174.
dementia. Dementia and Geriatric Cognitive Disorders, Ferris, S.H. and Mittelman, M.S. (1996). Behavioral
37: 181–195. treatment of Alzheimer’s disease. International
Davis, K.L., Marin, D.B., Kane, R. et al. (1997). The Psychogeriatrics, 8: 87–90.
Caregiver Activity Survey (CAS): Development and Folstein, M.F., Folstein, S.E. and McHugh, P.R. (1975).
validation of a new measure for caregivers of persons Mini-mental state. A practical method for grading the
with Alzheimer’s disease. International Journal of cognitive state of patients for the clinician. Journal of
Geriatric Psychiatry, 12: 978–988. Psychiatric Research, 12: 189–198.
Deeken, J.F., Taylor, K.L., Mangan, P. et al. (2003). François, C., Sintonen, H., Sulkava, R. and Rive, B. (2004).
Care for the caregivers: A review of self-report Cost effectiveness of memantine on moderately
­instruments developed to measure the burden, severe to severe Alzheimer’s disease. A Markov model
needs, and quality of life of informal caregivers. in Finland. Clinical Drug Investigation, 24: 373–384.
Journal of Pain and Symptom Management, 26: Garfield, F.B., Getsios, D., Caro, J.J. et al. (2002).
922–953. Assessment of health economics in Alzheimer’s dis-
Dolan, P. (2000). A note on QALYs versus HYEs. Health ease (AHEAD). Treatment with galantamine in Sweden.
states versus health profiles. International Journal Pharmacoeconomics, 20: 629–637.
of Technology Assessment in Health Care, 16: Geldmacher, D.S., Provenzano, G., McRae, T. et al. (2003).
1220–1224. Donepezil is associated with delayed nursing home
Drummond, M.F. (2003). The use of health economic infor- placement in patients with Alzheimer’s disease.
mation by reimbursement authorities. Rheumatology Journal of the American Geriatric Society, 51: 937–944.
(Oxford), 42 (Suppl. 3): S60–S63. Getsios, D., Blume, S., Ishak, J. and Maclaine, G.D.H.
Drummond, M.F., Mohide, E.A., Tew, M. et al. (1991). (2010). Cost effectiveness of donepezil in the treat-
Economic evaluation of a support program for care- ment of mild to moderate Alzheimer’s disease:
givers of demented elderly. International Journal of A UK evaluation using discrete-event simulation.
Technology Assessment in Health Care, 7: 209–219. Pharmacoeconomics, 28: 411–427.
Drummond, M.F., Sculper, M.J., Torrance, G.W. et al. Getsios, D., Caro, J.J., Caro, G. and Ishak, K. (2001).
(2004). Methods for the Economic Evaluation of Health Assessment of health economics in Alzheimer’s
Care Programmes. Oxford, United Kingdom: Oxford disease (AHEAD): Galantamine treatment in Canada.
University Press. Neurology, 57: 972–978.
Ekman, M., Berg, J., Wimo, A. et al. (2007). Health utilities Graff, M.J., Adang, E.M., Vernooij-Dassen, M.J. et al.
in mild cognitive impairment and dementia: A popula- (2008). Community occupational therapy for older
tion study in Sweden. International Journal of Geriatric patients with dementia and their care givers: Cost
Psychiatry, 22: 649–655. effectiveness study. BMJ, 336: 134–138.
Health economic aspects of dementia 383

Gray, A. (2002). Health economics. In: Qizilbash, N. et al. of people with dementia. International Journal of
(eds.), Evidence-Based Dementia Practice. Oxford, Geriatric Psychiatry, 13: 674–681.
United Kingdom: Blackwell publishing. Johannesson, M., Johansson, P.O. and Jönsson, B. (1992).
Green, C. (2007). Modelling disease progression Economic evaluation of drug therapy: A review of the
in Alzheimer’s disease: A review of modelling contingent valuation method. Pharmacoeconomics, 1:
methods used for cost-effectiveness analysis. 325–337.
Pharmacoeconomics, 25: 735–750. Johannesson, M., Jönsson, B. and Karlsson, G. (1996).
Grutzendler, J. and Morris, J.C. (2001). Cholinesterase Outcome measurement in economic evaluation. Health
inhibitors for Alzheimer’s disease. Drugs, 61: 41–52. Economics, 5: 279–296.
Guo, S., Getsios, D., Hernandez, L. et al. (2012). Joling, K.J., Bosmans, J.E., Van Marwijk, H.W. et al. (2013).
Florbetaben PET in the early diagnosis of Alzheimer’s The cost-effectiveness of a family meetings interven-
disease: A discrete event simulation to explore its tion to prevent depression and anxiety in family care-
potential value and key data gaps. International givers of patients with dementia: A randomized trial.
Journal of Alzheimer’s Disease, 2012: 548157. Trials, 14: 305.
Guo, S., Getsios, D., Revankar, N. et al. (2014). Evaluating Jones, R.W., McCrone, P. and Guilhaume, C. (2004). Cost
disease-modifying agents: A simulation framework for effectiveness of memantine in Alzheimer’s disease: An
Alzheimer’s disease. Pharmacoeconomics, 32: 1129–1139. analysis based on a probabilistic Markov model from a
Gustavsson, A., Brinck, P., Bergvall, N. et al. (2011). UK perspective. Drugs & Aging, 21: 607–620.
Predictors of costs of care in Alzheimer’s disease: A Jönsson, B., Jönsson, L. and Wimo, A. (2000). Cost
multinational sample of 1222 patients. Alzheimer’s & of dementia. In May, M. and Sartorius, N. (eds.),
Dementia: The Journal of the Alzheimer’s Association, Dementia. WPA Series Evidence and experience in
7: 318–327. Psychiatry. London, United Kingdom: John Wiley &
Gustavsson, A., Jönsson, L., McShane, R. et al. (2010). Sons.
Willingness-to-pay for reductions in care need: Jönsson, L. (2003a). Economic Evaluation of Treatments
Estimating the value of informal care in Alzheimer’s for Alzheimer’s Disease. Neurotec. Stockholm,
disease. International Journal of Geriatric Psychiatry, Sweden: Karolinska Institutet.
25: 622–632. Jönsson, L. (2003b). Pharmacoeconomics of cholineester-
Gustavsson, A., Jönsson, L., Parmler, J. et al. (2012). ase inhibitors in the treatment of Alzheimer’s disease.
Disease progression and costs of care in Alzheimer’s Pharmacoeconomics, 21: 1025–1037.
disease patients treated with donepezil: A longitudinal Jönsson, L. (2005). Cost-effectiveness of memantine for
naturalistic cohort. The European Journal of Health moderate to severe Alzheimer’s disease in Sweden.
Economics, 13: 561–568. American Journal of Geriatric Pharmacotherapy, 3:
Gustavsson, A., Svensson, M., Jacobi, F. et al. (2013). Cost 77–86.
of disorders of the brain in Europe 2010. European Jönsson, L., Andreasen, N., Kilander, L. et al. (2006a).
Neuropsychopharmacology, 21: 718–779. Patient- and proxy-reported utility in Alzheimer
Hauber, A.B., Gnanasakthy, A. and Mauskopf, J.A. disease using the EuroQoL. Alzheimer Disease and
(2000). Savings in the cost of caring for patients with Associated Disorders, 20: 49–55.
Alzheimer’s disease in Canada: An analysis of treat- Jönsson, L., Eriksdotter Jonhagen, M., Kilander, L. et al.
ment with rivastigmine. Clinical Therapeutics, 22: (2006b). Determinants of costs of care for patients
439–451. with Alzheimer’s disease. International Journal of
Hill, S. and McGettigan, P. (1998). Drug authorities’ policy Geriatric Psychiatry, 21: 449–459.
on the assessment of drugs for dementia. In Wimo, Jönsson, L., Lindgren, P., Wimo, A. et al. (1999a). Costs
A., Karlsson, G., Jönsson, B. and Winblad, B. (eds.), of mini mental state examination-related cognitive
The Health Economics of dementia. London, United impairment. Pharmacoeconomics, 16: 409–416.
Kingdom: Wiley. Jönsson, L., Lindgren, P., Wimo, A. et al. (1999b). The cost-
Hurd, M.D., Martorell, P. and Langa, K.M. (2013). effectiveness of donepezil therapy in Swedish patients
Monetary costs of dementia in the United States. New with Alzheimer’s disease: A Markov model. Clinical
England Journal of Medicine, 369: 489–490. Therapeutics, 21: 1230–1240.
Hux, M.J., O’Brien, B.J., Iskedjian, M. et al. (1998). Relation Jönsson, L. and Wimo, A. (2009). The cost of dementia in
between severity of Alzheimer’s disease and costs of Europe: A review of the evidence, and methodological
caring. CMAJ, 159: 457–465. considerations. Pharmacoeconomics, 27: 391–403.
Ikeda, S., Yamada, Y. and Ikegami, N. (2002). Economic Jutkowitz, E., Gitlin, L.N. and Pizzi, L.T. (2010). Evaluating
evaluation of donepezil treatment for Alzheimer’s willingness-to-pay thresholds for dementia caregiv-
disease in Japan. Dementia and Geriatric Cognitive ing interventions: Application to the tailored activity
Disorders, 13: 33–39. program. Value Health, 13: 720–725.
Jansson, W., Almberg, B., Grafstrom, M. and Winblad, Karlsson, G., Jönsson, B., Wimo, A. and Winblad, B.
B. (1998). The Circle Model – Support for relatives (1998). Methodological issues in health economics of
384 Dementia

dementia. In Wimo, A. Jönsson, B., Karlsson, G. and Lyseng-Williamson, K.A. and Plosker, G.L. (2002).
Winblad, B. (eds.), Health Economics of Dementia. Galantamine: A pharmacoeconomic review of its
London, United Kingdom: John Wiley & Sons. use in Alzheimer’s disease. Pharmacoeconomics, 20:
Kasuya, M. and Meguro, K. (2010). Health economic effect 919–942.
of donepezil treatment for CDR 0.5 converters to Lyseng-Williamson, K.A. and Plosker, G. L. (2003).
Alzheimer’s disease as shown by the Markov model. Spotlight on galantamine in Alzheimer’s disease.
Archives of Gerontology Geriatrics, 50: 295–299. Disease Management and Health Outcomes, 11:
Kerner, D.N., Patterson, T.L., Grant, I. and Kaplan, R.M. 125–128.
(1998). Validity of the quality of well-being scale for Maas, M.L., Specht, J.P., Weiler, K. et al. (1998). Special
patients with Alzheimer’s disease. Journal of Aging care units for people with Alzheimer’s disease. Only
and Health, 10: 44–61. for the privileged few? Journal of Gerontological
Knapp, M., Iemmi, V. and Romeo, R. (2013). Dementia care Nursing, 24: 28–37.
costs and outcomes: A systematic review. International Martikainen, J., Valtonen, H. and Pirttila, T. (2004).
Journal of Geriatric Psychiatry, 28: 551–561. Potential cost-effectiveness of a family-based program
Knapp, M.J., Knopman, D.S., Solomon, P.R. et al. (1994). in mild Alzheimer’s disease patients. The European
A 30-week randomized controlled trial of high-dose Journal of Health Economics, 5: 136–142.
tacrine in patients with Alzheimer’s disease. The Mauskopf, J.A., Paul, J.E., Grant, D.M. and Stergachis,
Tacrine Study Group. The Journal of the American A. (1998). The role of cost-consequence analysis in
Medical Association, 271: 985–991. healthcare decision-making. Pharmacoeconomics, 13:
Knapp, M. and Prince, M. (2007). Dementia UK. London, 277–288.
United Kingdom: Alzheimer’s Society. Max, W. (1996). The cost of Alzheimer’s disease. Will drug
Knopman, D., Schneider, L., Davis, K. et al. (1996). Long- treatment ease the burden? Pharmacoeconomics, 9:
term tacrine (Cognex) treatment: Effects on nursing 5–10.
home placement and mortality, Tacrine Study Group. Max, W., Webber, P. and Fox, P. (1995). Alzheimer’s dis-
Neurology, 47: 166–177. ease. The unpaid burden of caring. Journal of Aging
Konig, M. and Wettstein, A. (2002). Caring for relatives with and Health, 7: 179–199.
dementia: Willingness-to-pay for a reduction in care- Melis, R.J., Adang, E., Teerenstra, S. et al. (2008).
giver’s burden. Expert Review of Pharmacoeconomics Multidimensional geriatric assessment: Back to the
and Outcomes Research, 2: 535–547. future cost-effectiveness of a multidisciplinary inter-
Koopmanschap, M.A. (1998). Indirect costs and cost- vention model for community-dwelling frail older peo-
ing informal care. In Wimo, A., Karlsson, G., Jönsson, ple. The Journals of Gerontology. Series A, Biological
B. and Winblad, B. (eds.), The Health Economics of Sciences and Medical Sciences, 63: 275–282.
Dementia. London, United Kingdom: John Wiley & Mesterton, J., Wimo, A., By, A. et al. (2010). Cross sectional
Sons. observational study on the societal costs of Alzheimer’s
Lamb, H.M. and Goa, K.L. (2001). Rivastigmine. A pharma- disease. Current Alzheimer Research, 7: 358–367.
coeconomic review of its use in Alzheimer’s disease. Migliaccio-Walle, K., Getsios, D., Caro, J.J. et al. (2003).
Pharmacoeconomics, 19: 303–318. Economic evaluation of galantamine in the treatment
Langa, K.M., Chernew, M.E., Kabeto, M.U. et al. (2001). of mild to moderate Alzheimer’s disease in the United
National estimates of the quantity and cost of informal States. Clinical Therapeutics, 25: 1806–1825.
caregiving for the elderly with dementia. Journal of Mohide, E.A., Torrance, G.W., Streiner, D.L. et al. (1988).
General Internal Medicine, 16: 770–778. Measuring the wellbeing of family caregivers using
Leung, G.M., Yeung, R.Y., Chi, I. and Chu, L.W. (2003). the time trade-off technique. Journal of Clinical
The economics of Alzheimer disease. Dementia and Epidemiology, 41: 475–482.
Geriatric Cognitive Disorders, 15: 34–43. Moore, M.J., Zhu, C.W. and Clipp, E.C. (2001). Informal
Lindgren, B. (1981). Costs of Illness in Sweden 1964-1975. costs of dementia care: Estimates from the National
Lund, Sweden: Liber. Longitudinal Caregiver Study. The Journals of
Lindholm, C., Gustavsson, A., Jönsson, L. and Wimo, A. Gerontology. Series B, Psychological Sciences and
(2013). Costs explained by function rather than diagno- Social Sciences, 56: S219–S228.
sis – Results from the SNAC Nordanstig elderly cohort Muangpaisan, W., Praditsuwan, R., Assanasen, W. et al.
in Sweden. International Journal of Geriatric Psychiatry, (2010). Caregiver burden and needs of dementia care-
28: 454–462. givers in Thailand: A cross-sectional study. Journal of
Logsdon, R.G., Gibbons, L.E., McCurry, S.M. and Teri, L. the Medical Association of Thailand, 93: 601–607.
(2002). Assessing quality of life in older adults with cog- Mulhern, B., Rowen, D., Brazier, J. et al. (2013).
nitive impairment. Psychosomatic Medicine, 64: 510–519. Development of DEMQOL-U and DEMQOL-PROXY-U:
Lovestone, S. (2002). Can we afford to develop treatments Generation of preference-based indices from DEMQOL
for dementia? Journal of Neurology, Neurosurgery & and DEMQOL-PROXY for use in economic evaluation.
Psychiatry, 72: 685. Health Technology Assessment, 17 (5): 1–140.
Health economic aspects of dementia 385

Neumann, P.J., Hermann, R.C., Kuntz, K.M. et al. (1999). Reisberg, B., Doody, R., Stoffler, A. et al. (2003). Memantine
Cost-effectiveness of donepezil in the treatment of in moderate-to-severe Alzheimer’s disease. New
mild or moderate Alzheimer’s disease. Neurology, 52: England Journal of Medicine, 348: 1333–1341.
1138–1145. Reisberg, B., Ferris, S.H., De Leon, M.J. and Crook,
Neumann, P.J., Sandberg, E.A., Araki, S.S. et al. (2000). T. (1988). Global Deterioration Scale (GDS).
A comparison of HUI2 and HUI3 utility scores in Psychopharmacology Bulletin, 24: 661–663.
Alzheimer’s disease. Medical Decision Making, 20: Rice, D.P., Fox, P.J., Max, W. et al. (1993). The economic
413–422. burden of Alzheimer’s disease care. Health Affairs
NICE (2001). Guidance on the Use of Donepezil, (Millwood), 12: 164–176.
Rivastigmine and Galantamine for the Treatment of Rice, D.P., Hodgson, T.A. and Kopstein, A.N. (1985). The
Alzheimer’s Disease. London, UK: National Institute for economic costs of illness: A replication and update.
Clinical Excellence (NICE). Health Care Financing Review, 7: 61–80.
NICE (2009). Donepezil, Galantamine, Rivastigmine Rigaud, A.S., Fagnani, F., Bayle, C. et al. (2003). Patients
(Review) and Memantine for the Treatment of with Alzheimer’s disease living at home in France:
Alzheimer’s Disease (Amended). London, UK: National Costs and consequences of the disease. Journal of
Institute for Clinical Excellence (NICE). Geriatric Psychiatry and Neurology, 16: 140–145.
Nocera, S., Bonato, D. and Telser, H. (2002). The con- Rosen, W.G., Mohs, R.C. and Davis, K.L. (1984). A new
tingency of contingent valuation. How much are rating scale for Alzheimer’s disease. The American
people willing to pay against Alzheimer’s disease? Journal of Psychiatry, 141: 1356–1364.
International Journal of Health Care Finance and Rosenheck, R.A., Leslie, D.L., Sindelar, J.L. et al. (2007).
Economics, 2: 219–240. Cost-benefit analysis of second-generation anti-
Nordberg, G., Von Strauss, E., Kareholt, I. et al. (2005). psychotics and placebo in a randomized trial of the
The amount of informal and formal care among non- treatment of psychosis and aggression in Alzheimer
demented and demented elderly persons-results disease. Archives of General Psychiatry, 64: 1259–1268.
from a Swedish population-based study. International Salek, S.S., Walker, M.D. and Bayer, A.J. (1998). A review
Journal of Geriatric Psychiatry, 20: 862–871. of quality of life in Alzheimer’s disease. Part 2: Issues
O’Brien, B.J., Goeree, R., Hux, M. et al. (1999). in assessing drug effects. Pharmacoeconomics, 14:
Economic evaluation of donepezil for the treatment 613–627.
of Alzheimer’s disease in Canada. Journal of the Sano, M., Ernesto, C., Thomas, R.G. et al. (1997). A con-
American Geriatric Society, 47: 570–578. trolled trial of selegiline, alpha-tocopherol, or both as
OECD (2013). Public spending on health and long-term treatment for Alzheimer’s disease. The Alzheimer’s
care: A new set of projections. In OECD (ed.), OECD Disease Cooperative Study. New England Journal of
Economic Policy Papers. Paris, France: OECD. Medicine, 336: 1216–1222.
OECD (2015). Addressing dementia. In OECD (ed.), OECD Sano, M., Zhu, C.W., Whitehouse, P.J. et al. (2006). ADCS
Health Policy Studies. Paris, France: OECD. Prevention Instrument Project: Pharmacoeconomics:
OECD PPP (2003). Prices and purchasing power pari- Assessing health-related resource use among healthy
ties (PPP). Available at http:/www.oecd.org/std/ppp/. elderly. Alzheimer Disease and Associated Disorders,
Accessed 14 June 2016. 20: S191–S202.
Olin, J. and Schneider, L. (2003). Galantamine for SBU (2008). Dementia. A Systematic Review. Stockholm,
Alzheimer’s Disease (Cochrane Review). Oxford, Sweden: Staten beredning för medicinsk utvärder-
United Kingdom: The Cochrane Library. ing (SBU) (The Swedish Council on Technology
Pouryamout, L., Dams, J., Wasem, J. et al. (2012). Assessment in Health Care).
Economic evaluation of treatment options in patients Schaller, S., Mauskopf, J., Kriza, C. et al. (2015). The
with Alzheimer’s disease: A systematic review of cost- main cost drivers in dementia: A systematic review.
effectiveness analyses. Drugs, 72: 789–802. International Journal of Geriatric Psychiatry, 30: 111–129.
Prince, M., Knapp, M., Guerchet, M. et al. (2014). Schneider, L. (1998). Designing phase III trials of anti-
Dementia UK: An Update. London, United Kingdom: dementia drugs with a view towards pharmacoeconomi-
Alzheimer’s Society. cal considerations. In Wimo, A., Jönsson, B., Karlsson, G.
Quentin, W., Riedel-Heller, S.G., Luppa, M. et al. (2009). and Winblad, B. (eds.), Health Economics of Dementia.
Cost-of-illness studies of dementia: A systematic London, United Kingdom: John Wiley & Sons.
review focusing on stage dependency of costs. Acta Schneider, L.S., Olin, J.T., Lyness, S.A. and Chui, H.C. (1997).
Psychiatrica Scandinavica, 121: 243–259. Eligibility of Alzheimer’s disease clinic patients for clinical
Raccichini, A., Castellani, S., Civerchia, P. et al. (2009). The trials. Journal of American Geriatric Society, 45: 923–928.
caregiver’s burden of Alzheimer patients: Differences Schulz, R. and Beach, S.R. (1999). Caregiving as a risk fac-
between live-in and non-live-in. American Journal tor for mortality: The caregiver health effects study.
of Alzheimer’s Diseases and Other Dementia, 24: The Journal of the American Medical Association, 282:
377–383. 2215–2219.
386 Dementia

Schwarzkopf, L., Menn, P., Kunz, S. et al. (2011). Costs of Torrance, G. (1996). Designing and conducting
care for dementia patients in community setting: An cost–utility analysis. In Spilker, B. (ed.), Quality
analysis for mild and moderate disease stage. Value of Life and Pharmacoeconomics in Clinical Trials.
Health, 14: 827–835. Philadelphia, PA: Lippincott-Raven Publishers,
Selai, C., Vaughan, A., Harvey, R.J. and Logsdon, R. (2001). pp. 1105–1112.
Using the QOL-AD in the UK. International Journal of Torrance, G.W. (1997). Preferences for health outcomes
Geriatric Psychiatry, 16: 537–538. and cost-utility analysis. The American Journal of
Selai, C.E., Elstner, K. and Trimble, M.R. (2000). Quality of Managed Care, 3 (Suppl. 1): S8–S20.
life pre and post epilepsy surgery. Epilepsy Research, Torrance, G.W., Feeny, D.H., Furlong, W.J. et al. (1996).
38: 67–74. Multiattribute utility function for a comprehensive
Shukla, V.K., Otten, N. and Coyle, D. (2000). Drug health status classification system. Health Utilities
Treatments for Alzheimer’s Disease III. A Review of Index Mark 2. Medical Care, 34: 702–722.
Published Pharmacoeconomic Evaluations. Ottawa, Tsuchiya, A., Dolan, P. and Shaw, R. (2003). Measuring
Canada: Canadian Coordinating Office for Health people’s preferences regarding ageism in health:
Technology Assessment (CCOHTA). Some methodological issues and some fresh evidence.
Siegel, J.E., Torrance, G.W., Russell, L.B. et al. (1997). Social Science and Medicine, 57: 687–696.
Guidelines for pharmacoeconomic studies. Walker, M.D., Salek, S.S. and Bayer, A.J. (1998). A review
Recommendations from the panel on cost effectiveness of quality of life in Alzheimer’s disease. Part 1: Issues
in health and medicine. Panel on cost-effectiveness in in assessing disease impact. Pharmacoeconomics, 14:
Health and Medicine. Pharmacoeconomics, 11: 159–168. 499–530.
Sintonen, H. (2001). The 15D instrument of health-related Wallin, A.K., Gustafson, L., Sjogren, M. et al. (2004). Five-
quality of life: Properties and applications. Annals of year outcome of cholinergic treatment of Alzheimer’s
Medicine, 33: 328–336. disease: Early response predicts prolonged time until
Skoldunger, A., Johnell, K., Winblad, B. and Wimo, A. nursing home placement, but does not alter life expec-
(2013). Mortality and treatment costs have a great tancy. Dementia of Geriatric Cognitive Disorders, 18:
impact on the cost-effectiveness of disease modifying 197–206.
treatment in Alzheimer’s disease – A simulation study. Walters, S.J. and Brazier, J.E. (2003). What is the relation-
Current Alzheimer Research, 10: 207–216. ship between the minimally important difference and
Smith, S.C., Lamping, D.L., Banerjee, S. et al. (2007). health state utility values? The case of the SF-6D.
Development of a new measure of health-related Health and Quality of Life Outcomes, 1 (1): 4.
quality of life for people with dementia: DEMQOL. van den Berg, B., Brouwer, W., Van Exel, J. et al. (2006).
Psychological Medicine, 37: 737–746. Economic valuation of informal care: Lessons from
Sonnenberg, F.A. and Leventhal, E.A. (1998). Modeling the application of the opportunity costs and proxy
disease progression with Markov models. In Wimo, A., good methods. Social Science and Medicine, 62:
Jönsson, B., Karlsson, G. and Winblad, B. (eds.), Health 835–845.
Economics of Dementia. London, United Kingdom: Ward, A., Caro, J.J., Getsios, D. et al. (2003). Assessment
John Wiley & Sons. of Health Economics in Alzheimer’s Disease (AHEAD):
Spilker, B. (1996). Quality of Life and Pharmacoeconomics Treatment with galantamine in the UK. International
in Clinical Trials. Philadelphia, PA: Lippincott-Raven Journal of Geriatric Psychiatry, 18: 740–747.
Publishers. Ware, J.E., Jr. and Sherbourne, C.D. (1992). The MOS
Stewart, A. and Brod, M. (1996). Measuring health related 36-item short-form health survey (SF-36). I. Conceptual
quality of life in older and demented people. In Spilker, framework and item selection. Medical Care, 30:
B. (ed.), Quality of Life and Pharmacoeconomics in 473–483.
Clinical Trials. Philadelphia, PA: Lippincott-Raven Weinstein, M.C. and Fineberg, H.V. (1980). Clinical
Publishers, pp. 819–830. Decision Analysis. Philadelphia, PA: W.B. Saunders.
Stewart, A., Phillips, R. and Dempsey, G. (1998). Wimo, A. (2009). The art of cost of illness. Journal of
Pharmacotherapy for people with Alzheimer’s disease: Alzheimer’s Disease, 19 (2): 617–619.
A Markov-cycle evaluation of five years’ therapy using Wimo, A., Ballard, C., Brayne, C. et al. (2014a). Health
donepezil. International Journal of Geriatric Psychiatry, economic evaluation of treatments for AD: Impact of
13: 445–453. new diagnostic criteria. Journal of Internal Medicine,
Stommel, M., Collins, C.E. and Given, B.A. (1994). The 275 (3): 304–316.
costs of family contributions to the care of persons Wimo, A., Johansson, L. and Jönsson, L. (2009).
with dementia. Gerontologist, 34: 199–205. [Prevalence study of societal costs for dementia 2000-
Suh, G.H., Wimo, A., Gauthier, S. et al. (2009). 2005. More demented people--but somewhat reduced
International price comparisons of Alzheimer’s drugs: costs per person]. Läkartidningen, 106: 1277–1282.
A way to close the affordability gap. International Wimo, A., Jönsson, L., Bond, J. et al. (2013a). The world-
Psychogeriatrics, 21 (6): 1116–1126. wide economic impact of dementia 2010. Alzheimer’s
Health economic aspects of dementia 387

& Dementia: The Journal of the Alzheimer’s Wimo, A. and Winblad, B. (2003b). Resource utilisation in
Association, 9: 1–11. dementia: RUD Lite. Brain Aging, 3: 48–59.
Wimo, A., J Jönsson, L., Fratiglioni, L. et al. (2014b). Wimo, A., Winblad, B., Engedal, K. et al. (2003a). An
Demenssjukdomarnas samhällskostnader i Sverige economic evaluation of donepezil in mild to moderate
2012. In Socialstyrelsen. Stockholm, Sweden: Alzheimer’s disease: Results of a 1-year, double-blind,
Socialstyrelsen. randomized trial. Dementia of Geriatric Cognitive
Wimo, A., Jönsson, L. and Gustavsson, A. (2008). The Disorders, 15: 44–54.
cost of illness and burden of dementia in Europe. In Wimo, A., Winblad, B. and Grafstrom, M. (1999b). The
Europe, A. (ed.), Dementia in Europe Yearbook 2008. social consequences for families with Alzheimer’s
Luxembourg, Europe: Alzheimer Europe. disease patients: Potential impact of new drug treat-
Wimo, A., Jönsson, B., Karlsson, G. and Winblad, B.E. ment. International Journal of Geriatric Psychiatry, 14:
(1998a). The Health Economics of Dementia. London, 338–347.
United Kingdom: John Wiley & Sons. Wimo, A., Winblad, B., Stoffler, A. et al. (2003b). Resource
Wimo, A., Johansson, L., Von Strauss, E. and Nordberg, utilisation and cost analysis of memantine in patients
G. (1999a). Formal and informal home care to Swedish with moderate to severe Alzheimer’s disease.
demented patients, an application of RUD (Resource Pharmacoeconomics, 21: 327–340.
Utilization in Dementia) (abstract). International Wimo, A., Witthaus, E., Rother, M. and Winblad, B.
Psychogeriatrics, 11 (Suppl. 1): S197. (1998c). Economic impact of introducing propentofyl-
Wimo, A., Mattsson, B., Krakau, I. et al. (1994). Cost- line for the treatment of dementia in Sweden. Clinical
effectiveness analysis of day care for patients with Therapeutics, 20: 552–566.
dementia disorders. Health Economics, 3: 395–404. Winblad, B., Hill, S., Beermann, B. et al. (1997). Issues in
Wimo, A., Mattson, B., Krakau, I. et al. (1995). Cost-utility the economic evaluation of treatment for dementia.
analysis of group living in dementia care. International Position paper from the International Working Group
Journal of Technology Assessment in Health Care, 11: on Harmonization of Dementia Drug Guidelines.
49–65. Alzheimer Disease and Associated Disorders, 11:
Wimo, A., Nordberg, G., Jansson, W. and Grafstrom, M. 39–45.
(2000). Assessment of informal services to demented Winblad, B., Kilander, L., Eriksson, S. (2006). Donepezil
people with the RUD instrument. International Journal in patients with severe Alzheimer’s disease: Double-
of Geriatric Psychiatry, 15: 969–971. blind, parallel-group, placebo-controlled study.
Wimo, A. and Prince, M. (2010). World Alzheimer Report Lancet, 367: 1057–1065.
2010. The Global Economic Impact of Dementia. Winblad, B., Wimo, A., Mobius, H.J. et al. (1999). Severe
London, United Kingdom: Alzheimer’s Disease dementia: A common condition entailing high costs at
International. individual and societal levels. International Journal of
Wimo, A., Reed, C.C., Dodel, R. et al. (2013b). The GERAS Geriatric Psychiatry, 14: 911–914.
Study: A prospective observational study of costs and Wolfs, C.A., Dirksen, C.D., Kessels, A. et al. (2007).
resource use in community dwellers with Alzheimer’s Economic evaluation of an integrated diagnostic
disease in three European countries – Study design approach for psychogeriatric patients: Results of
and baseline findings. Journal of Alzheimer’s Disease, a randomized controlled trial. In Wolfs, C. A. (ed.),
36 (2): 385–399. An Integrated Approach to Dementia. A Clinical
Wimo, A., Von Strauss, E., Nordberg, G. et al. (2002). Time and Economic Evaluation (thesis). Maastricht, The
spent on informal and formal care giving for persons Netherlands: Neuropsych Publishers.
with dementia in Sweden. Health Policy, 61: 255–268. Wolfson, C., Oremus, M., Shukla, V. et al. (2002). Donepezil
Wimo, A., Wallin, J.O., Lundgren, K. et al. (1990). Impact and rivastigmine in the treatment of Alzheimer’s
of day care on dementia patients – Costs, well-being disease: A best-evidence synthesis of the published
and relatives’ views. Family Practice, 7: 279–287. data on their efficacy and cost-effectiveness. Clinical
Wimo, A., Wetterholm, A.L., Mastey, V. and Winblad, B. Therapeutics, 24: 862–886; discussion 837.
(1998b). Evaluation of the resource utilization and care- Zarit, S.H., Reever, K.E. and Bach-Peterson, J. (1980).
giver time in anti-dementia drug trials – A quantitative Relatives of the impaired elderly: Correlates of feelings
battery. In Wimo, A., Jönsson, B., Karlsson, G. and of burden. Gerontologist, 20: 649–655.
Winblad, B. (eds.), The Health Economis of Dementia. Zhang, Y., Kivipelto, M., Solomon, A. and Wimo, A. (2011).
London, United Kingdom: John Wiley & Sons. Cost-effectiveness of a health intervention program
Wimo, A. and Winblad, B. (2003a). Pharmacoeconomics with risk reductions for getting demented: Results of a
of mild cognitive impairment. Acta Neurologica Markov model in a Swedish/Finnish setting. Journal of
Scandinavica, 107 (Suppl. 179): S94–S99. Alzheimer’s Disease, 26: 735–744.
38
Global challenge of dementia: What can be
done?

CLEUSA P. FERRI, MAËLENN GUERCHET AND MARTIN PRINCE

38.1 GLOBAL CHALLENGE OF Lack of awareness directly affects help-seeking behaviour


DEMENTIA and hence the recognition and management of the condi-
tion, which in turn contribute to the burden experienced by
the family and other main carers of people with dementia.
Demographic ageing has been a much more rapid process in
all world regions than anticipated, with the population aged
60 and over increasing at the fastest pace ever in developing 38.1.1 PREVALENCE
countries (World Population Prospects, 2003). Globally, the
numbers and proportion of older people are sharply increas- Alzheimer’s Disease International (ADI), the umbrella
ing. In 2012, 23% of the population in high income countries organization for national Alzheimer’s associations (AA),
was aged 60 years or over, and this is predicted to increase has been collating evidence since 2004 on the global preva-
to 32% by 2050. This proportion in low- and ­middle‑income lence of dementia, the projected increases over time and its
countries (LMIC) was just 9% in 2012 but will be more than impact worldwide.
double by 2050, reaching 19%. Currently, 66% of the world’s In 2004, ADI convened a panel of experts to review
older population live in LMIC, and by 2050, 79% will do so the global evidence on the prevalence, numbers of people
(World Population Prospects, 2013). Chronic non-commu- affected and its impact. The figures from this consensus
nicable diseases, which have been under-prioritized in the exercise among experts (Ferri et al., 2005) were revisited for
public health agenda for a long time, are now assuming a the 2009 World Alzheimer’s Report (Alzheimer’s Disease
greater significance in LMIC. International, 2009) when a systematic review of the world
Co-morbid health conditions are particularly common literature on the prevalence of dementia was conducted and
among older people with physical conditions affecting dif- a quantitative meta-analysis was attempted to synthesize
ferent organ systems coexisting with mental and cogni- the available evidence for some regions, rather than only
tive disorders. Dementia has a disproportionate impact on relying on expert consensus as had been done previously.
dependency and disability and yet its global public health Since the consensus estimates were published, the global
importance is still underappreciated. There is a general lack evidence-base has expanded considerably. There have been
of awareness of Alzheimer’s and other dementias as medi- new studies from Europe (Lobo et al., 2007; Fernandez
cal conditions. Dementia is under-recognized, even in high- et al., 2008) and the United States (Plassman et al., 2007),
income countries such as the United Kingdom, where the and an explosion of studies from LMIC and other regions
national under-detection rate is about 52% (Department of and groups previously under-represented in the literature
Health, 2013). This under-recognition seems to be accen- (Alzheimer’s Disease International, 2009) (Latin America,
tuated in LMIC, where they are commonly perceived as a India, China, Korea, Thailand, Australia [indigenous peo-
normal part of ageing (Patel and Prince, 2001; Lopez et al., ple], Guam, Poland and Turkey). By 2009, 41% (64/158) of
2014). In India, Dias and Patel (2009) reported that 90% dementia prevalence studies had been conducted in LMIC.
of patients with dementia recruited into a trial in primary A very different situation compared to 1998, when the 10/66
care had not been diagnosed, and in Brazil rates of under- Dementia Research Group was founded – its title being
diagnosis might be as high as 77% (Nakamura et al., 2015). derived from the estimate that only 10% of population-based
388
Global challenge of dementia: What can be done? 389

research had been conducted in LMIC, relative to the two- Worldwide, 7.7 million new cases of dementia were antici-
thirds of people with dementia living in those regions. The pated each year, implying one new case every 4.1 seconds;
total number of people with dementia was estimated at 3.6 million (46%) would impact in Asia, 2.3 million (31%) in
35.6 million in 2010, 48.1 million in 2020 and 90.3 million in Europe, 1.2 million (16%) in the Americas and 0.5 million (7%)
2040, with a projection of numbers doubling every 20 years in Africa.
(Alzheimer’s Disease International, 2009). Those estimates For the year 2010, prevalence (35.6 million cases) was 4.6
were about 10% higher than the previous ones (Ferri et al., times annual incidence, suggesting an approximate average
2005). In 2013, an update used a new systematic review of survival from onset of 4.6 years, broadly consistent with
the prevalence of dementia in China (Chan et al., 2013) com- earlier estimates from case series (Fitzpatrick et al., 2005).
prising 75 studies, and identified seven studies from five sub-
Saharan African countries (Hendrie et al., 1995; Guerchet 38.1.3 BURDEN
et al., 2009; Guerchet et al., 2010; Paraiso et al., 2011; Yusuf
et al., 2011; Longdon et al., 2013; Guerchet et al., 2014), where The number of people worldwide affected by dementia
previously only one study from Nigeria had been available provides one indicator of the impact of the disease, with-
(Hendrie et al., 1995). The regional prevalence of dementia out fully capturing the problem for the individual affected,
estimated from China and sub-Saharan Africa studies were their families and society. The impact of dementia can be
substantially higher than those used in 2009 (Prince et al., understood at three levels: (1) The person with dementia,
2013b). As more data becomes available, the variation in who experiences ill health, disability, impaired quality of
prevalence between world regions is reduced. life and reduced life expectancy; (2) The family and friends
The 2013 updated estimates were higher than the origi- of the person with dementia, who in all world regions, pro-
nal estimates reported in the 2009 World Alzheimer Report, vide the cornerstone of care and support for the person with
with the number of people living with dementia worldwide dementia; (3) Wider society, health and long-term care sys-
estimated at 44.3 million in 2013, reaching 75.4 million in tems and lost productivity.
2030 (15% higher) and 135.5 million in 2050 (17% higher) The Global Burden of Disease study shows that non-
(Prince et al., 2013b). Over the next 20 years, the number communicable diseases are rapidly becoming the dominant
of people with dementia is anticipated to increase by 35% causes of ill-health in all developing regions except sub-
in Europe, 59% in North America, 65% in Southern Latin Saharan Africa (Fuster and Voute, 2005). Dementia is one
America and 69% in developed Asia-Pacific. In comparison, of the main causes of disability in later life, but it is impor-
the percentage increase is expected to be 114% in South East tant to understand the contribution of dementia, relative
Asia, 106%–120% in Latin America and 88% in Africa. From to that of other chronic diseases. The estimates contained
2013 to 2050, the number of people with dementia will have in the WHO’s Global Burden of Disease (GBD) Study, first
increased slightly less than twofold in Europe, more than two- published in 1996 and updated to 2010, provide important
fold in North America, threefold in Asia and fourfold in Latin evidence in this respect (World Health Organization, 1996;
America and Africa. While 38% of people with dementia live World Health Organization, 2006; Murray et al., 2012a).
in high-income countries, 62% live in LMICs. By 2050, this These are highly influential in terms of prioritization for
proportion will have increased to 71% (Prince et al., 2013b). policymaking and planning at national, regional and inter-
national levels. The key indicator is the disability-adjusted
38.1.2 INCIDENCE life year (DALY), a single integrated measure of disease
burden calculated as the sum of years lived with disability
The most recent available estimates for the global inci- (YLD) and years of life lost (YLL). In the 2010 GBD study,
dence of dementia come from the systematic review con- disability weights were measured for 220 unique health
ducted for the 2012 World Health Organization (WHO)/ states after a large-scale empirical survey using a scale from
ADI joint report (World Health Organization, 2012) where 0 (implying no loss of health) to 1 (implying a health loss
34 fully eligible studies were identified. While the evidence equivalent to death). Disability from dementia was accorded
base from Europe and North America dominated, 13 of the a disability weight from 0.082 for mild dementia to 0.438
34 studies were from outside these regions, and 10 studies for severe dementia (Salomon et al., 2012), a lower weight
were conducted in countries with low or middle income. than the one (0.67) attributed in the previous GBD report
The incidence of dementia increased exponentially with (World Health Organization, 2004a). These weights were
increasing age. For all the studies combined, the incidence equivalent to the disability weights for having tuberculosis
of dementia doubled with every 5.9-year increase in age, with HIV infection, untreated epilepsy or a major depres-
from 3.1/1000 person years (pyr) at age 60–64 to 175.0/1000 sive disorder, and slightly lower than those for metastatic
pyr at age 95 or more. Estimates of the global annual num- cancer and severe stroke (Salomon et al., 2012). A weight
bers of new cases were derived for the year 2010. Number of of 0.44 signifies that each year lived with severe dementia
new cases increased and then declined with increasing age entails the loss of almost a half of one DALY. According to
in each region; in Europe and the Americas peak incidence the GBD, at the global level, dementia accounted for 11.3
was among those aged 80–89 years, in Asia it was among million DALYs and was one of the 15 causes with the largest
those aged 75–84 and in Africa among those aged 70–79. increase. Alzheimer’s disease (AD) and other dementia were
390 Dementia

ranked the 49th leading cause of DALYs in 2010 globally, Costs in LMIC are likely to rise faster than in high-
but were the 11th and 12th in Western Europe and high- income countries, for several reasons. Wage levels are rising
income North America, respectively (Murray et al., 2012b). rapidly, and hence, so is the opportunity cost or replacement
Around half of all people with dementia need personal cost of informal care. While currently very few people with
care and the others will develop such needs over time. People dementia in those regions live in care homes, this sector is
with dementia have special needs for care. Compared with expanding rapidly, particularly in urban settings in mid-
other long-term care users they need more personal care, dle-income countries, boosted by demographic and social
more hours of care and more supervision, all of which is changes that reduce the availability of family members to
associated with greater caregiver strain, and higher costs provide care. Medical help-seeking is relatively unusual;
of care. Inevitably, the number of dependent older people, demand for medical care is likely to increase in the future,
including those with dementia, will increase markedly in with improved awareness, better coverage of evidence-based
the coming decades, particularly in middle-income coun- interventions, and, possibly, more effective treatments.
tries (Prince et al., 2013c). The 10/66 Dementia Research Group has also examined
The economic costs of dementia are enormous. These the economic impact of dementia in its pilot study of 706
can include the costs of (1) ‘formal care’ – healthcare, social/ persons with dementia, and their caregivers, living in Latin
community care, respite and long-term residential or nurs- America, India, China and Nigeria (Prince, 2004). One of the
ing home care and (2) ‘informal care’ – unpaid care by family key findings from this study, from the development perspec-
members including their lost opportunity to earn income. tive, was that caregiving in the developing world is associated
The total estimated worldwide costs of dementia were with substantial economic disadvantage. A high proportion
US$604 billion in 2010, equivalent to 1% of the world’s gross of caregivers had to cut back on their paid work to care. Many
domestic product (Wimo et al., 2013). Low-income countries carers needed and obtained additional support, and while
accounted for just less than 1% of total worldwide costs (but this was often informal unpaid care from friends and other
14% of the prevalence), middle-income countries for 10% family members, paid caregivers were also relatively com-
of the costs (but 40% of the prevalence) and high-income mon. People with dementia were heavy users of health ser-
countries for 89% of the costs (but 46% of the prevalence). vices, and associated direct costs were high. Compensatory
About 70% of the global costs occurred in just two regions: financial support was negligible; few older people in devel-
Western Europe and North America. These discrepancies oping countries receive government or occupational pen-
are accounted for by the much lower costs per person in sions, and virtually none of the people with dementia in the
lower-income countries – US$868 in low-income countries, 10/66 study received disability pensions. Carers were com-
US$3,109 in lower-middle-income, US$6,827 in upper-mid- monly in paid employment, and almost none received any
dle-income and US$32,865 in high-income countries. form of carer allowance (Prince, 2004). The combination of
These costs are driven mainly by social care needs; reduced family incomes and increased family expenditure
healthcare costs account for a small proportion of the total, on care is obviously particularly stressful in lower-income
given the low diagnosis rate, limited therapeutic options countries where so many households exist at or near to
and the underutilization of existing evidence-based inter- subsistence level. While healthcare services are cheaper in
ventions (Prince et al., 2011b). In all world regions, informal low-income countries, in relative terms, families from the
care provided by family, friends and the community is the poorer countries spend a greater proportion of their income
cornerstone of the care system. In lower-income countries, on healthcare for the person with dementia. They also tend
these informal care costs predominate, accounting for 58% selectively to use the more expensive services of private doc-
and 65% of all costs in low-income and middle-income tors. Numerous studies have shown that the costs of caring
countries, respectively, compared with 40% in high-income can extend beyond demands on time and money and can
countries. Conversely, in high-income countries, the direct affect the carer’s health and morale for a longer term.
costs of social care (professional care in the community, and
the costs of residential and nursing home care) account for
the largest element of costs – 42%, compared with only 14% 38.1.4 FUTURE TRENDS IN THE
in LMIC where such services are not generally available. In PREVALENCE, INCIDENCE OF
LMIC, despite larger, extended families, the psychologi- DEMENTIA AND SURVIVAL WITH
cal and economic strain on family caregivers is substan- DEMENTIA
tial (Honyashiki et al., 2011; Prince et al., 2012). Typically,
around a fifth of carers have cut back on paid work, and All current projections of the scale of the coming dementia
paid carers are becoming common in some cities adding to epidemic, including those published by ADI (Alzheimer’s
the economic burden; compensatory benefits are practically Disease International, 2009; Prince et al., 2013b), assume
non-existent (Alzheimer’s Disease International, 2009). that the age- and gender-specific prevalence of dementia
Worldwide, the costs of dementia are set to soar. The will not vary over time, and that population ageing alone
World Alzheimer Report 2010 tentatively estimated an 85% (increasing the number of older people at risk) drives the
increase in costs by 2030, based only on predicted increases projected increases (Ferri et al., 2005; Alzheimer’s Disease
in the numbers of people with dementia (Wimo et al., 2010). International, 2009; Prince et al., 2013a; Prince et al., 2013b).
Global challenge of dementia: What can be done? 391

The basis for this assumption is uncertain, since preva- et al., 2008; Matthews et al., 2013), or incidence of dementia
lence is a product of incidence and survival with dementia, (Schrijvers et al., 2012; Gao et al., 2014; Satizabal et al., 2014)
and a fall in either or both of these indicators would lead this has not been consistently replicated in all other studies
to a fall in age-specific prevalence (Alzheimer’s Disease of this type (Sekita et al., 2010; Mathillas et al., 2011; Rocca
International, 2009). A decline in age-specific incidence, at et al., 2011; Qiu et al., 2013; Gao et al., 2014).
least in high-income countries, was theoretically possible, The relationship between trends in prevalence, inci-
driven by changes in exposure to suspected developmental, dence and mortality are particularly unclear, particularly
lifestyle and cardiovascular risk factors for dementia. since in none of the studies were all three of these param-
The 2014 World Alzheimer Report focused upon demen- eters directly observed. If the onset of dementia occurs
tia risk reduction; the evidence-base for modifiable risk close to the end of the natural life span, fewer years will
factors for dementia (Prince et al., 2014a). The strongest be lived with dementia. Langa et al. (2008) described this
evidence for possible causal associations with dementia was phenomenon as ‘the compression of cognitive morbidity’,
those of low education in early life, hypertension in midlife, a desirable outcome for public health and individual qual-
and smoking and diabetes across the life course. ity of life, resulting in longer, healthier lives, with fewer
In most world regions, each generation is better edu- years spent in a state of reduced independence and need-
cated than the one before. Although trends differ between ing care.
countries, genders, age groups and time periods, there has Two studies suggest that decline in incidence may be
been a general trend in high-income countries towards less greater in younger age groups, suggesting that the incidence
smoking, falling total cholesterol and blood pressure levels of dementia may be being deferred into older age (Gao
and increasing physical activity. However, the prevalence et al., 2014; Satizabal et al., 2014). In two other studies, sur-
of obesity and diabetes has been increasing in most devel- vival with dementia seemed to be decreasing (Langa et al.,
oped countries. The trends in cardiovascular health among 2008; Doblhammer et al., 2014), supporting the notion of
older people in many low- and particularly middle-income ‘compression of cognitive morbidity’ (Langa et al., 2008).
countries are in an adverse direction (Prince et al., 2015), However, in other studies survival with dementia seemed to
with a pattern of increasing stroke (Feigin et al., 2009) and be increasing (Schrijvers et al., 2012; Qiu et al., 2013), or this
ischaemic heart disease (Critchley et al., 2004; Gupta et al., could be inferred from a pattern of stable prevalence with
2008; Gaziano et al., 2010) morbidity and mortality, linked declining incidence (Gao et al., 2014; Rocca et al., 2011).
to an epidemic of obesity, and increasing blood pressure lev- There is some suggestion that favourable trends are less
els (Anand and Yusuf, 2011). evident in more deprived high-income country populations
After a lag period, to the extent that these factors are gen- (Rocca et al., 2011). Only two studies have been conducted
uinely causally associated with dementia, one would expect outside of Europe or the United States; in Japan, prevalence
to see corresponding reductions (or increases) in the inci- had increased (Sekita et al., 2010), while in Nigeria incidence
dence of dementia. The net effect on survival with dementia was stable (Gao et al., 2014).
is harder to estimate, and other factors, for example, stan- Evidence from systematic reviews of prevalence studies
dards of health and social care for people with dementia, in China and other East Asian countries suggests a worry-
and provision of life-prolonging critical interventions, ing trend towards an increase in the prevalence of dementia
might also be expected to have an influence. over the last 20 years (Chan et al., 2013; Wu et al., 2014). This
In 2009, very few data that were available from certain would be consistent with adverse changes in cardiovascular
high-income countries did not suggest any clear pattern of health in this region. A recent modelling exercise assessed
a decline or increase over time in either the incidence or the likely impact of recent increases in obesity among
prevalence of dementia (Alzheimer’s Disease International, middle-aged Chinese on dementia prevalence, assuming
2009). Meta-analyses of European studies conducted since a causal link with dementia; it concluded that the future
1980 did not suggest any secular trend in prevalence (Prince dementia prevalence in China may have been underesti-
et al., 2013a, 2014). mated by up to 19% given the additional impact of epide-
Just a few years later, and linked to a greatly increased miologic transition (Loef and Walach, 2012).
interest in the potential for prevention of dementia by The future course of the global dementia epidemic,
targeting modifiable risk factors (Barnes and Yaffe, 2011; through to 2050 is therefore likely to depend upon the suc-
Lincoln et al., 2014), the quality and extent of the evidence cess or otherwise of continuing efforts to improve public
has expanded greatly, with several studies monitoring health (Prince et al., 2014; Lincoln et al., 2014). Those who
trends in prevalence and/or incidence and dementia mor- will be old in 2050, were born around the 1970s, and have
tality within defined populations, using identical or similar already received their basic education. They are now in
research methodology over time. their third and fourth decades of life, a crucial ‘sensitive
Findings across these studies conducted, mainly, in period’ where, evidence suggests, efforts to prevent, detect
high-income countries, are currently too inconsistent to and control obesity, hypertension, diabetes and high cho-
reach firm and generalizable conclusions regarding under- lesterol are likely to have maximum positive impact upon
lying trends. While some studies have suggested substantial brain health and dementia risk in late-life (Prince et al.,
recent declines in the prevalence (Lobo et al., 2007; Langa 2014; Lincoln et al., 2014).
392 Dementia

Previous modelling exercises have sought to predict Table 38.1. This focuses initial attention on awareness and
what might happen to the prevalence of dementia, given understanding and from this, moves on to risk reduction
our best estimates of risk associations, and possible changes and the underlying issues of capacity building and resource
in those risk factor profiles over time (Loef and Walach, development. Service development starts in primary care,
2012; Norton et al., 2014). An alternative approach is to before secondary care can be considered because this will be
observe and correlate actual changes in risk factor profiles more equitable, benefiting more people with dementia and
and dementia incidence over time. This is a well-estab- their families. It has the potential to prevent or at least delay
lished modelling approach in the cardiovascular disease the need for expensive institutional services that few families
field and has contributed greatly to our understanding of can afford.
the potential for prevention, and the attribution of changes
in disease incidence to specific factors (Laatikainen et al., 38.2.1 INCREASING AWARENESS –
2005; Ford et al., 2007; Vartiainen et al., 2010). Similar DECREASING STIGMA
studies could, in the future, be carried out to monitor the
impact of prevention programmes on the future scale of Awareness raising and increasing understanding are impor-
the dementia epidemic. tant to counter the fatalism and stigma that are often asso-
ciated with dementia. The heart of awareness raising is to
explain that dementia is an illness/disease, that it is not an
inevitable consequence of ageing, and that it is worth assist-
38.2 WHAT CAN BE DONE? ing family carers and people with dementia to live as normal
a life as possible for as long as possible. Efforts to increase
The increasing global burden of dementia will require strong awareness must be accompanied by health system and ser-
commitment and leadership from different parts of society, vice reform, so that help-seeking is met with a supply of
especially from policymakers and health professionals to better-prepared and more responsive services. Awareness
place dementia higher up in the global health agenda, to raising and increasing understanding is also essential for all
improve the lives of people with dementia. Concrete actions those involved in decision making about dementia services.
can be taken to build quality into the process of care and Without a basic understanding of the nature of the illness,
support for people with dementia and their caregivers. its social and economic consequences and the practical steps
Dementia has already become and is becoming a health that can be taken, it is unlikely that either existing services
priority in an increasing number of countries, including will be redesigned or enhanced or that new services will be
Australia, Denmark, England, Finland, France, Israel, the considered.
Netherlands, Norway, Scotland, South Korea, Switzerland, ADI identified raising awareness of dementia among
Taiwan and the United States. The key elements of these people in general, carers and health workers as a global pri-
countries plans are: to raise awareness; to improve diagno- ority (Graham and Brodaty, 1997) as follows:
sis and treatment; and to increase the capacity of healthcare
systems to respond to the challenge of the increasing num- 1. A critical mass of informed carers can assist aware-
ber of people with dementia. ness-raising, provide advice and support to families,
The key elements for countries with limited resources and can work with Alzheimer’s associations to lobby
will differ in its contents and process of implementation and for more services that better meet their needs.
they should start with initiatives that have the maximum 2. Community solidarity can effect change through sup-
impact for as many people as possible. ADI has developed port for policies based on equity and justice – equal
a graduated approach for countries with limited resources access to health and social care, and welfare benefits.
(Alzheimer’s Disease International, 2009), illustrated in Aware communities can provide support, and reduce

Table 38.1 A graduated approach to dementia service development


Awareness and
understanding Capacity building Risk reduction Service development Service development
The public • Family carers • Diet • Primary care • Secondary care
Health and care • Primary healthcare • Exercise • Community • Institutional care
service decision workers • Social interaction support for
makers • Resource families
acquisition
• Legislative and
policy
framework
Source: Alzheimer’s Disease International, World Alzheimer Report 2009, Alzheimer’s Disease International, London, 2009.
Global challenge of dementia: What can be done? 393

stigma and exclusion of those with dementia and media. Also, help-seeking can be promoted by proac-
their carers. Policymakers can be held to account tive case finding. In India and Brazil, community health
by media campaigns and advocacy from committed workers (CHWs), after brief training, were able to identify
non-governmental organizations (NGOs). In high- likely cases of dementia in the community with a posi-
income countries awareness is growing rapidly, with tive predictive value of 66% (Shaji et al., 2002; Ramos-
the media playing an important part (Prince et al., Cerqueira et al., 2005).
2008). Recent evidence-based reports from the United Another step in recognizing/diagnosing dementia is test-
Kingdom and the Australian Alzheimer associations ing. Population screening is not considered cost effective,
garnered media attention and were instrumental in even in high-income countries (National Collaborating
making dementia a national priority. Media in LMIC Centre for Mental Health, 2007). Only a few trials evalu-
can be receptive to these stories, informing the public ating dementia screening in primary care have been per-
and stimulating debate, but efforts are required to formed, with little evidence on its benefits and potential
alert them to the importance of ageing and demen- harms (Brayne et al., 2007). Selective screening of those
tia, and to build their capacity to report the problem having a prior index of suspicion can be carried out either
(Prince et al., 2008). by cognitive testing, or informant report of cognitive and
3. Intergenerational solidarity can be promoted through functional decline. The mini-mental state examination
awareness-raising among children and young adults. (MMSE) (Folstein et al., 1975) is the most widely used cog-
In many LMIC, children or children-in-law are the nitive screen, and adapted versions have been normed for
most frequent carers for people with dementia (Choo use in many LMIC (Bird et al., 1987; Ganguli et al., 1995;
et al., 2003; Prince, 2004; Dias et al., 2004), and the Brucki et al., 2003; Xu et al., 2003; Castro-Costa et al., 2008).
most likely to initiate help-seeking. A high proportion However, it takes 10 minutes to administer and is prone to
of people with dementia in LMIC live in multigen- educational and cultural bias (Black et al., 1992; Ng et al.,
erational households with young children, and their 2007). A systematic review of 16 brief screening assess-
carers will have divided responsibilities. Children ments identified three (the General Practitioner Assessment
will be future carers of older parents, and future older of Cognition, Mini-Cog, and Memory Impairment Screen),
people. They are receptive, and easily accessed through taking less than five minutes to administer, that were con-
schools. sidered suitable and valid for routine use in general practice,
4. The provision of disability pensions and carer ben- but they have not been validated in poorer settings (Brodaty
efits will inevitably increase requests for diagnostic et al., 2006).
assessment. In high-income countries, diagnosis would typically
5. Increasingly the voices of people who have been be carried out by specialist dementia services, not widely
diagnosed with dementia are being heard, either singly available in LMIC. The 10/66 Dementia Research Group’s
(Taylor, 2009) or collectively (Group SDW, 2009). culture and education-fair diagnostic protocol is validated
By describing their experiences of how it feels to be across many LMIC countries, cultures and languages
diagnosed with dementia and their need for support- (Prince et al., 2003; Liu et al., 2005), but would need adap-
ive services, they are challenging the stigma associated tation for routine clinical use. Good practice guidelines
with dementia and the lack of public understanding of (National Collaborating Centre for Mental Health, 2007)
dementia. advocate a ‘dementia screen’ to exclude treatable causes
of dementia; haematology, biochemistry (electrolytes, cal-
cium, glucose, renal and liver function), thyroid function
tests, serum vitamin B12 and folate levels are routinely rec-
38.2.2 IMPROVING THE CAPACITY OF ommended, testing for syphilis or HIV is indicated only
EXISTING SERVICES for those at risk. Feasibility and cost effectiveness needs to
be tested in LMIC.
38.2.2.1 Recognition and diagnosis Disclosure of dementia diagnosis is considered by pri-
mary care physicians the most difficult area in dementia
Dementia is a hidden problem, especially in LMICs, with management (Bamford et al., 2004). A recent systematic
little help-seeking despite the high burden it poses to review identified eight domains of good practice for dis-
carers and families. Dementia is in many settings seen closing dementia diagnosis – preparation; integrating fam-
as ‘normal’ ageing and there is reluctance from family, ily members; exploring the patient’s perspective; disclosing
patients and health workers to dementia diagnosis as it the diagnosis; responding to patient reactions; focusing on
carries an important burden of stigma. However, recogni- quality of life and well-being; planning for the future; and
tion is the first step in enhancing the capacity of family communicating effectively (Lecouturier et al., 2008) The
carers to look after their relative with dementia, and to person being assessed should be asked if they, and/or oth-
empower them in their interaction with service provid- ers wish to be told the diagnosis. They should then be pro-
ers. It can be boosted by community dissemination of vided with information about the signs and symptoms of
information from government, healthcare providers and dementia, course and prognosis, available treatments, local
394 Dementia

care and support services. Medicolegal issues may be dis- risk factors were eliminated, then a total of up to 28.2%
cussed, including financial and testamentary capacity and of all cases of AD worldwide might be prevented. This is
driving safety. implausible – a more realistic 10% reduction in the preva-
lence of the risk exposures would lead to an 8% reduction
in the prevalence of dementia through to 2050, while a
38.2.2.2 Prevention 20% reduction in exposure prevalence would lead to a 15%
reduction in dementia prevalence (Norton et al., 2014).
Emerging evidence, based mainly upon systematic Prevention could in principle be highly cost effective,
reviews and meta-analyses of observational epidemiologi- considering the high cost attached to the care and treatment
cal data supports the notion that the future scale of the of those with dementia. With changing lifestyles, expo-
dementia epidemic may be reduced by action taken now sure levels to cardiovascular disease risk factors and rates
to reduce exposure to modifiable risk factors. In January of cardiovascular disease are rising rapidly in many LMICs
2014, a meeting in Blackfriars, London, United Kingdom (Prince et al., 2015).
was hosted by the UK Health Forum and Public Health In comparison with the situation in most high-income
England to discuss the relationship between risk factors countries, efforts to prevent and control the coming epi-
for dementia and cardiovascular diseases. The result- demic of cardiovascular and other chronic diseases in
ing ‘Blackfriars Consensus’ statement was published in most of the LMICs are in their infancy (Epping-Jordan
the Lancet (Lincoln et al., 2014), and is supported by 60 et al., 2005). Advocated measures include the implementa-
experts and organizations in dementia and non-commu- tion of tobacco-free policies, taxation of tobacco products,
nicable diseases (NCD) prevention. The statement includes comprehensive bans on advertising of tobacco products,
the recommendation that salt reduction through voluntary agreements with the food
industry, reduction in alcohol intake, increases in activ-
Recent evidence suggests that risk in the popu- ity levels and combination drug therapy for those at high
lation might be reduced so that fewer people at risk of cardiovascular disease (Beaglehole et al., 2012). The
particular ages develop dementia. The scientific detection and control of hypertension, hyperlipidaemia,
evidence is evolving rapidly and sufficient to jus- diabetes and metabolic syndrome is poorly implemented
tify considered action and further research on by overstretched primary care services that struggle to
dementia risk reduction, both by reducing the cope with the double burden of historic priorities (mater-
modifiable risk factors and improving the recog- nal, child and communicable diseases) and the increase of
nised protective factors. chronic disease in adults.

In September 2014, ADI launched the World Alzheimer 38.2.2.3 Improving treatment


Report 2014 – Dementia and Risk Reduction: An Analysis
of Modifiable Risk Factors (Prince et al., 2014). The report The 10/66 Dementia Research Group’s carer pilot study
systematically reviews the evidence in four domains: devel- conducted in several developing countries (World Health
opmental, psychological and psychosocial, lifestyle and Organization, 2006) indicated that the people with demen-
cardiovascular risk factors. The conclusion is that there is tia were using primary and secondary care health services.
particularly robust evidence that low education, hyperten- Only 33% of people with dementia in India, 11% in China
sion in midlife and diabetes and smoking from mid- to late- and South East Asia and 18% in Latin America had used
life increase the risk of developing dementia. no health services at all in the previous 3 months. There
A recent systematic review of the epidemiological evi- was also a heavy use of private medical services, which
dence for AD risk reduction focusing on seven risk fac- might be a reflection of the carer’s perception of the unre-
tors for which there was evidence of independent effects sponsiveness of services provided by the government.
on AD incidence; diabetes, midlife hypertension and Many developing countries have in place a comprehensive
obesity, depression, physical inactivity, smoking and low community-based primary care system staffed by doctors,
education; assessing evidence pertaining to the United nurses and generic multipurpose health workers, yet pri-
States and the population worldwide (Norton et al., 2014). mary healthcare services in LMIC fail older people with
Relative risks were combined with the prevalence of the dementia as they are clinic-based and preoccupied with
risk factor in the population to compute a population simple curative interventions, orientated towards acute
attributable risk (PAR) – the proportion of cases of AD in rather than chronic conditions (Patel and Prince, 2001;
the population that might be prevented if the risk factor Shaji et al., 2002; Dias et al., 2004; Prince et al., 2007). A
could be removed entirely. From the worldwide perspec- paradigm shift is needed to encompass continuing care
tive, the most promising strategies for prevention were and support as part of a wider chronic disease strategy.
the elimination of physical inactivity (12.7% of AD cases Given the frailty of many older people with dementia,
prevented), smoking (13.9%) and low education (19.1%). there is also a need for outreach, assessing and managing
This is because these factors are both relatively common patients in their own homes. The WHO Innovative Care for
and strongly associated with incident AD. If all of the Chronic Conditions (ICCC) framework (Epping-Jordan
Global challenge of dementia: What can be done? 395

et al., 2004) makes no mention of dementia, but the princi- of any attempt to increase coverage of services for people
ples are highly applicable; a dialogue to build commitment with dementia in resource poor settings (Lund et al., 2012;
for change; extended, regular healthcare contact; a multi- Hanlon et al., 2014). These non-specialist health workers
sectoral approach; centring care on patients and families, would be trained to deliver evidence-based ‘packages of
supporting patients in the community; and an emphasis care’. Packages of care for people with dementia (Prince
on prevention. Dementia care should be an essential com- et al., 2009) (and other priority mental and neurologi-
ponent of chronic disease care strategies. cal disorders, including depression) have been developed
Needs for care and support differ for each families, and by WHO as part of their Mental Health Gap Action
ways to make care more person-centred need to be found, Programme (mhGAP, http://www.who.int/mental_health/
packages of care should be more flexible and individual- mhgap/). However, as yet, these have not been taken up,
ized. Families need to be guided and supported in accessing implemented or evaluated for older people. Such single
information and exercising choice, with case managers who condition, vertical programmes have limited appeal to pol-
can provide continuity and advocacy from early stages and icymakers who struggle to provide equitable access even to
across the disease progression (Prince et al., 2013c). basic age-appropriate medical care for frail and dependent
older people. Structural barriers to progress include health
38.2.2.4 Reducing costs of treatment systems tailored to the provision of acute rather than con-
tinuing care, out-of-pocket user fees that mount up with
Costs of cholinesterase inhibitors (ChEIs) are now reim- chronic disease, and staff who are neither trained nor
bursed in many middle-income countries, and prices have experienced in holistic needs-based assessment of older
fallen as agents come off patent protection and cheaper clients (Albanese et al., 2011). Crucially, access to services
generics become available. This is an active field for drug depends upon the ability to attend a primary care facility,
development, particularly following the launch of the G7 often at some distance from the home.
Global Action Against Dementia, with the stated the aim Since dementia is very often co-morbid with depres-
of finding a disease modifying treatment or cure by 2025 sion and physical impairments (including undernutrition,
(RDD/10495, 2013). Any newly developed and effective anti- reduced mobility, pain, hearing and vision problems, and
dementia drugs are likely to be very expensive, and scaling incontinence) (Prince et al., 2011), horizontally struc-
up coverage globally, in an equitable way, would raise huge tured comprehensive assessment and intervention, target-
ethical and practical challenges, most particularly to ensure ing frailty and dependence may be the best and simplest
access to the two-thirds of people with dementia in LMIC. approach to increase coverage of care for older people with
The challenges would be similar in many ways to those dementia and mood disorders. A new WHO evidence-based
addressed by the Global Fund and President’s Emergency clinical guideline, WHO-COPE (Care for Older People)
Plan for AIDS Relief (PEPFAR) initiatives for HIV/AIDS. is currently being developed by the WHO Department of
This would, of necessity, include strengthening the capac- Ageing and Life Course to address this need (http://www.
ity of primary care to diagnose dementia and deliver treat- who.int/ageing/about/who_activities/en/index1.html).
ment and care, as part of wider improvements in primary WHO-COPE comprises: (1) community case finding of
healthcare for older people (World Health Organization frail dependent older adults by CHWs; (2) structured com-
Maximizing Positive Synergies Collaborative Group et al., prehensive home-based assessment of mental, cognitive and
2009). Costs of primary care and community interven- physical impairments, care arrangements and carer strain;
tions can be minimized through task shifting to non- (3) evidence-based interventions for low mood, confusion,
specialist and paraprofessional staff and integration with behaviour disturbance, undernutrition, reduced mobility,
more broadly based chronic disease (Epping-Jordan et al., incontinence, low vision and hearing problems.
2004), disability and elder care programmes (World Health Wider adoption of packages of care, including invest-
Organization, 2004; United Nations, 2002). ment and scaling up will only occur with political will
supported by effective dissemination of evidence on cost
effectiveness, detailed manualization, and information
38.2.3 DELIVERING EFFICACIOUS on planning and resource implications for implementa-
TREATMENT tion. Cultural factors will impact on care arrangements,
and how these are best supported, and on the design of
38.2.3.1 General principles to promote some intervention components to enhance acceptability.
scaling up coverage of dementia Health system factors, particularly human resources and
care regulation will impact on feasibility, with different levels
and types of CHW cadres each with their own capacities
In resource-poor countries with few specialists, as for and training needs. It will thus be necessary to test feasi-
other mental and neurological priority conditions, task bility, acceptability and effectiveness in a range of settings,
shifting and sharing to trained and supported non-spe- making necessary culture and system-specific adaptations
cialist health workers is likely to be an essential feature before full implementation.
396 Dementia

Some of the key features of packages of care for people sound research before any firm conclusion can be drawn.
with dementia are outlined in the following text. Day care and residential care are often more expensive than
targeted home care supplementing family support and care,
and unlikely to be a priority for government investment,
38.2.3.2 Practice-based programmes when the housing conditions of the general population are
poor. Nevertheless, even in some of the poorest developing
Although many LMIC aim to have in place comprehen- countries (e.g. China and India), nursing and residential
sive community-based primary care system staffed by care homes are opening up in the private sector to meet
doctors, nurses and generic multipurpose health workers, the demand from the growing affluent middle class. Good
most have insufficient specialists (psychiatrists, neurolo- quality, well-regulated residential care has a role to play
gists, elderly care physicians) dedicated to dementia care in all societies, for those with no family support, and for
to provide national front line dementia services (Prince those where family support capacity is exhausted, both as
et al., 2009). Initial diagnosis and needs assessment can temporary respite and for provision of longer-term care.
be conducted in primary care, although specialist input Nevertheless over investment in residential care reduces
would be desirable. A commitment to continuing care is the capacity to develop community- and home-based sup-
essential, with regular review, coupled with updated and port services, especially in settings with poor resources. It is
holistic assessments of health and social care needs. Case therefore important to balance the development of residen-
managers can coordinate this process; this collaborative tial care and community services for people with dementia.
care model has been shown to enhance person-centred Absence of regulation, staff training and quality assurance
care, improve outcomes for people with dementia and
is a serious concern in developed and developing countries
reduce inefficient service use, in some but not all studies,
alike. Important priorities would include a system of regis-
conducted exclusively in high-income country settings
tration and inspection of homes, training of care workers
(Prince et al., 2013c). Specialists, where available, can help
and provision of medical services for residents to create the
with advice, inpatient or outpatient review of refractory
best possible quality care.
behavioural and psychotic symptoms of dementia (BPSD)
Family carers and paid caregivers share much in
and respite care.
common, carrying out difficult, demanding and socially
useful roles, with minimal training and preparation.
Paid carers are usually paid around the minimum wage
and experience low job satisfaction while caregivers are
38.2.3.3 Community-based programmes paid nothing and often experience out-of-pocket costs.
Undervaluing caregivers have a negative impact on qual-
Those with dementia are mostly cared for by their families,
ity of care. All caregivers should be valued and recog-
especially in LMIC. The 10/66 pilot study (World Health
nized, and recompensed appropriately: incentives to
Organization, 2006) shows that levels of caregiver strain and
psychological morbidity are at least as high as in the devel- encourage family to provide care at home, skills devel-
oped world. Carer support programmes are essential for the opment and career progression among paid care workers
management of dementia, and can be delivered by trained (Prince et al., 2013c).
CHWs or peer-to-peer by more experienced carers. Carer Table 38.2 shows ADI’s vision for the pattern of demen-
strain should trigger more intensive intervention compris- tia service development (Alzheimer’s Disease International,
ing, as indicated, psychological assessment and treatment 2009). It is based on the graduated approach to service
for the carer, respite, and carer education and training. In development (see Table 38.1) and recognizes that progress
practice, such interventions, especially in LMIC, may be will depend on the resources available.
usefully incorporated into horizontally constructed com- We began by stating that the healthcare needs of older
munity-based programmes addressing the generic needs people have been under-prioritized. The full implications
of frail, dependent older people and their carers, whether of demographic ageing, the prevalence of dementia and
arising from cognitive, mental or physical disorders (Prince their impact on both people with dementia and health-
et al., 2009). care systems are only just beginning to be recognized.
Respite care might be an important level of care, as it Nevertheless, there are grounds for optimism: some gov-
aims to decrease caregiver burden and contribute to ernments have made dementia a national health priority
increase the time the person with dementia can live at and we have shown that it is possible for LMICs to take
home. Despite the satisfaction manifested by carers, there is steps to improve dementia care through primary care
no clear evidence on its benefits. A recent systematic review and support for people with chronic illnesses. Now is the
(Lee and Cameron, 2004) that included three randomized time for governments to act and learn from their emerg-
control trials found no evidence of efficacy of respite care ing experiences. Future research will play an important
for people with dementia or for their caregivers. However, role in evaluating new dementia initiatives and filling
the authors found methodological problems in the trials our gaps in knowledge about how to prevent and treat
and suggest that there is a need for more methodologically this disease.
Global challenge of dementia: What can be done? 397

Table 38.2 Alzheimer’s Disease International’s vision for the pattern of dementia service development
LMICs High-income countries
Awareness and Public education – dementia as an illness, not an Public education – dementia as an illness, not an
understanding inevitable part of old age; where to go for help inevitable part of old age; where to go for help
and information and information
Risk reduction as part of general health Risk reduction as part of general health promotion
promotion Campaigns for early help-seeking and recognition
Campaigns against stigma and discrimination of dementia
Campaigns against stigma and discrimination
Capacity Integrate dementia awareness and understanding National plans for dementia services, from
building into primary healthcare planning diagnosis to palliative care
Education/training for healthcare professionals, Budgets specifically for dementia
auxiliary health workers and care workers Education/training for healthcare professionals,
Information and training for family carers auxiliary health workers and care workers
Integrate dementia into curricula Information and training for family carers
Integrate dementia into curricula for healthcare
professionals
Services Detection and diagnosis in primary healthcare Detection and diagnosis shared between primary
Information, advice and support for family carers and secondary healthcare
through auxiliary health workers Treatment of cognitive impairment with ChEIs
Treatment of cognitive impairment with ChEIs Post-diagnosis support for people with dementia
where affordable Information, advice and support for family carers
Care workers employed by families of people through self-help groups and specialist
with dementia dementia workers
Advice and support from primary care if BPSD Community-based services for people with
become problematic dementia to provide stimulation and help
maintain skills while providing respite for family
carers
Extra support for people with dementia
experiencing BPSD including secondary medical
care
Continuing care in the person with dementia’s
own home for as long as possible
High-quality care homes for people with dementia
no longer able to care for themselves in their
own homes
End of life palliative care
Source: Alzheimer’s Disease International, World Alzheimer Report 2009, Alzheimer’s Disease International, London, 2009.
Abbreviations: BPSD, behavioural and psychological symptoms of dementia; ChEIs, cholinesterase inhibitors; LMICs, low- and middle-income
countries.

REFERENCES Barnes, D.E. and Yaffe, K. (2011). The projected effect of


risk factor reduction on Alzheimer’s disease preva-
Albanese, E., Liu, Z., Acosta, D. et al. (2011). Equity in the lence. The Lancet Neurology, 10 (9): 819–828.
delivery of community healthcare to older people: Beaglehole, R., Bonita, R., Horton, R. et al. (2012).
Findings from 10/66 Dementia Research Group cross- Measuring progress on NCDs: One goal and five tar-
sectional surveys in Latin America, China, India and gets. The Lancet, 380 (9850): 1283–1285.
Nigeria. BMC Health Services Research, 11: 153. Bird, H.R., Canino, G., Stipec, M.R. and Shrout, P.
Alzheimer’s Disease International (2009). World Alzheimer (1987). Use of the mini-mental state examination in
Report 2009. London: Alzheimer’s Disease International. a probability sample of a Hispanic population. The
Anand, S.S. and Yusuf, S. (2011). Stemming the global Journal of Nervous and Mental Disease, 175 (12):
tsunami of cardiovascular disease. The Lancet, 377 731–737.
(9765): 529–532. Black, S.A., Espino, D.V., Mahurin, R. et al. (1992). The
Bamford, C., Lamont, S., Eccles, M. et al. (2004). Disclosing a influence of noncognitive factors on the mini-mental
diagnosis of dementia: A systematic review. International state examination in older Mexican-Americans:
Journal of Geriatric Psychiatry, 19 (2): 151–169. Findings from the Hispanic EPESE. Established
398 Dementia

Population for the Epidemiologic Study of the Fernandez, M., Castro-Flores, J., Perez-de las, H.S. et al.
Elderly. Journal of Clinical Epidemiology, 52 (11): (2008). Prevalence of dementia in the elderly aged
1095–1102. above 65 in a district in the Basque Country. Revista
Brayne, C., Fox, C. and Boustani, M. (2007). Dementia de Neurologia, 46 (2): 89–96.
screening in primary care: Is it time? JAMA, 298 Ferri, C.P., Prince, M., Brayne, C. et al. (2005). Global
(20): 2409–2411. prevalence of dementia: A Delphi consensus study.
Brodaty, H., Low, L.F., Gibson, L. and Burns, K. (2006). The Lancet, 366 (9503): 2112–2117.
What is the best dementia screening instrument for Fitzpatrick, A.L., Kuller, L.H., Lopez, O.L. et al. (2005).
general practitioners to use? American Journal of Survival following dementia onset: Alzheimer’s disease
Geriatric Psychiatry, 14 (5): 391–400. and vascular dementia. Journal of the Neurological
Brucki, S.M., Nitrini, R., Caramelli, P. et al. (2003). Sciences, 229: 43–49.
Suggestions for utilization of the mini-mental state Folstein, M.F., Folstein, S.E. and McHugh, P.R. (1975).
examination in Brazil. Arquivos de Neuro-psiquiatria, ‘Mini-mental state’: A practical method for grading the
61 (3B): 777–781. cognitive state of patients for the clinician. Journal of
Castro-Costa, E., Fuzikawa, C., Uchoa, E. et al. (2008). Psychiatric Research, 12(3): 189–198.
Norms for the mini-mental state examination: Ford, E.S., Ajani, U.A., Croft, J.B. et al. (2007). Explaining
Adjustment of the cut-off point in population-based the decrease in U.S. deaths from coronary disease,
studies (evidences from the Bambui health aging study). 1980–2000. The New England Journal of Medicine.
Arquivos de Neuro-psiquiatria, 66 (3A): 524–528. 356 (23): 2388–2398.
Chan, K.Y., Wang, W., Wu, J.J. et al. (2013). Epidemiology Fuster, V. and Voute, J. (2005). MDGs: Chronic diseases are
of Alzheimer’s disease and other forms of dementia in not on the agenda. The Lancet, 366 (9496): 1512–1514.
China, 1990–2010: A systematic review and analysis. Ganguli, M., Ratcliff, G., Chandra, V. et al. (1995). A Hindi
The Lancet, 381 (9882): 2016–2023. Version of the MMSE: The development of a cognitive
Choo, W.Y., Low, W.Y., Karina, R. et al. (2003). Social sup- screening instrument for a largely illiterate rural elderly
port and burden among caregivers of patients with population in India. International Journal of Geriatric
dementia in Malaysia. Asia-Pacific Journal of Public Psychiatry, 10(5): 367–377.
Health, 15 (1): 23–29. Gao, S., Ogunniyi, A., Hall, K.S. et al. (eds.) (2014).
Critchley, J., Liu, J., Zhao, D. et al. (2004). Explaining Alzheimer’s Disease Incidence Declined in African
the increase in coronary heart disease mortal- Americans, but not in Yoruba. Copenhagen: AAIC.
ity in Beijing between 1984 and 1999. Circulation, Gaziano, T.A., Bitton, A., Anand, S. et al. (2010). Growing
110 (10): 1236–1244. epidemic of coronary heart disease in low- and
Dementia: A State of the Nation Report on ­middle-income countries. Current Problems in
Dementia Care and Support in England. Cardiology, 35 (2): 72–115.
London, UK: Department of Health. Graham, N. and Brodaty, H. (1997). Alzheimer’s dis-
Dias, A. and Patel, V. (2009). Closing the treat- ease international. International Journal of Geriatric
ment gap for dementia in India. Indian Journal of Psychiatry, 12 (7): 691–692.
Psychiatry, 51 (Suppl. 1): S93–S97. Group SDW (2009). Scottish Dementia Working Group.
Dias, A., Samuel, R., Patel, V. et al. (2004). The impact Available at http://www.sdwg.org.uk/. Accessed
associated with caring for a person with dementia: 2 October 2009.
A report from the 10/66 Dementia Research Group’s Guerchet, M., Houinato, D., Paraiso, M.N. et al. (2009).
Indian network. International Journal of Geriatric Cognitive impairment and dementia in elderly people
Psychiatry, 19 (2): 182–184. living in rural Benin, West Africa. Dementia and
Doblhammer, G., Fink, A. and Zylla, S. (eds.) (2014). Geriatric Cognitive Disorders, 27 (1): 34–41.
Short-term Trends in German Dementia Prevalence, Guerchet, M., M’Belesso, P., Mouanga, A.M. et al. (2010).
Incidence, and Mortality. Copenhagen: AAIC. Prevalence of dementia in elderly living in two cities
Epping-Jordan, J.E., Galea, G., Tukuitonga, C. and of Central Africa: The EDAC survey. Dementia and
Beaglehole, R. (2005). Preventing chronic diseases: Taking Geriatric Cognitive Disorders, 30 (3): 261–268.
stepwise action. The Lancet, 366 (9497): 1667–1671. Guerchet, M., M’Belesso, P., Ndamba-Bandzouzi, B. et al.
Epping-Jordan, J.E., Pruitt, S.D., Bengoa, R. and Wagner, (2014). Epidemiology of dementia in Central Africa
E.H. (2004). Improving the quality of health care for (EPIDEMCA): Protocol for a multicentre population-
chronic conditions. Quality and Safety in Health Care, based study in rural and urban areas of the Central
13 (4): 299–305. African Republic and the Republic of Congo.
Feigin, V.L., Lawes, C.M., Bennett, D.A. et al. (2009). SpringerPlus, 3: 338.
Worldwide stroke incidence and early case fatality Gupta, R., Joshi, P., Mohan, V. et al. (2008). Epidemiology
reported in 56 population-based studies: A systematic and causation of coronary heart disease and stroke in
review. The Lancet Neurology, 8 (4): 355–369. India. Heart, 94 (1): 16–26.
Global challenge of dementia: What can be done? 399

Hanlon, C., Luitel, N.P., Kathree, T. et al. (2014). Lund, C., Tomlinson, M., De Silva, M. et al. (2012). PRIME:
Challenges and opportunities for implementing inte- A programme to reduce the treatment gap for mental
grated mental health care: A district level situation disorders in five low- and middle-income countries.
analysis from five low- and middle-income countries. PLoS Medicine, 9 (12): e1001359.
PloS One, 9 (2): e88437. Mathillas, J., Lovheim, H. and Gustafson, Y. (2011).
Hendrie, H.C., Osuntokun, B.O., Hall, K.S. et al. (1995). Increasing prevalence of dementia among very old
Prevalence of Alzheimer’s disease and dementia people. Age and Ageing, 40 (2): 243–249.
in two communities: Nigerian Africans and African Matthews, F.E., Arthur, A., Barnes, L.E. et al. (2013). A
Americans. American Journal of Psychiatry, 152 two-decade comparison of prevalence of dementia in
(10): 1485–1492. individuals aged 65 years and older from three geo-
Honyashiki, M., Ferri, C.P., Acosta, D. et al. (2011). graphical areas of England: Results of the Cognitive
Chronic diseases among older people and co- Function and Ageing Study I and II. The Lancet,
resident psychological morbidity: A 10/66 382 (9902): 1405–1412.
Dementia Research Group population-based Murray, C.J., Ezzati, M., Flaxman, A.D. et al. (2012a). GBD
­survey. International Psychogeriatrics, 23 2010: Design, definitions, and metrics. The Lancet, 380
(9): 1489–1501. (9859): 2063–2066.
Laatikainen, T., Critchley, J., Vartiainen, E. et al. (2005). Murray, C.J., Vos, T., Lozano, R. et al. (2012b). Disability-
Explaining the decline in coronary heart disease adjusted life years (DALYs) for 291 diseases and injuries
mortality in Finland between 1982 and 1997. American in 21 regions, 1990–2010: A systematic analysis for the
Journal of Epidemiology, 162 (8): 764–773. Global Burden of Disease Study 2010. The Lancet, 380
Langa, K.M., Larson, E.B., Karlawish, J.H. et al. (2008). (9859): 2197–2223.
Trends in the prevalence and mortality of cognitive Nakamura, A.E., Opaleye, D., Tani, G. and Ferri,
impairment in the United States: Is there evidence of a C.P. (2015). Dementia underdiagnosis in Brazil. The
compression of cognitive morbidity? Alzheimer’s and Lancet, 385 (9966): 418–419.
Dementia: The Journal of the Alzheimer’s Association, National Collaborating Centre for Mental Health (2007).
4 (2): 134–144. Dementia. A NICE-SCIE Guideline on supporting peo-
Lecouturier, J., Bamford, C., Hughes, J.C. et al. (2008). ple with dementia and their carers in health and social
Appropriate disclosure of a diagnosis of dementia: care – National Clinical Practice Guideline Number 42.
Identifying the key behaviours of ‘best practice’. BMC Leicester,UK: The British Psychological Society.
Health Services Research, 8: 95. Norton, S., Matthews, F.E., Barnes, D.E. et al. (2014).
Lee, H. and Cameron, M. (2004). Respite care for peo- Potential for primary prevention of Alzheimer’s
ple with dementia and their carers. The Cochrane disease: An analysis of population-based data. The
Database of Systematic Reviews, 2: CD004396. Lancet Neurology, 13 (8): 788–794.
Lobo, A., Saz, P., Marcos, G. et al. (2007). Prevalence of Ng, T.P., Niti, M., Chiam, P.C. and Kua, E.H. (2007). Ethnic
dementia in a southern European population in two and educational differences in cognitive test perfor-
different time periods: The ZARADEMP Project. Acta mance on mini-mental state examination in Asians.
Psychiatrica Scandinavica, 116 (4): 299–307. American Journal of Geriatric Psychiatry, 15 (2): 130–139.
Loef, M. and Walach, H. (2012). Midlife obesity and Paraiso, M.N., Guerchet, M., Saizonou, J. et al. (2011).
dementia: Meta-analysis and adjusted forecast of Prevalence of dementia among elderly people living
dementia prevalence in the US and China. Obesity, in Cotonou, an urban area of Benin (West Africa).
21 (1): E51–E55. Neuroepidemiology, 36 (4): 245–251.
Longdon, A.R., Paddick, S.M., Kisoli, A. et al. (2013). The Patel, V. and Prince, M. (2001). Ageing and mental health
prevalence of dementia in rural Tanzania: A cross‑­ in a developing country: Who cares? Qualitative studies
sectional community-based study. International from Goa, India. Psychological Medicine, 31 (1): 29–38.
Journal of Geriatric Psychiatry, 28 (7): 728–737. Plassman, B.L., Langa, K.M., Fisher, G.G. et al. (2007).
Lopez, A.D., Williams, T.N., Levin, A. et al. (2014). Prevalence of dementia in the United States:
Remembering the forgotten non-communicable dis- The aging, demographics, and memory study.
eases. BMC Medicine, 12 (1): 200. Neuroepidemiology, 29 (1–2): 125–132.
Lincoln, P., Fenton, K., Alessi, C. et al. (2014). The Prince, M. (2004). Care arrangements for people with
Blackfriars Consensus on brain health and dementia. dementia in developing countries. International
The Lancet, 383 (9931): 1805–1806. Journal of Geriatric Psychiatry, 19 (2): 170–177.
Liu, S.I., Prince, M., Chiu, M.J. et al. (2005). Validity and Prince, M., Acosta, D., Albanese, E. et al. (2008). Ageing
reliability of a Taiwan Chinese version of the com- and dementia in low and middle income countries-
munity screening instrument for dementia. American Using research to engage with public and policy mak-
Journal of Geriatric Psychiatry, 13 (7): 581–588. ers. International Review of Psychiatry, 20 (4): 332–343.
400 Dementia

Prince, M.J., Acosta, D., Castro-Costa, E. et al. (2009). RDD/10495 (2013). G8 Summit Declaration 2013. Available
Packages of care for dementia in low- and middle- at https://www.gov.uk/government/publications/
income countries. PLoS Medicine, 6 (11): e1000176. g8-dementia-summit-agreements/g8-dementia-
Prince, M., Acosta, D., Chiu, H. et al. (2003). Dementia summit-declaration. Accessed 11 March 2015.
diagnosis in developing countries: A cross-cultural Rocca, W.A., Petersen, R.C., Knopman, D.S. et al. (2011).
validation study. The Lancet, 361 (9361): 909–917. Trends in the incidence and prevalence of Alzheimer’s
Prince, M., Acosta, D., Ferri, C.P. et al. (2011a). The asso- disease, dementia, and cognitive impairment in the
ciation between common physical impairments and United States. Alzheimer’s and Dementia: The Journal
dementia in low and middle income countries, and, of the Alzheimer’s Association, 7 (1): 80–93.
among people with dementia, their association with Salomon, J.A., Vos, T., Hogan, D.R. et al. (2012). Common
cognitive function and disability. A 10/66 Dementia values in assessing health outcomes from disease and
Research Group population-based study. International injury: Disability weights measurement study for the
Journal of Geriatric Psychiatry, 26 (5): 511–519. Global Burden of Disease Study 2010. The Lancet, 380
Prince, M., Albanese, E., Guerchet, M. and Prina, (9859): 2129–2143.
M. (2014a). World Alzheimer Report 2014. Dementia Satizabal, C., Beiser, A., Chene, G. et al. (eds.) (2014).
and Risk Reduction. An Analysis of Protective and Temporal trends in Dementia Incidence in the
Modifiable Risk Factors. London: Alzheimer’s Disease Framingham Study. Copenhagen: AAIC.
International. Schrijvers, E.M., Verhaaren, B.F., Koudstaal, P.J. et al.
Prince, M., Brodaty, H., Uwakwe, R. et al. (2012). Strain (2012). Is dementia incidence declining? Trends in
and its correlates among carers of people with dementia incidence since 1990 in the Rotterdam
dementia in low-income and middle-income countries. Study. Neurology, 78 (19): 1456–1463.
A 10/66 Dementia Research Group population-based Sekita, A., Ninomiya, T., Tanizaki, Y. et al. (2010). Trends
survey. International Journal of Geriatric Psychiatry, 27 in prevalence of Alzheimer’s disease and vascular
(7): 670–682. dementia in a Japanese community: The Hisayama
Prince, M., Bryce, R., Albanese, E. et al. (2013a). The Study. Acta Psychiatrica Scandinavica, 122 (4): 319–325.
global prevalence of dementia: A systematic review Shaji, K.S., Arun Kishore, N.R., Lal, K.P. and Prince,
and metaanalysis. Alzheimer’s and Dementia the M. (2002). Revealing a hidden problem. An evaluation of
Journal of the Alzheimer’s Association, 9 (1): 63–75. a community dementia case-finding program from the
Prince, M., Bryce, R. and Ferri, C. (2011b). World Indian 10/66 dementia research network. International
Alzheimer Report 2011: The Benefits of Early Diagnosis Journal of Geriatric Psychiatry, 17 (3): 222–225.
and Intervention. London: Alzheimer’s Disease Shaji, K.S., Smitha, K., Lal, K.P. and Prince, M. (2002).
International. Caregivers of patients with Alzheimer’s disease: A quali-
Prince, M., Guerchet, M., Prina, M. and Alzheimer’s Disease tative study from the Indian 10/66 dementia research
International. (2013b). The Global Impact of Dementia network. International Journal of Geriatric Psychiatry,
2013–2050. Policy Brief for Heads of Governments. 18: 1–6.
London: Alzheimer’s Disease International. Taylor, R. (2009). Alzheimer’s From the Inside Out.
Prince, M., Knapp, M., Guerchet, M. et al. (2014b). Baltimore, MD: Health Profession Press Inc.
Dementia UK: Update, Second Edition. London: United Nations (2002). Report of the Second World
Alzheimer’s Society. Assembly on Ageing. Madrid, 8–12 April, 2002. New
Prince, M., Livingston, G. and Katona, C. (2007). Mental York, NY: United Nations.
health care for the elderly in low-income countries: A Vartiainen, E., Laatikainen, T., Peltonen, M. et al. (2010).
health systems approach. World Psychiatry, 6 (1): 5–13. Thirty-five-year trends in cardiovascular risk factors in
Prince, M., Prina, M., Guerchet, M. and Alzheimer’s Disease Finland. International Journal of Epidemiology, 39 (2):
International. (2013c). World Alzheimer Report 2013. 504–518.
Journey of Caring: Analysis of Long-Term Care for Wimo, A., Jonsson, L., Bond, J. et al. (2013). The world-
Dementia. London: Alzheimer’s Disease International. wide economic impact of dementia 2010. Alzheimer’s
Prince, M.J., Wu, F., Guo, Y. et al. (2015). The burden of and Dementia: The Journal of the Alzheimer’s
disease in older people and implications for health Association, 9 (1): 1–11.
policy and practice. The Lancet, 385 (9967): 549–562. Wimo, A., Prince, M. and Alzheimer’s Disease
Qiu, C., von Strauss, E., Backman, L. et al. (2013). Twenty- International. (2010). World Alzheimer Report 2010:
year changes in dementia occurrence suggest The Global Economic Impact of Dementia. London:
decreasing incidence in central Stockholm, Sweden. Alzheimer’s Disease International.
Neurology, 80 (20): 1888–1894. World Health Organization (1996). The Global Burden of
Ramos-Cerqueira, A.T., Torres, A.R., Crepaldi, A.L. et al. Disease: A Comprehensive Assessment of Mortality and
(2005). Identification of dementia cases in the com- Disability from Diseases, Injuries and Risk Factors in 1990
munity: A Brazilian experience. Journal of American and Projected to 2020. Cambridge, MA: The Harvard
Geriatric Society, 53 (10): 1738–1742. School of Public Health, Harvard University Press.
Global challenge of dementia: What can be done? 401

World Health Organization (2004a). Global Burden of World Population Prospects (2013). The 2012 Revision,
Disease 2004 Update: Disability Weights for Diseases Highlights and Advance Tables. New York, NY:
and Conditions. Geneva: World Health Organization. United Nations, Department of Economic and Social
World Health Organization (2004b). Towards Age- Affairs PD.
Friendly Primary Health Care. Geneva: World Health Wu, Y.T., Lee, H.Y., Norton, S. et al. (2014). Period, birth
Organization. cohort and prevalence of dementia in mainland China,
World Health Organization (2006). Neurological Disorders: Hong Kong and Taiwan: A meta-analysis. International
Public Health Challenge. Geneva: World Health Journal of Geriatric Psychiatry, 29 (12): 1212–1220.
Organization. Xu, G., Meyer, J.S., Huang, Y. et al. (2003). Adapting
World Health Organization (2012). Dementia: A Public mini-mental state examination for dementia screen-
Health Priority. Geneva: World Health Organization. ing among illiterate or minimally educated elderly
World Health Organization Maximizing Positive Chinese. International Journal of Geriatric Psychiatry,
Synergies Collaborative Group, Samb, B., Evans, T. 18 (7): 609–616.
et al. (2009). An assessment of interactions between Yusuf, A.J., Baiyewu, O., Sheikh, T.L. and Shehu, A.U.
global health initiatives and country health systems. (2011). Prevalence of dementia and dementia sub-
The Lancet, 373 (9681): 2137–2169. types among community-dwelling elderly people in
World Population Prospects (2003). The 2002 Revision – northern Nigeria. International Psychogeriatrics, 23
Highlights. New York, NY: United Nations, Department (3): 379–386.
of Economic and Social Affairs PD.
39
The international challenge of dementia

KARIM SAAD

dementia somewhere in the world being diagnosed every 4


39.1 
INTRODUCTION seconds (Alzheimer Disease International, 2013). If we do
nothing, this will increase to an estimated 75.6 million in
It may be over a century since Alois Alzheimer published 2030 and 135.5 million in 2050.
his report ‘about a peculiar disease of the cerebral cor- Although, dementia mainly affects older people, 2%–10%
tex’, yet the global challenges of dementia in general and of all cases start before the age of 65. Furthermore, 62% of this
Alzheimer’s disease (AD) in particular are only just being rising prevalence is occurring in developing countries and
recognized as urgent healthcare priorities. by 2050, this will rise to a staggering 71%. The fastest growth
The last decade has ushered in progressive international in the elderly population is occurring in China, India, their
engagement and acknowledgement although disease-mod- south Asian and western Pacific neighbours and Sub-Saharan
ifying treatments, let alone a cure, remain inaccessible, and African regions. In the next few decades, the global burden of
inaction is not an option. dementia will shift inexorably to poorer countries, particularly,
What follows is a sample of initiatives, driven by global, rapidly developing middle-income countries that are members
continental and/or visionary individuals, organizations and of the G20, but not the Group of Seven countries (G7).
countries, propelled by the rising phenomenon of state-
owned dementia plans.
39.3 GLOBAL ECONOMIC IMPACT

39.2 A GLOBAL PUBLIC HEALTH The worldwide costs of dementia, estimated at $US604 bil-
lion in 2010, amounted to more than 1% of global gross
PRIORITY
domestic product (GDP), varying from 0.24% of GDP in
low-income countries to 1.24% in high-income countries
Alzheimer Disease International (ADI), a worldwide fed- (Alzheimer Disease International, 2010). Based on cur-
eration of Alzheimer associations representing people with rent financial estimates, if dementia care were a country, it
dementia and their families, has been instrumental in fixing would be the world’s eighteenth largest economy, ranking
a global spotlight on AD and other dementias. Established somewhere between Turkey and Indonesia. Social care and
in 1984, ADI now comprises over 80 member organizations. informal care altogether account to over 80% of those costs.
ADI has released five World Alzheimer Reports since
2009, outlining the global plight of people with dementia
and the demography of AD and other dementias both in
developed and developing countries (Prince, 2009; Prince,
39.4 GLOBAL BURDEN OF CARE
Bryce and Ferri, 2011). The 10/66 group, refers to the two-
thirds (66%) of people with dementia in low- to middle- Dementia is one of the main causes of dependence and dis-
income countries and the 10% of population-based research ability in old age. As the world population ages, the tra-
carried out in those regions. ditional system of ‘informal’ care by family, friends and
In 2013, the global prevalence of dementia, described as community will require much greater support (World
an epidemic, was estimated at 44.4 million people with an Health Organization, 2012). Globally, 13% of people aged
incidence of 7.7 million new cases annually; a new case of 60 or above require long-term care, which is predominantly
402
The international challenge of dementia 403

about caring for people with dementia; around half of all individual jurisdiction’s initiatives. This provided an oppor-
older people who need personal care and 80% of older peo- tunity to create a strategic, collaborative and cost effective
ple in nursing homes are living with dementia. response to dementia across Australia.
In 2008, WHO launched the Mental Health Gap Action Australia’s second Framework (Alzheimer’s Australia,
Programme (mhGAP) (World Health Organization, 2008), 2013) builds on the achievements of the first plan and fol-
which has since then included dementia as a priority condi- lows the stages of dementia care most people will experi-
tion. In 2011, the United Nations General Assembly on pre- ence in their journey including: risk reduction, awareness;
vention and control of non-communicable diseases adopted assessment, diagnosis, post-diagnostic support and end-of-
a political declaration acknowledging that the global burden life care.
of non-communicable diseases (NCDs), including AD, con- Alzheimer’s Australia has established useful resources,
stituted a major challenge for development in this century. including support groups, a support hotline and the world’s
first risk reduction ‘Brainy App’ co-developed with Bupa
Health Foundation for smartphones (Brainy App, 2011).
39.5 EMERGENCE OF NATIONAL
DEMENTIA PLANS 39.5.2 ASIA
With a huge ageing population, dementia care is recognized
Stigma and widespread misconceptions mean that many as a major public health challenge in this region. Over half of
people currently living with dementia cannot access a for- the global dementia population will be part of Asia by 2030.
mal diagnosis and therefore, lack access to treatment, care In China alone, where an estimated 9.2 million people
and advance planning. were living with dementia in 2010, determinants of under-
Missed diagnosis rates range from 20% to 50% (Olafsdottir detection reported to be 93.1% (Chan et al., 2013) and
et al., 2000; Valcour et al., 2000; Boustani et al., 2005; Wilkins included low education levels and living in rural areas,
et al., 2007) and the estimates are as high as 93% (Dias and highlighting the need to target these populations in future
Patel, 2009; Chen et al., 2013). The 2012 World Alzheimer public health campaigns (Chen et al., 2013).
Report (Alzheimer Disease International, 2012) called for the In many Asian countries, dementia is regarded as a
creation of high-level governmental plans customized to the shameful illness and carries a stigma that may lead to reluc-
unique culture and demographics of each country. Key rec- tance in seeking diagnosis and treatment.
ommendations include the following:

1. Every country should have a national dementia strat- 39.5.3 JAPAN


egy promoting earlier diagnosis, raising awareness and In 2004, the term for dementia used in Japan was officially
adequate workforce training. changed from chihō (foolish, stupid, absent minded) to
2. All primary care services should have basic compe- ninchishō (cognitive symptoms and syndrome) (Miyamoto
tency in early detection of dementia, making and et al., 2011).
imparting a provisional dementia diagnosis and initial New nomenclature had to be simple, short, devoid of
management of dementia. pejorative connotations, easily understandable and per-
3. Where feasible, networks of specialist diagnostic and tinent in medicine, government and the public spheres. A
treatment centres should be established. public opinion survey subsequently clinched one out of a
possible six replacement names.
Consequently, the last decade marked an unprecedented In 2005, the Alzheimer’s Association of Japan changed
emergence of dementia plans. Australia, England, France, its name to accommodate the new terminology, replacing
Norway, Netherlands, Denmark and Republic of Korea were the words ‘elderly’ (recognizing the increase in young onset
among the first to launch their plans. France, Australia, of dementia) with ‘persons with dementia’ as well as their
Netherlands and Scotland are examples of countries imple- ‘family caregivers’, thus becoming ‘the Association of per-
menting subsequent plans. sons with dementia and their families’.
In many countries, sub-national plans are also in process A ‘Campaign to Understand Dementia and Build
as the most appropriate level of government to plan strategi- Community Networks’ in Japan comprising four major
cally include Australia, Canada, Switzerland and the United projects concentrated on the following:
States.
1. A nationwide initiative to train 1 million supporters
39.5.1 AUSTRALIA for dementia
2. An initiative to build a dementia-friendly community
Australia’s first national dementia plan (2006–2010) focused 3. The development of support groups for people with
on healthcare and support dimensions that can best be dementia and their caregivers
achieved nationally, with the cooperation of the Australian 4. Care management fully involving people with demen-
State and Territory Governments, rather than on any tia and their families
404 Dementia

39.5.4 REPUBLIC OF KOREA 39.5.5.2.1 


THE EU JOINT PROGRAMMING
NEURODEGENERATIVE DISORDERS
A low birth rate, rapidly ageing society, rapid escalation in (JPND)
medical costs and dwindling informal support provided by Launched in 2008, the JPND is an initiative pooling together
family caregivers have led the government to place dementia European research funds and aligning national research
as a national healthcare priority, setting up long-term care programmes. JPND comprises 28 countries, including non-
insurance and in 2010, the declaration of ‘War on Dementia’ EU states such as Canada. As such, it constitutes one of the
took place. largest global research initiatives in the field of neurodegen-
Early diagnosis, a key objective in the Korean plan (Lee, erative disorders. Priorities of the programme include find-
2010), was enabled by expanding public health centres and ing causes, developing cures and improving effective care
increasing the number of trained dementia specialists to structures (EU JPND research, 2014).
6000 by 2012.
39.5.5.2.2 
EUROPEAN INNOVATION PARTNERSHIP
ON ACTIVE AND HEALTHY AGEING (EIP
39.5.5 EUROPE AHA)
An estimated 6.37 million people live with dementia in the Launched in 2011, this collaboration is designed to identify
European Union (EU) costing €105 million per year with promising regional innovations and scale them accordingly
total societal costs per case assessed to be eight times higher to benefit EU countries. The EIP AHA aims to improve the
in Northern Europe than in Eastern Europe (Wimo et al., quality of life of older people in Europe, including those with
2011). Regional variation across European memory clinics cognitive impairment and to increase the average healthy
(Hausner et al., 2010) suggest that Northern European coun- life years of EU citizens by 2 years by the year 2020. The
tries will see the healthiest dementia populations present- partnership brings together key stakeholders in the innova-
ing earlier stages in dementia, with the lowest concomitant tion cycle (end users, public authorities, industry).
prescription of psychotropics. Contrast this with southern
states where people with dementia will have the shortest 39.5.5.2.3 ALZHEIMER COOPERATIVE VALUATION IN
education period, presenting late stages in the disease, with EUROPE (ALCOVE)
least access to cholinesterase inhibitors or the ability to live The ALzheimer COoperative Valuation in Europe (2013)
with family caregivers. Joint Action is a multinational European collaboration
co-financed by the EU Health Programme 2008–2013. Its
39.5.5.1 The Paris declaration 53 recommendations were presented in Paris on 28 March
2013 and are available online, which covers the following:
A century after the description of AD, Alzheimer’s Europe
called upon European countries (Alzheimer Europe, 2006, 39.5.5.2.3.1 Timely diagnosis
2008a) to accord all forms of dementia and offer the politi- ALCOVE defined the factors contributing to under-
cal priority they deserve. diagnosis or late diagnosis, including stigma, variable
Countries including Cyprus, Greece, Malta, Portugal, post-diagnostic support and inadequate training of general
Slovenia and Spain are developing national plans (Alzheimer practitioners in diagnosing dementia and recognizing its
Europe Survey, 2014), with Italy the latest to launch a national early signs.
strategy at the time of writing the report on the survey. Early medical assessment when cognitive changes ensue
Other countries, e.g. Croatia, Estonia, Germany, Latvia, is supported because various conditions can cause cogni-
have no dedicated national strategies, yet dementia is tive impairment, many of which are treatable. Conversely,
included within other policy initiatives. undergoing an assessment for dementia may predispose the
person and their family to face fears of the disease and the
39.5.5.2 The 2009 European Parliament stigma of suffering from the same.
written declaration Early diagnosis should be ‘timely’ to those seeking it
(Brooker et al., 2014), occurring at a point when the person
In 2009, the European Parliament adopted a written decla- and their family are ready to undergo assessment. ALCOVE
ration calling upon the European Commission and Member proposes incremental strategies to support timely and accu-
States to develop a European Action Plan and to collaborate rate diagnosis at local and national levels.
in order to improve early diagnosis and quality of life for Case finding is supported to identify people to increase
people with dementia and their caregivers. The European detection rates in circumstances where there are services
Commission responded with a commitment to support available that will benefit the person at risk and their fam-
member states in ensuring effective and efficient recogni- ily. As such, targeted or opportunistic case finding within
tion, prevention, diagnosis, treatment, care and research primary care, acute hospitals or care homes should be
for dementia. Consequently, three initiatives were funded, undertaken and steps taken to ensure that adequate post-
which are as follows: diagnostic services and support is available.
The international challenge of dementia 405

General population screening is not recommended until particular, psycho-educational programmes and multicom-
there is better evidence of the reliability of screening along- ponent interventions.
side ways of preventing or delaying dementia. Guidelines on BPSD management made available to
A consensus is required on how early cognitive changes patients, family caregivers and the workforce providing
(e.g. mild cognitive impairment) are to be managed in clini- care for people with dementia are integral to quality and
cal practice. Meanwhile, when people are informed that safety programmes.
they may have early cognitive changes, advice and support
should be given alongside clear pathways for monitoring 39.5.5.2.3.4 Ethics
and subsequent follow-up of the symptoms.
ALCOVE upholds the principle of respecting the rights of
39.5.5.2.3.2 Epidemiology people with dementia, including balancing autonomy and
protection. To achieve this, recommendations were identi-
ALCOVE reviews the prevalence rate for dementia in
fied for the assessment of competence (including four mod-
Europe, using the same terms that the EuroCoDe proj-
els for competence assessments) and the drawing up and use
ect (2006–2008) had adopted for systematic reviews
of advance directives.
(Alzheimer Europe, 2008). On the basis of the highest
A second ALCOVE joint action, commencing March
quality epidemiological studies, ALCOVE estimated a
2015 for 3 years included a focus on post-diagnostic sup-
new European dementia prevalence rate of 7.23% in the
port, improvement of care pathways, safe prescribing, care-
population aged 65 years or older, corresponding to an
giver support, workforce skills and quality residential care.
estimated number of 6.37 million cases (EU-27 Countries,
2011). This estimate was 22% lower than earlier estimates
from EuroCoDe (prevalence rate: 9.28%, 8.3 million 39.5.6 FRANCE
cases). Based on this, ALCOVE identified recommenda-
tions for future data collection on estimates of prevalence France became the first European country to launch a
of dementia in Europe. national dementia plan (2001–2005) to address the rising
ALCOVE estimates that antipsychotics are overpre- prevalence of dementia, of which half its cases remained
scribed for 35.6% of people with dementia residing in undiagnosed. AD is now recognized as a chronic disease by
European nursing homes (ranging from 25.8% in Norway the French social security system. The second French Plan
to 60% in Italy) compared to an estimated 10.6% of the gen- concluded in 2007.
eral elderly population. The United Kingdom, Sweden and President Nicolas Sarkozy launched the third French
France have carried out successful campaigns to reduce Alzheimer plan in 2008 at the arrival of the recession and
exposure to antipsychotics. commented that dementia cannot wait for the worst eco-
Furthermore, overuse of antidepressants, prolonged use nomic crises (Alzheimer’s Europe 2008b). Even doing very
of antipsychotics, concomitant use (two antipsychotics or little is not an option and thereby, 1.6 billion of public funds
with other psychotropics, e.g. hypnotics) or absence of use pledged to a 5-year French plan that sought to understand,
as second-line after a non-pharmacological approach, are cure and care.
also reported. Specialists (geriatricians or neurologists) were given the
The Joint Action underscored the global safety and task to diagnose dementia in France. Service improvements
ethical concerns resulting from overprescribing of antipsy- included a reduced average waiting time for diagnosis and sub-
chotics for behaviour that is one of the biggest challenges sequent access to a simplified personalized integrated demen-
in dementia and makes a number of recommendations tia pathway delivered via an expansion of memory units with
to prevent and reduce inappropriate prescribing, includ- dedicated case managers. Quality improvements included
ing an online toolbox created as a resource to identify risk reduced hospitalization, reduced exposure to antipsychotic
exposure and reduction measures, linking safer prescrib- drugs and specialist cognitive rehabilitation to improve
ing to timely diagnosis, ethical principles underpinning autonomy at home and enhance quality of life post-diagnosis.
prescribing and sharing European programmes to prevent Support for people with dementia in crisis (behavioural units
and manage behavioural and psychological symptoms of in hospital) reported that over a half of the population were
dementia (BPSD). successfully discharged back home and 18,000 caregivers
received education (2-day training programmes) to improve
39.5.5.2.3.3 Strategy for BPSD quality of life and reduce institutionalization.
ALCOVE reviewed all intervention models for BPSD man- National centres for early onset of dementia (in Rouen,
agement at home, in hospital and in care homes, including Lille and Paris) were created to focus research, improve
ambulatory structures and care organizations to prevent awareness and improve care environments.
and manage BPSD. Evaluations of the French plan’s impact on research
Psychosocial individualized interventions for people (Ankri and Broeckhoven, 2013; Devos et al., 2014) suggested
with dementia and their carers were found to be effective a link between national funding and scientific research
in preventing BPSD and delaying institutionalization, in output.
406 Dementia

In November 2014, France issued a Fourth National Plan of the greatest enemies of humanity’, set out to address three
incorporating dementia with other chronic neurodegenera- challenges, which are as follows:
tive diseases (Stratégie National de Sante, 2014).
1. Driving improvements in health and care
39.5.7 THE NETHERLANDS 2. Creating dementia-friendly communities
3. Improving dementia research
The 3-year Dutch Dementia Care Plan was announced
in 2008 outlining three key aims: creating a coordinated By 2014, improvements were evident including the fol-
range of personalized care options, delivering sufficient lowing advancements (Annual Report of Progress, 2013):
guidance and support for people with dementia and
their carers, and measuring the quality of dementia care 1. A scheme enabling towns, cities and villages across
annually. England to become dementia-friendly environments,
The plan called for easily accessible information, sup- where people with dementia are empowered to have
ported by Alzheimer Nederland, to facilitate identifica- high aspirations and feel confident, knowing they can
tion at an individual level. Municipalities under the Social contribute and participate in personally meaningful
Support Act played a crucial role in policy development for activities.
early-detection of dementia. For every person with dementia who is able to live at
General practitioners (GPs) regarded as pivotal in achiev- home within a dementia-friendly community rather
ing earlier diagnosis, were to be knowledgeable in detecting than in residential care, an evaluation of such com-
early symptoms, supported by financial incentives involved munities (Green and Lakey, 2013) estimated savings
in diagnosing dementia. of £11,296 per year or £941 per month. Delaying 5%
In 2013 Dementia Care Standards, financed by insur- admissions to care homes for 1 year was estimated to
ers, reflected a steady increase in service user satisfaction. save the United Kingdom £55 million annually.
Deltaplan Dementia, a second 4-year plan, with public/pri- At the time of writing, over 70 communities across
vate funding is built on three pillars, which are as follows: England had registered to become ‘Dementia-Friendly’
and over 500,000 ‘Dementia Friends’ were trained.
1. Developing an e-health portal Other innovations include a dementia education pio-
2. Establishing a national dementia registry neer programme launched in schools across England
3. Investing in and implementing new programmes for (Saad, 2012) educating young people about demen-
scientific research tia, its causes and the challenges faced by caregivers.
Outputs and resources from this initiative are driv-
39.5.8 NORWAY ing a campaign to spread intergenerational exchange
opportunities across England (Department of Health,
Regarded as the best place to grow older in the world 2013; Alzheimer’s Society Website, 2015).
(Global Age Watch Index, 2014), Norway was one of the 2. A national ambition to identify 67% of people with
first countries to launch its national strategy (Dementia dementia while improving post-diagnosis support
Plan 2015, 2007) and is in the process of launching its was achieved by March 2015. In 2012, 42% of people
2015–2020 plan. in England were able to access a formal diagnosis,
One notable innovation from Norway has been the increasing to 58% in January 2015.
development of ‘Active Care’ day programmes supported by 3. Over 108,000 National Health Service (NHS) staff
municipality grants promoting cognitive stimulation, cul- have received foundation level dementia training with
tural activities and ‘green’ care on farms. The aim is to pre- a further 250,000 NHS staff who received training by
vent institutionalization and support well-being and social March 2015.
integration while offering relief to family caregivers. 4. In hospital settings, a commissioning incentive gener-
When it is necessary to move to a care home, these care ated around 4000 hospital referrals of people worried
centres are typically for small groups with direct access to about their memory signposts for subsequent diagno-
adapted outdoor activities, run by small organizations and sis each month.
a skilled workforce (Engedal and Haugen, 2009). 5. Research spending on dementia has increased by
nearly 50% since 2010, with an ambition to further
39.5.9 ENGLAND spend double the amount over the subsequent decade.
The Medical Research Council, United Kingdom,
A 5-year National Strategy, (Department of Health, 2009) announced the world’s biggest research sample
was superseded by the Prime Minister’s Challenge on (Medical Research Council, 2014) involving a 2 mil-
Dementia (Department of Health, 2012). Mr. Cameron, who lion strong cohort, while the Alzheimer’s Society
commented: ‘dementia now stands alongside cancer as one pledged £100 million towards research.
The international challenge of dementia 407

The G8 Dementia Summit in London 2013, during the Dementia diagnosis rates in Scotland currently stand at
UK presidency of the EU, initiated an unprecedented Global 64%, higher than other parts of the United Kingdom. All
Action Against Dementia, uniting international efforts to those diagnosed from April 2014 are entitled to support
fight dementia (Department of Health, 2013), subsequently workers to help them and their families understand demen-
establishing the World Dementia Council and the appoint- tia and make advance plans.
ment of a World Dementia Envoy.
The World Dementia Council now comprising of 18
members, appointed by the World Dementia Envoy, will 39.5.10.1 The Glasgow Declaration
work to build an international effort to fund doubly for (October 2014)
dementia research, increase the number of people involved
This declaration calls for the creation of a European Dementia
in clinical trials and to set an ambition to identify a cure or
Strategy and for world leaders to recognize dementia as a
disease-modifying therapy for dementia by 2025 (Annual
public health priority and to develop a global action plan on
Report of Progress, 2013).
dementia. It affirms that every person living with dementia
Given that, during 1998–2011, only four dementia drugs
has the right to
successfully made it to market out of over 100 experimental
drugs, the Council set four priorities in its drive to speed up
1. A timely diagnosis
research, which are as follows:
2. Access quality post-diagnostic support
3. Person-centred, coordinated quality care
1. Optimizing the path of medicines from research to
market, so that new treatments are developed in a 4. Equitable access to treatments and therapeutic
interventions
timely manner.
5. Be respected as an individual in their community
2. Exploring ways to increase investment in funding
dementia innovation by exploring new types of fund-
ing products. 39.5.11 THE UNITED STATES
3. Unleashing the potential of open science for sharing
information and knowledge to accelerate progress President Obama signed into law the National Alzheimer’s
in developing new treatments and care approaches, Project Act (NAPA) (2011). NAPA’s aims are as follows:
avoiding duplication of effort, assisted by the World
Health Organization (WHO) and Organization for 1. Prevent and effectively treat AD by 2025
Economic Cooperation and Development (OECD). 2. Enhance care quality and efficiency
4. Reviewing the evidence into existing research on 3. Expand support for people with dementia and their
dementia risk reduction and strengthening public families
health messaging on lifestyle and prevention. 4. Enhance public awareness and engagement
5. Improve data to track progress
The global action against dementia has since expanded
to include academic institutions, non-governmental organi- The term ‘dementia’ has now been replaced in American
zations and civil societies, and now embraces commitment medical terminology. The Diagnostic and Statistical Manual
from developing countries. As the burden of dementia is of Mental Disorders, Fifth Edition (American Psychiatric
heavier in non-G7 countries, all outputs need to be global. Association, 2013) has included a category named neuro-
Learning from other conditions, e.g. HIV and cancer, cognitive disorder, formally known in The Diagnostic and
and aligning with emerging initiatives, e.g. JPND and the Statistical Manual of Mental Disorders, Fourth Edition
ALCOVE Joint Action, are recognized as useful global (DSM-IV) as ‘dementia, delirium, amnestic and other cog-
strategies. nitive disorders’.
The global action against dementia has since held a series
of legacy events (each focusing on a specific theme), the last 39.5.12 CANADA
of which was in Tokyo, Japan, in November 2014.
In 2014, funding was allocated for the creation of the
39.5.10 SCOTLAND Canadian Consortium on Neurodegeneration in Aging
(CCNA) to focus on dementia. The CCNA is supported with
Quick to adopt national dementia plans, Scotland launched funding over 5 years from the Government of Canada and a
its second 3-year National Strategy in 2013 (Scottish public/private partnership, including the Alzheimer Society
Government, 2010). of Canada.
Key improvement areas are diagnosis and post-diagnos- Canada, a leader in dementia research, co-hosted one of
tic support, hospitals and modelling proactive eight-pillar four global actions against dementia legacy events (together
community-based support. with France).
408 Dementia

Effective international collaboration to harness and


39.6 PREVENTION AND RISK maintain momentum is necessary between and within
REDUCTION countries to provide continuity, promote a common pur-
pose and implement improvements in care, cure and pre-
Best estimates (World Alzheimer Report, 2014) combining vention synchronously, globally and locally. Pursuing a cure
a comprehensive approach from all sectors collaborating or disease-modifying treatment must not be at the expense
to reduce the risks associated with dementia, suggest that of developing and improving care and prevention for the
the future scale of the dementia epidemic may be blunted patients.
through improvements in population health.
There is mounting evidence that the population at risk of REFERENCES
suffering from dementia can be modified through a reduc-
tion in tobacco use, alongside better treatment for hyperten- Alzheimer’s Australia (2013). Dementia: A Public Health
sion and diabetes during mid-life. Education and life-long Priority. Available at http://campaign.fightdementia.
learning may improve the brain health of those entering old org.au/dementia-a-public-health-priority/. Accessed 6
age and reduce the incidence of dementia (Barnes and Yaffe, June 2016.
2011; Matthews et al., 2013). ALzheimer COoperative Valuation in Europe
The European Dementia Prevention Initiative (EDPI), (2013). ALCOVE Project. Available at http://www.
launched in 2011 in Stockholm, Sweden, allows researchers alcove-project.eu. Accessed 6 June 2016.
to collaborate in prevention studies. EDPI incorporates three Alzheimer Disease International (2010). World Alzheimer
large randomized controlled trials with a fourth prevention Report 2010, The Global Economic Impact of
study (Healthy Ageing Through Internet Counseling in the Dementia. London, United Kingdom: Alzheimer’s
Elderly) adopting innovative internet-based interventions Disease International. Available at http://www.alz.
recently added (HATICE, 2014). co.uk/research/files/WorldAlzheimerReport2010.pdf.
The International Database on Aging and Dementia Accessed 6 June 2016.
(IDAD) and EDPI are examples of ongoing international Alzheimer Disease International (2012). National
efforts aiming to improve the methodology of preventive Alzheimer and Dementia Plans Planned Policies and
studies and provide the basis for larger intervention trials Activities: Early Detection, Access to Diagnosis, and
(Solomon et al., 2014). Stigma Reduction. London, UK: Alzheimer’s Disease
Public health authorities must seize the opportunities International.
to do more in promoting the message on risk reduction. Alzheimer Europe (2006). Paris Declaration. Available at
Emerging research suggests some dementias are prevent- http://www.alzheimer-europe.org/Policy-in-Practice2
able diseases and that it is never too early or too late to pro- /Paris-Declaration-2006. Accessed 6 June 2016.
mote public health messages. The UK Department of Health Alzheimer Europe (2008a). European Collaboration on
now adopts new principles (Lincoln et al., 2014) outlining Dementia. Available at http://www.alzheimer-europe.
the need for future policy to promote a range of risk reduc- org/Research/European-Collaboration-on-Dementia.
tion strategies enshrined in service planning. Accessed 6 June 2016.
Alzheimer Europe (2008b). National Plan for "Alzheimer
and Related Diseases" 2008–2012. France: Alzheimer
Europe. Available at http://www.alzheimer-europe.
39.7 CONCLUSIONS
org/EN/Policy-in-Practice2/NationalDementia-Plans/
France. Accessed 6 June 2016.
An unprecedented increasingly robust global platform Alzheimer Europe Survey (2014). The Glasgow
for dementia is extending across Europe, Australasia and Declaration. Luxembourg, Europe: Alzheimer Europe.
North America. Whether countries change the wording for American Psychiatric Association (2013). The Diagnostic
dementia or decide to tackle the stigma of ‘dementia’ head- and Statistical Manual of Mental Disorders. Fifth
on, a long overdue upsurge is accruing at present, during Edition. Washington, DC: American Psychiatric
what could be dubbed the decade of dementia. Association.
The plight of individuals with dementia, their caregiv- Ankri, J. and Van Broeckhoven, C. (2013). Evaluation du
ers and their governments as a result of inadequate health Plan Alzheimer 2008–2012. France: Ministère des
and care provision are played out across the world. National affaires sociales et de la santé.
plans have shown the way and now people with dementia Annual Report of Progress (2013). Prime Minister’s
must be empowered, supported by appropriate communi- Challenge on Dementia; Delivering Major
cation within the public domain, media and educational Improvements in Dementia Care and Research by 2015.
institutions. London, United Kingdom: Annual Report of Progress.
The international challenge of dementia 409

Australian Government Department of Health (2006). Grossetête, F., Bowis, J., Lévai, K. et al. (2008). 0080/2008
National Framework for Action on Dementia, Written Declaration on Priorities in the Fight against
2006–2010. Canberra, Australia: Australian Alzheimer’s Disease. European Parliament, 8 October
Government Department of Health. 2008.
Barnes, D.E. and Yaffe, K. (2011). The projected effect of HATICE (2014). Healthy Ageing Through Internet
risk factor reduction on Alzheimer’s disease preva- Counseling in the Elderly (HATICE). Available at http:
lence. The Lancet Neurology, 10: 819–828. //www.hatice.eu. Accessed 6 June 2016.
Boustani, M., Callahan, C.M., Unverzagt, F.W. et al. (2005). Hausner, L., Frolich, L., Gardette, V. et al. (2010). Regional
Implementing a screening and diagnosis program for variation on the presentation of Alzheimer’s disease
dementia in primary care. Journal of General Internal patients in memory clinics within Europe: Data from
Medicine, 20: 572–577. the ICTUS study. Journal of Alzheimer’s Disease,
Brainy App (2011). Free Brain Health App. Available at 21: 155–165.
http://www.bupa.co.uk/brainyapp. Accessed 6 June Lee, S. (2010). Korea’s dementia strategy. International
2016. Journal of Geriatric Psychiatry, 25: 931–932.
Brooker, D., Fontaine, J.L., Evans, S. et al. (2014). Public Lincoln, P., Fenton, K., Alessi, C. et al. (2014). The
health guidance to facilitate timely diagnosis of Blackfriars consensus on brain health and dementia.
dementia: ALzheimer’s COoperative Valuation in The Lancet, 383: 1805–1806.
Europe recommendations. International Journal of Matthews, F., Arthur, A., Barnes, L. et al. (2013). A two-
Geriatric Psychiatry, 29: 682–693. decade comparison of prevalence of dementia in indi-
Canadian Institute of Health Research (2014). viduals aged 65 years and older from three geographical
Canadian Consortium on Neurodegeneration in areas of England: Results of the cognitive function and
Aging (CCNA). Available at http://www.cihr-irsc ageing study I and II. The Lancet, 382: 1405–1412.
.gc.ca/e/46475.html. Accessed 6 June 2016. Medical Research Council (2014). Dementias Platform UK
Chen, R., Hu, Z., Chen, R.L. et al. (2013) Determinants for (DPUK). Available at http://www.mrc.ac.uk/research
undetected dementia and late-life depression. The /facilities/dementias-platform-uk/. Accessed 6 June
British Journal of Psychiatry, 203: 203–208. 2016.
Norwegian Ministry of Health and Care Services (2015). Miyamoto, M., George, D. and Whitehouse, P. (2011).
Dementia Plan 2015. Oslo, Norway: Government of Government, professional and public efforts in
Norway. Japan to change the designation of dementia (chihō).
Department of Health (2009). Living Well With Dementia: Dementia, 10 (4): 475–486.
A National Dementia Strategy. London, UK: Olafsdottir, M., Skoog, I. and Marcusson, J. (2000).
Department of Health. Detection of dementia in primary care: The Linkoping
Department of Health (2012). Prime Minister’s Challenge study. Dementia and Geriatric Cognitive Disorders,
on Dementia. Delivering Major Improvements in 11: 223–229.
Dementia Care and Research by 2015. London, UK Prince, M. (2009). World Alzheimer Report 2009. London,
Department of Health. UK: Alzheimer’s Disease International.
Devos, P., Haeffner-Cavaillon, N., Ledoux, S. et al. (2014). Prince, M., Bryce, R. and Ferri, C. (2011). World
Evaluation of French Alzheimer Plan. The Lancet, Alzheimer Report 2011. Benefits of Early Diagnosis
383: 1805. and Intervention. London, UK: Alzheimer’s Disease
Dias, A. and Patel, V. (2009). Closing the treatment gap International.
for dementia in India. Indian Journal of Psychiatry, Saad, K. (2012). Dementia awareness starts in schools.
51 (5): 93–97. Nursing Times, 108 (43): 29.
Engedal, K. and Haugen, P.K. (2009). Demens. Fakta og The Scottish Government (2010). Scotland’s National
utfordringer. Tønsberg, Norway: Forlaget Aldring og Dementia Strategy. Edinburgh, Scotland: The Scottish
helse. Government.
European Commission (2014). European Innovation Smith, D. and Yaffe, K. (2014). Dementia (including
Partnership on Active and Healthy Ageing. Available Alzheimer’s disease) can be prevented: Statement sup-
at http://ec.europa.eu/research/innovation-union ported by International Experts. Journal of Alzheimer’s
/index_en.cfm?section=active-healthy-ageing. Disease, 38: 699–703.
The EU Joint Programme – Neurodegenerative Disease Stratégie Nationale de Sante (2014). Plan Maladies
Research (2014). JPND Research. Available at http: Neurodégénératives, 2014–2019. Paris, France:
//www.neurodegenerationresearch.eu. Accessed 6 Ministère des Affaires Sociales et de la Santé.
June 2016. Solomon, A., Mangialasche, F., Richard, E. et al. (2014).
Green, G. and Lakey, L. (2013). Building Dementia-Friendly Advances in the prevention of Alzheimer’s dis-
Communities: A Priority for Everyone. London, UK: ease and dementia. Journal of Internal Medicine,
Alzheimer’s Society. 275: 229–250.
410 Dementia

Valcour, V.G., Masaki, K.H., Curb, J.D. and World Dementia Council (2014). Global Action Against
Blanchette, P.L. (2000). The detection of demen- Dementia. Available at http://dementiachallenge.
tia in the primary care setting. Archives of Internal dh.gov.uk/category/wdc/.
Medicine, 160: 2964–2968. World Health Organization (2008). Mental Health
Wilkins, C.H., Wilkins, K.L., Meisel, M. et al. (2007). Gap Action Programme (mhGAP), Scaling up Care
Dementia undiagnosed in poor older adults with func- for Mental, Neurological, and Substance Use
tional impairment. Journal of the American Geriatrics Disorders. Geneva, Switzerland: World Health
Society, 55: 1771–1776. Organization.
Wimo, A., Jönsson, L., Gustavsson, A. et al. (2011). Cost World Health Organization and Alzheimer’s Disease
of illness and burden of dementia: The base option. International (2012). Dementia: A Public Health Priority.
International Journal of Geriatric Psychiatry, 26: 825–832. Geneva, Switzerland: World Health Organization,
World Alzheimer Report (2013). Policy Brief for Heads pp. 92–93.
of Government: The Global Impact of Dementia
(2013–2050). London, UK: World Alzheimer Report.
40
The pharmaceutical industry and dementia:
How can clinicians, researchers and industry
work together for the betterment of people
with dementia and their families? Continuous
vigilance and transparency is the answer

MARTIN M. BEDNAR AND LEON FLICKER

If I have seen further it is by standing on ye shoulders of Giants.

—Sir Isaac Newton, 1676

and memory of AD patients. In parallel, the pharmaceutical


40.1 ACADEMIC HEALTHCARE industry, recognizing the importance of the structural and
INVESTIGATORS, THE biochemical derangements in the brains of AD patients, was
PHARMACEUTICAL INDUSTRY able to produce a class of therapeutics known as acetylcho-
AND DEMENTIA linesterase inhibitors. Initial reports (e.g. Summers et al.,
1986) necessitated a standardized approach to evaluation of
dementia drugs (Leber, 1990). After successful multicentre
40.1.1 THE RELATIONSHIP CAN BE randomized trials (e.g. Davis et al., 1992), tacrine was the
PRODUCTIVE first drug approved in 1993 by the United States Food and
Drug Administration (FDA) and heralded a major advance
The therapeutic advances that have improved the lives of in the treatment of people with AD.
individuals with dementia, most notably Alzheimer’s dis- The history of the development of acetylcholinesterase
ease (AD), are the culmination of scientific innovation inhibitors for the treatment of AD is notable for several rea-
emanating from very distinct sectors of the scientific com- sons. It is an example of rational drug design that has domi-
munity. The successful ‘handoff’ of scientific information, nated drug development since the 1950s, largely (though
often obtained from publicly funded endeavours seemed, in not completely) replacing accidental drug discovery that
a sense, well-orchestrated albeit not pre-planned. While aca- was prevalent in the first half of the twentieth century and
demic healthcare investigators demonstrated a critical defi- the reliance on folklore prior to the twentieth century. It
ciency of the neurotransmitter acetylcholine in the brains of is also an excellent example of how the cross-fertilization
AD patients in the 1970s (Davies and Maloney, 1976; Davies of diverse groups of scientists and clinicians within both
and Verth, 1977), the use of available agents such as cho- academia and pharma, with different skill sets and goals,
line supplements did not meaningfully improve the lives resulted in a significant therapeutic advance to the benefit

411
412 Dementia

of patients. What is clear is that neither group could have patent protection, market share and revenue? Thus, there
accomplished this goal anywhere near as effectively on may be a key conflict in the academia–pharma relation-
their own. It is also clear that each group contributed to ship: on one hand, pharma’s advancement of applied science
the success of the other by providing critical data, access to create and market new medicines that also create share-
to patients and feedback (forward and back translation) holder value versus academia’s pursuit of advancements in
over the 15-plus years it took from concept to the first drug basic and clinical science as well as the fiduciary responsibil-
approval for AD as well as for the 15-plus years since that ity of the healthcare professional to do what is in the best
first drug approval. This is but one example of many where interest of the patient.
academia and pharma have complemented, enriched and The group of academic researchers who are also active
enhanced the professional roles of each other for the bet- clinicians, are particularly prone to a duality of interests,
terment of society. which often are congruent but sometimes conflicting. They
The key aspect of the academic investigator–pharma have simultaneous duties to develop new medications that
relationship outlined above, defining disease targets (mech- will provide their sponsors with an increase in share value
anisms) that, in turn, facilitate drug design, has been primar- and society with better treatments, but also a duty to their
ily opportunistic and has not required prospective codified patients to provide the best available treatment for that indi-
guidelines (rules) for engagement. However, more inter- vidual. This duality of interests frequently coexist without
dependent activities readily demonstrate points of friction conflict, but sometimes there is conflict between these com-
within this success story. Some of these include defining the peting demands.
value, financial and medical, of acetylcholinesterase inhibi- With this historical backdrop, the path forward should
tors, for a given country or world region or for a particular acknowledge that a relationship between academic health-
stage of illness (mild, moderate and severe). This was par- care investigators and pharma is in the best interest of soci-
ticularly important with drugs such as tacrine, which not ety and the patient. Each has complementary skill sets (see
only had major financial costs but large numbers of patients Table 40.1), resources and goals that, if appropriately har-
who developed hepatotoxicity (Ames et al., 1990). Variable nessed, can compensate for knowledge and resource gaps of
patient access to needed medicines, usually on the basis of each party. The key issue is how to effectively and efficiently
cost, is another issue that deserves attention to ensure that use these resources, skills and knowledge in a way that
needed medicines are available to the patients in need of acknowledges potential conflicts (both between and within
them. Increasingly there are mechanisms to evaluate the pharma and academia; see Table 40.2), minimizes or elimi-
cost-effectiveness so as to determine which medications will nates the potential for personal gain to supersede societal
be supported by third-party payers, although decisions can benefit and provides the needed transparency.
vary widely from country to country (Bae et al., 2015). Thus, it is reasonably certain that pharma will continue
to rely on academic healthcare investigators to conduct basic
40.1.2 THERE IS A POTENTIAL FOR research that better defines the pathophysiology of disease
CONFLICT OF INTERESTS, and that identifies new therapeutic targets, paving the way
PARTICULARLY WITH CLINICIAN for the development of innovative medicines. Pharma will
RESEARCHERS also continue to look to academic healthcare investigators
to facilitate access to consenting patients necessary for the
Another aspect is the question of innovation ver- conduct of clinical trials for these potential new medicines.
sus ­incremental improvement of a medicine or class of A significant perturbation of these ‘traditional’ roles for
medicines – e.g. pharmacokinetics, route of delivery, safety pharma and academia seems both unlikely to occur and of
and efficacy advantages within this class of therapeutics. Is doubtful value. The current limited infrastructure of aca-
each of these characteristics truly driving innovation and demia does not lend itself to forging new pathways for the
providing enhanced value for the patient or are they simply academic healthcare investigator to translate promising
strategies used by the pharmaceutical industry to maintain therapeutic targets into proven treatment strategies.

Table 40.1 Academia–pharma: complementarya skill sets

Academia Pharmaceutical Industry


Original basic science idea, e.g. novel receptor, Applied creativity, e.g. druggable chemical series
biochemical action
Research focus allows for digression to follow novel Rational drug design maintains focus on operationalization
observations that may not be related to the original
research question
Novel observations typically anecdotal Rigor of safety and efficacy of a therapeutic must meet
regulatory (societal) demands before being marketed
a Though not intended to suggest, they are mutually exclusive.
The pharmaceutical industry and dementia 413

Table 40.2 Confounders in the healthcare–pharma relationship


Does disclosure of conflict of interest by healthcare professionals genuinely alleviate the potential of bias or simply serve
to remove attention from it (Kassirer, 2009)?
Limited numbers of opinion leaders who sway national and international prescribing. Alternatively, pharma seek prominent
healthcare professionals to improve credibility. The potential for coercion and abuse exist.
Loss of potential healthcare leaders, the clinician–scientist, due to both reduced funding and the dedicated time to do so
(vicious cycle).
Physicians must remain unbiased and devoted to patient care, but they are already influenced by federal constraints,
managed care regulations and reimbursement.
Primary goals of an academic institution (e.g. physician hours involved with direct patient care, the rise of for-profit
academic medical centres and also private practice consortiums that conflict with the traditional charitable/altruistic
service of the physician) may not align with the goals of the healthcare professional, where publications and grants can
be the most significant factor in promotions.
The need for pharma to sponsor/subsidize healthcare research, meetings, etc. has increased as traditional sources of
funding diminish. Some degree of regulation has now been brought into play.
Academia-led research does not undergo the same degree of scrutiny as that in pharma-reverse bias (Doogan, 2009).
The potential for data fraud both by healthcare professionals and pharma for secondary gain.
Knowledge sharing versus the protection of intellectual property (both academia and pharma).
Some companies are interested in orphan diseases that may not generate blockbuster profits or significant shareholder
value either as part of their overall mission (typically larger pharma) or because even a modest revenue is sufficient for
their business model (smaller, biotech companies).

On the other hand, to conclude that the responsibility 40.1.3 PARTICULAR ISSUES IN


of the academic healthcare investigator to always act in the DEALING WITH PEOPLE WITH
best interest of the patient is in opposition to pharma’s goal DEMENTIA – CHALLENGES FOR
to generate financial value from the creation of new medi-
BOTH CLINICIANS AND
cines is misleading, but nonetheless frequently cited. The
fiduciary responsibility of the academic healthcare inves- RESEARCHERS
tigator to their patients actually relies on the interaction There are a number of issues in the treatment of dementia
of the healthcare professional with pharma to facilitate that, if not unique, are certainly more prominent and they
improvements in health care, including the bidirectional add additional complexity to the relationship of healthcare
exchange of information regarding translational medicine
professionals and the pharmaceutical industry. The most
(bench-to-bedside and bedside-to-bench) and to provide
obvious of these is the disease itself. With varying degrees
(directly and through regulatory agencies and other gov-
of dementia, the patient may have limited or no capacity
ernance bodies) information sharing on marketed prod-
to understand the nature of the research and its risks and
ucts. Indeed, pharma has been one of leaders in public and
benefits. This results in family members and/or caregiv-
professional education of AD (and most other diseases),
ers serving as both the patient proxy for consent as well as
raising awareness and allowing therapies to reach many
more individuals than would otherwise have been possi- being involved in judging the efficacy of a potential new
ble. Thus, to a certain degree, it is this bidirectional flow of medicine. Does the relationship between patient and care-
information that helps to mitigate idiosyncratic physician giver alter the way that the physician provides informa-
practice patterns and that facilitates physician knowledge tion in seeking informed consent? If the trial is targeting
and competency, which serves as the foundation of the challenging behaviours associated with dementia this may
physician’s role in the care of the patient. Also of note is be particularly problematic. Similar issues are faced by
the emergence of the precompetitive consortia and public–­ clinicians treating people with dementia and challenging
private partnerships whereby pharma and academic behaviours. Whose symptoms are we managing and how
experts, in addition to government agencies and vari- valid is this?
ous societies/advocacy groups have openly collaborated The role of the physician and also of the family/care-
to achieve advances in the field that might not have been giver to act on behalf of the patient can be biased by the
possible or, at the very least, would have been significantly type of medical facility, which is chosen to be involved in
delayed. The Alzheimer’s Disease Neuroimaging Initiative clinical trials. Is it the physician or is it the site which is
(ADNI) and the Australian Imaging, Biomarkers and interested in participating in clinical trials? Are the ther-
Lifestyle Flagship Study of Ageing (AIBL) are two excel- apies studied at a particular site those showing greatest
lent examples of this collaborative effort in the field of AD, promise or those of most interest to a particular physician
which has since spawned similar initiatives worldwide. (the two are not mutually exclusive)? Many of the putative
414 Dementia

disease-­modifying therapies now in clinical development the population with a disease under study has the poten-
must be administered parenterally. If a hospital or clinic is tial to benefit everyone involved, from the patient to the
not equipped to support this paradigm, clinical trial options third-party payer by providing an increasing likelihood
for their patients to consider will be much more limited. that a new medicine will improve either patients at a spe-
The pharmaceutical industry must also struggle with cific stage of the disease and/or their specific set of signs
major pragmatic issues. One of these is the cost of clini- and symptoms in a much more precise manner. Federal
cal trials for dementia. For drugs that may be potentially regulators, third-party payers and the public must care-
disease-modifying, late-stage clinical trials are long (at least fully review the data to determine the societal benefit and
18–24 months), expensive and carry a very high risk of fail- risk, both in the short and long term, when approving
ure due to their unprecedented nature. Of course, the risk and paying for these new medicines. Critical participants
is not only to pharma but also to patients, where the true in this debate are the academic healthcare investigators.
risk of participating in the clinical trial is never certain and Their influence is considerable and speaks to the need for
the benefit for an AD disease-modifying agent is unclear. collaboration of academia and pharma, from defining the
The analogous experience with acute stroke is edifying, pathophysiology of disease to the education of society. Not
where the great risk and clinical trial expense are contribut- only will the academic healthcare investigators continue
ing factors to the current paucity of neuroprotective trials. to be recognized as the spokespersons for the patient and
The 1990s were heralded as the decade of the brain in the other clinicians (and, by extension, for society) but will
United States and the approval of tissue plasminogen activa- also have an important role in the creation, validation and
tor (tPA) in 1995 provided great promise. Now 20 years later, societal uptake for the various tests and scales that will be
not a single neuroprotective therapy has been approved for used to both assess patients during the various stages of
stroke. Similarly, not a single disease-modifying agent has their disease and judge the efficacy of potential new medi-
been proven to be effective for AD dementia to date. cines for treatment.
There is also the issue of when is the most favour-
able time to treat patients with cognitive impairment.
Traditionally, clinical trials had targeted patients who are 40.2 FUTURE DIRECTIONS
in the mild-to-moderate stage of the disease. This is, in
part, a carry over of the clinical development for symp- We have discussed the historical perspective of the
tomatic therapies such as acetylcholinesterase inhibi- academia–pharma relationship. This relationship has
­
tors where there was concern that patients at the severe evolved over the past several decades to create a significant
end of the spectrum may fail to benefit from therapy due degree of interdependency, despite the fact that at least some
to the widespread and prolonged nature of their disease. of their goals are neither mutual nor similarly prioritized.
However, the question regarding the timing and duration This has the potential to create an inefficient and ineffective
of therapy remains not only about late-stage disease but system filled with conflict.
also about earlier treatment. Most clinicians would hope In looking ahead, there is a need better to understand how
that patients could be diagnosed with AD well before they clinicians, academic healthcare investigators and pharma can
reach the stage of dementia. The terminology of mild cog- all benefit without issues of conflict of interest overwhelming
nitive impairment (MCI) or, more recently mild neuro- these relationships. This chapter has cited the development of
cognitive disorder due to AD seeks to characterize those acetylcholinesterase inhibitors as a successful, albeit opportu-
with objective but modest cognitive decline who are still nistic collaboration between pharma and academic healthcare
independent in everyday activities (American Psychiatric investigators. In the future, a more proactive strategic align-
Association, 2013). These people are likely to develop ment will assist in ensuring the timely and efficient develop-
dementia (convert) within a relatively brief period of sev- ment of improved models of healthcare and new medicines for
eral years. However, while many feel that there is clear merit patients, e.g. precompetitive consortia.
in the treatment of AD at earlier stages before there is sig-
nificant loss of brain function (much like treating patients 40.2.1 ARE THERE PROBLEMS THAT NEED
with heart disease before there is significant heart failure), TO BE FIXED?
there remain others who feel that this is an industry-led
example of ‘disease mongering’, an attempt to increase As can be seen, the relationship between prescribers, other
the commercial market and hence drug sales (Doran and health professionals, academia and the pharmaceutical
Henry, 2008). As mentioned with the precompetitive con- industry is complex and multilayered. Despite this, there
sortia such as ADNI, AIBL and now many others, there is are substantial benefits to be obtained for patients if this
strong agreement across pharma, academia and regulators collaboration can be managed successfully. A complex rela-
to treat this disease as soon as possible. There are a number tionship can still be very functional. Is there evidence that
of examples where either the criteria for diagnosing dis- these evolving interrelationships have become dysfunc-
ease have been redefined or where distinct ‘new’ diseases tional? If so, how these difficulties can be proactively solved?
have been introduced (e.g. fibromyalgia) followed by the Below are provided some examples where there have been
introduction of a medicine to treat it. This segmentation of issues of conflict and potential solutions.
The pharmaceutical industry and dementia 415

40.2.2 INCOMPLETE REPORTING OF analyses demonstrating that the risk of major bleeding


TRIAL EVIDENCE episodes could be reduced substantially if plasma levels
of the drug were monitored (Cohen, 2014). Is seems that
There are many people who benefit from the performance commercial interest was one of the reasons that these
of well-conducted scientific studies. Academics receive data were not made available (Cohen, 2014). As noted by
accolades and promotion by the conduct of major stud- Doshi et al. (2013), making raw data from clinical trials
ies particularly if the reports are published in prestigious widely publicly available should reduce selective report-
journals. Companies may make substantial profits if a ing biases and enhance the reproducibility of and trust
new class of drugs is shown to be therapeutic resulting in in clinical research.
blockbuster status for these drugs. Future generations of
patients will benefit from efficacious treatments to help 40.2.3 SOLUTION
prevent and alleviate their symptoms. The people who bear
the most risk from studies and have the least likelihood These concerns have resulted in the creation of trial reg-
of benefits are the subjects of these studies. These people istries, and the adoption of the position by many journals
consent to randomization to either ineffective treatments that trials will not be published unless they have been
(placebo) or drugs that potentially may yield no benefits prospectively registered. The problem is that there is no
and may have unknown side effects. This group of people compunction that the now known studies’ data should be
participates for many reasons but for most it is not just widely available. One solution would be that repositories
the desperation of seeking effective treatment. Many are for unpublished trials should become widely available
driven by an altruistic desire to benefit not just themselves, and a requirement from institutional ethics committees
but for future generations. The gift of participation of that simple tables of the results of the primary efficacy
these individuals places an obligation on all the organiza- variables be placed in such repositories within 5 years of
tions and study investigators who may potentially benefit study completion. The responsibility for completion of this
that, the participation of these subjects should firstly be requirement will need to be shared between the academic
reported, and the report should be an accurate and true researcher and the sponsoring company. It is imperative
record of the subjects’ participation. that all relevant human data are made available in a trans-
For this reason, it is most important that every subject parent fashion.
in a randomized trial (i.e. trials which are hypothesis test-
ing) should have their data incorporated into the results 40.2.4 DELIBERATE MANUFACTURE OR
of that trial and that all these trials should be reported. CONCEALMENT OF PATIENT DATA
Unfortunately whenever this is reviewed, the conclusion is
that negative trials are much less likely to be reported than For some years concerns have been raised regarding the
positive studies. For example, in the early trials of rivastig- deliberate manufacture of patient results. The situation is
mine, of the four pivotal phase 3 studies, only two were almost certainly very uncommon but the question of how
reported, and these two reported studies showed a greater often it occurs, as opposed to its detection, remains unre-
response than the two unreported studies (Birks et al., solved. An example of this practice was discovered in the
2009). It is not just the pharmaceutical industry who finds it Second European Stroke Prevention Study (ESPS-2), a
difficult to report negative or similar results. It is difficult to major clinical trial to prevent repeat strokes. It was found,
convince journals to publish such studies especially if there rather inadvertently, that one of the investigators had fab-
have been other studies, which have yielded positive results. ricated data on subjects. This occurred without the trial’s
However, one of the stated purposes of Cochrane is that sponsor’s knowledge (Enserink, 1996).
all randomized controlled trials (RCTs) will be placed in a The lure of benefits has not just affected those researchers
repository and then subsequently incorporated into at least involved in pharmaceutical studies. Some academics, have
one systematic review. This approach has heightened the also fabricated patient data so as to increase their impor-
imperative to report all RCTS and fulfil the obligation that tance in the field overall (e.g. Hagmann, 2000). Fortunately
all parties have to the patients who participated in these because this study’s results conflicted dramatically with
trials. This strategy may improve the reporting of clinical other investigators’ work, the fabrication was detected early
trials, which is typically noted to be at extremely low levels, and did not result in the inappropriate management of
both by academics (perhaps to a greater extent) and pharma patients been widely adopted. A more recent example from
(Anderson, 2015). an academic research group in Japan claiming a simplified
Since the last edition of this textbook, there have strategy to transform adult cells into pluripotent stem cells
been further examples of withholding of valuable anal- was uncovered in a similar manner (Nature Editorial, 2014).
yses from the public eye. An example of this is dabi- One of the difficulties with studies that arise from academia
gatran, one of the classes of novel oral anticoagulants is that because of their limited budgets such studies do not
(NOACs). Although the drug is safe and efficacious, have the extensive auditing procedures that are used in
internal documents from the manufacturer reported pharmaceutical studies. Continuous vigilance is required
416 Dementia

to ensure that such fraud does not become more common- expedite the review process as well as to ensure that those
place. The propensity to fraud will always exist when there engaged in misconduct are readily identified to spon-
are perverse incentives for the dishonest researcher who is sors of drug or device trials (website: http://www.gao.gov/
willing to cut corners and bow to the increasing pressure to assets/300/296033.html).
maintain ‘productivity’.
The issues become even more complex with pharma- 40.2.6 EDUCATION OF CLINICIANS
ceutical studies. Not only is there the perverse incentive of
academic advancement but also there is the added impe- The pharmaceutical industry not only has a legitimate
tus of monetary gain for the investigators for recruitment right to educate doctors and the general public about the
of subjects and also the requirement for all pharmaceu- advances in treatment that have been achieved but also has
tical companies to maximize the return to shareholders. a responsibility. It is the methods that are employed, which
The recent inexplicable failure to report deaths in a large have raised concerns.
study of rofecoxib may point to bias by the pharmaceutical
industry to suppress negative findings (Krumholz et al., 40.2.7 TREATMENT GUIDED BY EVIDENCE
2007). Many journals now have such safeguards in place, OR TRUSTED AUTHORITIES?
requiring acknowledgement that all writing was the effort
of the authors at the time the article is initially submitted. Both physicians and the general public believe that evi-
Such shortcomings have severe consequences for both the dence should guide rational prescribing. Unfortunately
general public but also the company itself if these short- when doctors are ‘shadowed’ this is not the mechanism by
comings are exposed. which doctors seek solutions to clinical problems. Doctors
seek solutions from other doctors and due to the hierarchi-
40.2.5 SOLUTION cal relationship of this transfer of information, a relatively
small number of opinion leaders guide national and inter-
The perverse incentives in academia need to be addressed national prescribing patterns. The pharmaceutical indus-
to discourage output for its own sake. Such pressure has try has a societal responsibility to contract such opinion
been found to underlie many cases of scientific fraud. This leaders, to educate them about their products and to seek
is an issue for academic departments whose oversight of their advice as to how to further investigate the benefit–risk
individual academic’s practices often ranges from cur- ratio for a particular medicine. One problem has been that
sory to non-existent. This is not the responsibility of the historically these contractual relationships are covert and
institutional ethics committee to police such practices, unregulated and this has led to questions such as ‘when
as in some instances the ethics committee may not even does reimbursement for services rendered become coer-
be aware that fraudulent ‘trials’ are even taking place cion’? More recently, hospitals and medical centres have
(Hagmann, 2000). The benefits of scientific publication begun to mandate the reporting of any perceived conflict
and grants are usually shared by the academic investiga- of interest, a policy that will improve transparency and the
tor’s institution, but these benefits should be tied to the perception of the relationship between pharma and health-
responsibility of ensuring proper academic conduct. care professionals.
The controversy surrounding rofecoxib has caused
greater scrutiny on the relationship between academic 40.2.8 SOLUTION
researchers and pharma. Late-stage clinical studies in
particular may require additional safeguards. These could There is a need for greater transparency in the relationships
include storage of data independently to the sponsor and between opinion leaders and industry. Learned colleges and
analysis by independent investigators. Independent data medical associations have argued for such transparency
and safety monitoring boards need not be under the con- but these organizations have not indicated how compliance
trol of the sponsor, and could be under the direct control with these guidelines will be monitored or what would be
of an independent academic institution. The practices of the consequences of non-compliance to the practitioner, the
‘ghostwriting’ and false claims of full access to all relevant industry and the colleges themselves. The level of transpar-
data should be eliminated (Krumholz et al., 2007). It is in ency required has to be sufficient so that those people reliant
the interests of the pharmaceutical industry to lead such on information can judge whether any reimbursement may
change as the risk for maintaining the status quo may ­­be have influenced the opinion leader and by how much. There
large. should be limits placed on the compensation provided for
Additionally, the FDA has implemented additional different tasks, e.g. speaking at a meeting. Unfortunately,
actions to ensure that investigators who are non-­compliant there is no evidence that such disclosure has any influence
with the FDA’s statutes and regulations intended to pro- on the prescribing habits of those who have been influenced
tect study subjects or who knowingly reporting inaccurate by individual opinion leaders. The provision of excessive
study findings are not allowed to continue to participate gifts or travel grants to nonspeaking participants at inter-
in clinical trials. These additional actions are aimed in to national conferences would appear to be frank coercion and
The pharmaceutical industry and dementia 417

these practices should cease. Fortunately, these safeguards Davies, P. and Maloney A.J.F. (1976). Selective loss of
are now commonplace, although not universal. cholinergic neurons in Alzheimer’s disease. Lancet,
2: 1403.
Davies, P. and Verth, A.H. (1977). Regional distribution
of muscarinic acetylcholine receptor in normal and
40.3 CONCLUSIONS Alzheimer-type dementia brains. Brain Research,
138: 385–392.
In general, the relationships between clinicians, academic Davis, K.L., Thal, L.J., Gamzu, E.R. et al. (1992). A double-
researchers and the pharmaceutical industry have been blind, placebo-controlled multicenter study of tacrine
healthy and conducive to the best interests of patients. They for Alzheimer’s disease. The Tacrine Collaborative
have produced results for patients which would have been Study Group. New England Journal of Medicine,
unachievable by the individual parties. There are particu- 327: 1253–1259.
lar issues regarding people with dementia that may make Doogan, D. (2009). In support of industry-sponsored
these relationships even more complex. There have been clinical research. In Snyder, P.J., Mayes, L.C. and
some recent incidents, which have resulted in some ten- Spencer, D. (eds.), Delgado’s Brave Bulls: The Complex
sions to appear in these relationships, which may erode the Relationship between Science and the Media. New
essential trust on which these relationships are founded. An York, NY: Elsevier, Inc.
ongoing dialogue between pharma, healthcare profession- Doran, E. and Henry, D. (2008). Disease mongering:
als and learned colleges and regulators is critical for both Expanding the boundaries of treatable disease.
understanding the issues and finding constructive solu- Internal Medicine Journal, 38: 858–861.
tions. Several changes are suggested that will allow greater Doshi, P., Goodman, S.N. and Ioannidis, J.P.A. (2013).
transparency for all the parties concerned. Raw data from clinical trials: Within reach? Trends in
Pharmacological Sciences, 34: 645–547.
REFERENCES Enserink, M. (1996). Fraud and ethics charges hit stroke
drug trial. Science, 274: 2004–2005.
American Psychiatric Association (2013). Desk Reference Hagmann, M. (2000). Scientific misconduct. Cancer
for the Diagnostic Criteria from DSM5. Arlington, researcher sacked for alleged fraud. Science,
VA: American Psychiatric Association. 287: 1901–1902.
Ames, D.J., Bhathal, P.S., Davies, B.M. et al. (1990). Kassirer, J.P. (2009). Medicine’s obsession with disclo-
Heterogeneity of adverse hepatic reactions to tetrahy- sure of financial conflicts: Fixing the wrong problem.
droaminoacridine. Australian and New Zealand Journal In Snyder, P.J., Mayes, L.C. and Spencer, D. (eds.),
of Medicine, 20: 193–195. Delgado’s Brave Bulls: The Complex Relationship
Anderson, M.L., Chiswell, K., Peterson, E.D. et al. (2015). between Science and the Media. New York, NY:
Compliance with results reporting at . New England Elsevier, Inc.
Journal of Medicine, 372: 1031–1039. Krumholz, H.M., Ross, J.S., Presler, A.H. and Egilman,
Bae, G., Bae, E.Y. and Bae, S. (2015). Same drugs, valued D.S. (2007). What have we learnt from Vioxx? British
differently? Comparing comparators and methods Medical Journal, 334: 120–123.
used in reimbursement recommendations in Australia, Leber, P. (1990). Draft Guidelines for the Clinical
Canada, and Korea. Health Policy, 119: 577–587. Evaluation of Antidementia Drugs. Washington, DC:
Birks, J., Grimley Evans, J., Iakovidou, V. and Food and Drug Administration (FDA publication no.
Tsolaki, M. (2009). Rivastigmine for Alzheimer’s F91–19331).
­disease. Cochrane Database of Systematic Reviews, Nature Editorial. (2014) STAP retracted. Nature, 511: 5–6.
(2): CD001191. Summers, W.K., Majovski, L.V., Marsh, G.M. et al. (1986).
Cohen, D. (2014). Dabigatran: How the drug company Oraltetrahydroaminoacridine in long-term treatment
withheld important analyses. British Medical Journal, of senile dementia, Alzheimer type. New England
349: g4670. Journal of Medicine, 315: 1241–1245.
2
Part    

Mild Cognitive Impairment

41 Mild cognitive impairment (MCI): A historical perspective 419


Karen Ritchie and Sylvaine Artero
42 Clinical characteristics of mild cognitive impairment 426
Yonas E. Geda, Janina Krell-Roesch, Anna Pink, Kathleen A. Spangehl and Ronald C. Petersen
43 Managing the patient with mild cognitive impairment 439
Nicola T. Lautenschlager and Alexander F. Kurz
41
Mild cognitive impairment (MCI): A historical
perspective

KAREN RITCHIE AND SYLVAINE ARTERO

he referred to as ‘depressive pseudo-dementia’, long-term


41.1 COGNITIVE IMPAIRMENT AS A follow-up of some of these patients showed that a significant
FEATURE OF NORMAL AGEING proportion went on to develop vascular dementia. BSF was
diagnosed by Kral by an open psychiatric interview and no
Alterations in cognitive functioning in the absence of formal algorithm was proposed. Formal diagnostic criteria
dementia have long been considered a normal aspect of age- for non-dementia cognitive impairment were first proposed
ing related to the changes in brain. Affecting, principally by Crook et al. in 1986 for the National Institute of Mental
episodic verbal memory, such changes are generally dis- Health. Referring to ‘age-associated memory impairment’
tinguished from neurodegenerative disorders by their far (AAMI), they defined these changes as subjective com-
slower progression, their lesser impact on ability to perform plaints of memory loss in elderly persons verified by a decre-
activities of daily living and the relative sparing of linguistic ment of at least one standard deviation on a formal memory
and visuospatial functions. Increased interest in the nature test in comparison with means established for young adults.
and long-term prognosis of ageing-related modifications Blackford and La Rue (1989) later suggested suitable tests
in cognitive performance has now led to the question as of non-verbal and verbal secondary memory tests, propos-
to what extent they may be considered ‘normal’. The past ing that AAMI be defined as at least one standard deviation
50 years have seen numerous attempts to define these sub- performance below the mean for young adults for one or
clinical alterations in cognitive functioning and to estab- more of these tests. They also differentiate AAMI from the
lish their aetiology with greater precision than the general more severe state of ‘late-life forgetfulness’ (LLF), defined
notion of ‘ageing brain changes’ to which they have formerly as performance between 1 and 2 standard deviations below
been attributed. the mean on at least 50% of a battery of minimum four tests.
One of the earliest attempts to clinically character- Levy et al. (1994) argued that there is little reason a
ize normal ageing-related cognitive change as opposed to priori that cognitive decline in normal old age should be
dementia (dotage) is Gulliver’s description of the eternal confined to memory functions exclusively and in collabo-
elderly of Luggnagg. ‘In talking, they forget the common ration with the International Psychogeriatric Association
appellation of things, and the names of persons, even of (IPA) and the World Health Organization (WHO), pro-
those who are their nearest friends and relations …. The least posed an alternative concept of ageing-associated cog-
miserable among them appear to be those who turn to dot- nitive decline (AACD). The criteria for AACD not only
age, and entirely lose their memories; these meet with more admits to the possibility of a wider range of cognitive
pity and assistance’ (Swift, 1726). Two centuries later Kral’s functions being affected (attention, memory, learning,
notion of benign senescent forgetfulness (BSF) (Kral, 1962) thinking, language, visuospatial function) but also stipu-
provides further clinical precision to Swift’s observation, lates that the deficit should be defined in relation to norms
describing normal cognitive ageing as patient complaints of for elderly and not young adults to avoid confounding
a persistent difficulty in recalling detail, commonly featur- of decline with age cohort effects. This refinement takes
ing depressive symptoms. While Kral initially considered into account the early observations of researchers such
such complaints to characterize a depressive state, which as Schaie and Willis (1991) who have demonstrated that

419
420 Dementia

much of the difference in cognitive performance observed forms of underlying pathology. The 10th revision of the
between young and elderly cohorts is attributable to gen- International Classification of Diseases (ICD-10) (World
eration differences (notably in education and healthcare) Health Organisation, 1993), described, for example, ‘mild
rather than in ageing-related changes in the brain. A com- cognitive disorder’ (MCD), which refers to disorders of
parison of the performance of AACD and AAMI criteria memory, learning and concentration, often accompanied
in the general practice setting concluded that they do not by mental fatigue, which must be demonstrated by formal
identify the same individuals within general population neuropsychological testing and attributable to cerebral
studies, AACD targeting a severe state of impairment disease or damage, or systemic physical disease known to
within a larger AAMI group (Richards et al., 1999). cause dysfunction. MCD is secondary to physical illness
Recognition of the major international classifications or impairment, excluding dementia, amnesic syndrome,
of disease of subclinical cognitive deterioration linked concussion, or post-encephalitic syndrome. The concept
to the normal ageing process began with the appear- of MCD, which was principally developed to describe the
ance in Diagnostic and Statistical Manual of Mental cognitive consequences of autoimmune deficiency syn-
Disorders, Fourth Edition (DSM-IV) (American Psychiatric drome, expanded to include other disorders in which cog-
Association, 1994) related to the concept of age-related cog- nitive change is secondary to another disease process; it is
nitive decline (ARCD). Like AACD, it refers to an objective applicable to all ages, not just the elderly. Attempts to apply
decline in cognitive functioning due to the physiological MCD criteria to population studies of elderly persons sug-
process of ageing, however, no operational criteria or cog- gest it to be of limited value in this context, casting doubt
nitive testing procedures are specified. It is rather loosely on its validity as a nosological entity (Christensen et al.,
defined as a complaint of difficulties in recalling names, 1995) for this age group. DSM-IV (American Psychiatric
appointments or in problem-solving, which cannot be Association, 1994) has proposed a similar entity, mild
related to a specific mental problem or a neurological dis- neurocognitive disorder (MNCD), which encompasses
order. The concepts of ARCD and AACD, while clinically not only memory and learning difficulties but also per-
meaningful, are now rarely used in research principally ceptual-motor, linguistic and central executive functions.
because of the difficulty in proving that such changes are While the concepts proposed by the two international clas-
‘normal’ and not part of an unidentified subclinical pathol- sifications, MCD and MNCD do not constitute adequate
ogy. Many early studies employing these categories found algorithms for application in a research context, they do
them to be subsequently associated with underlying vascu- provide formal recognition of subclinical cognitive dis-
lar pathology and high rates of evolution towards dementia order as a pathological state requiring treatment and as
on follow-up (Celsis et al., 1997; Di Carlo et al., 2000; Ritchie a source of handicap and are thus, likely to be important
et al., 2001). within a legal context.
A similar concept of cognitive change, secondary
to multiple underlying disease processes, but in this
41.2 DISEASE MODELS OF case referring to elderly populations, is that of cogni-
tive impairment not dementia (CIND). The concept was
SUBCLINICAL COGNITIVE
developed within the context of the Canadian Study of
IMPAIRMENT Health and Aging and is defined with reference to neuro-
psychological testing and clinical examination (Graham
The early conceptualizations of age-related cognitive loss et al., 1997). Persons with CIND, like MCD and MNCD,
described above had in common the theoretical assump- are considered to have cognitive impairment attributable
tion that such changes are distinct from dementia and to an underlying physical disorder, but may also have a
other pathologies, being the consequence of inevitable, ‘circumscribed memory impairment’, which is a modi-
thus normal, ageing-related cerebral changes, such as atro- fied form of AAMI. CIND encompasses a wider range of
phy and vascular fragility. As parallel research into the underlying pathologies (and therefore has higher preva-
causes of dementia and cerebrovascular disease has now lence rates) than MCD and MNCD, including disorders
led to a clearer understanding of their aetiology, it has such as delirium, substance abuse and psychiatric illness,
also been shown that many of the physiological abnor- which are excluded from the ICD and DSM categories.
malities seen in these disorders are also present to a lesser More recently (see below), a new entity has appeared in
extent in subjects identified as AAMI, ARCD and AACD. Diagnostic and Statistical Manual of Mental Disorders,
Consequently, elderly persons with subclinical cogni- Fifth Edition (DSM-5) (American Psychiatric Association,
tive deficits have become the subject of neurological as 2013) called ‘mild neurocognitive disorder (mNCD)’
well as psychogeriatric research, the underlying question defined by a noticeable decrement in cognitive function-
being whether cognitive deficits of this type may be due ing that goes beyond normal changes seen in ageing. It
to underlying brain pathology, which might be potentially is a disorder that may or may not progress to dementia
treatable. Alternative concepts have subsequently appeared because mNCD is recognized as a heterogeneous group
in the literature linking cognitive disorder to various of disorders occurring throughout life. Although mNCD
Mild cognitive impairment (MCI): A historical perspective 421

has been defined in relation to findings relating to mild


cognitive impairment (MCI), it clearly covers more than 41.4 OPERATIONALIZING MCI
a pre-dementia syndrome including stable states of poor CRITERIA
cognitive functioning.
Subsequent studies using MCI criteria encountered numer-
ous difficulties. These were principally due to a lack of con-
41.3 MCI – SUBCLINICAL COGNITIVE sensus as to whether the concept referred to memory loss is
CHANGE AS PRODROMAL a cause for dementia or specifically, Alzheimer’s disease and
DEMENTIA because of the lack of a working definition of memory loss.
The result has been that population prevalence, the clini-
cal features of subjects identified with MCI and their clini-
MCD, MNCD and CIND are constructs that have been cal outcomes vary widely between studies and even within
developed principally for research purposes and consider studies where there has been longitudinal follow-up. A con-
cognitive disorder in the elderly to be heterogeneous, not sensus conference held in Chicago in 1999 confronted the
necessarily progressive, with treatment being determined many difficulties facing the MCI concept (Petersen et al.,
by the nature of the underlying primary systemic dis-
2001), notably its two underlying assumptions that it should
ease. On the other hand, many clinical observations of the
be confined exclusively to isolated memory impairment and
long-term outcome of cognitive complaints, particularly
the dilemma whether it should refer exclusively to a prodro-
those presented to memory clinics and neurology depart-
mal form of Alzheimer’s disease, or alternatively to a more
ments, led to the general conclusion by many neurologists
clinically heterogeneous group with an increased risk of
that subclinical cognitive disorder in the elderly is in fact
dementia due to any cause. While there is some limited evi-
principally, if not exclusively, early-stage dementia. Thus,
dence that a purely amnesic syndrome may exist (Richards
whereas dementia was considered by many in the early
et al., 1999), this category appears to represent only a
1990s to be an upward extension of a ‘normal’ process of
very small proportion (6%) of elderly persons with cogni-
progressive ageing-related cognitive deterioration, by the
tive deficit when the full range of cognitive functions are
end of the decade subclinical cognitive deficit began to be
examined. Higher rates of ‘circumscribed memory deficit’
perceived as a downward extension of a neurodegenerative
process. In 1997, Petersen et al. proposed diagnostic crite- (31.7% of CIND cases) are observed within the Canadian
ria for MCI, initially defined as consisting of complaints of Longitudinal Study (Graham et al., 1997), but only subjects
defective memory and demonstration of abnormal memory with a score below cut-off on the MMSE are examined and
functioning for age, with normal general cognitive func- the test battery itself consists predominantly of memory
tioning and conserved ability to perform activities of daily tasks. Memory tests also involve cognitive capacities other
living, this was then considered to be a prodromal form of than memory, notably attentional and linguistic capacities,
Alzheimer’s disease. MCI criteria proved difficult to apply so that it is difficult in the absence of a detailed neuropsy-
as they referred to poor cognitive functioning as assessed at chological examination to conclude that an individual has
one point in time, thus precluding appreciation of decline an isolated mnesic impairment. Many of the studies which
over time and difficult to differentiate from cohort effects, observe principally memory deficits in MCI have used
low intelligence quotient (IQ) and education. A later defi- almost exclusively memory tests in their diagnostic exami-
nition refined the initial concept by referring to memory nation, a self-fulfilling prophecy.
impairment beyond that expected for both age and educa- The Chicago consensus group concluded that subjects
tion level (Petersen et al., 1999). An alternative approach with MCI have a condition which is different from normal
has been to define it in terms of early-stage dementia. For ageing and are likely to progress to Alzheimer’s disease at an
example, Krasuki et al. (1998) refer to cognitive impair- accelerated rate; however, they may also progress to another
ment with a score of 20 or more on the Mini–Mental State form of dementia or their condition might improve. This
Examination (MMSE) and Zaudig (1992) defines MCI group, thus proposed subtypes of MCI according to the type
with a score of more than 22 on MMSE or from 34 to 47 of cognitive deficit and clinical outcome, distinguishing
on the Score from Structured Interview for Diagnosis of MCI-amnestic (MCI with pronounced memory impairment
Dementia of the Alzheimer Type (SIDAM) scale. Others progressing to Alzheimer’s disease), MCI multiple domain
have referred to criteria based on the Clinical Dementia (slight impairment across several cognitive domains leading
Rating Scale or Global Deterioration Scale scores (Flicker to Alzheimer’s disease, vascular dementia or stabilizing in
et al., 1991; Kluger et al., 1997). The concept of MCI sub- the case of normal brain ageing changes) and MCI single
sequently moved from being a non-benign, non-specific non-memory domain (significant impairment in a cognitive
form of memory impairment to a dementia prodromal domain other than memory leading to Alzheimer’s disease
form. As such, it became the focus of considerable interest or another form of dementia). It has subsequently been sug-
as it opened up the possibility of wide scale treatment of a gested that MCI can be further subdivided according to the
subclinical state by the new anticholinergic therapy being suspected aetiology of the cognitive impairment, in keeping
developed at this time for Alzheimer’s disease. with international classifications of dementia disorders: for
422 Dementia

example, mild cognitive impairment–Alzheimer’s disease cardiovascular and respiratory disorders (Ritchie et al.,
(MCI-AD), mild cognitive impairment with Lewy body 1996; Lopez et al., 2003; Palmer et al., 2003; Matthews et al.,
dementia (MCI-LBD), mild cognitive impairment and fron- 2008). However, it is the identification of those cases likely
totemporal dementia (MCI-FTD) and so on, however, clini- to evolve towards dementia which has been given priority,
cal studies have been unable to provide consistent empirical especially, given the d ­ evelopment of treatments that may
evidence for these subdivisions. On the basis of evidence delay onset of dementia. The ­potential ­treatment window for
from population studies presented at this conference, it was dementia is large, with twin ­studies ­indicating that insidi-
also accepted that MCI subjects could have difficulties in ous changes in ­cognitive ­performance may occur up to 20
everyday functioning. A study has confirmed functional years of age before disease onset (La Rue and JarviK, 1987).
loss to be a characteristic of all MCI subtypes (Aretouli and For this reason, at a conceptual level, MCI has been the
Brandt, 2010) and application of the revised criteria to a gen- principal focus of interest as ­longitudinal ­studies ­confirm
eral population cohort has been shown to greatly increase the high risk rates of MCI for dementia, with conversion
the ability of the algorithms to differentiate a pre-dementia estimates ­ranging from 10% to 15% per year (Petersen
syndrome for other syndromes (Artero et al., 2006). et al., 1997), 40% over every 2 years (Johnson et al., 1998),
20% over every 3 years (Wolf et al., 1998), 30% over 3 years
(Black, 1999), 53% over 3 years (McKelvey et al., 1999),
to 100% over 4.5 years (Krasuki et al., 1998). Population
41.5 THE FUTURE OF MCI ­studies show, ­however, lower ­dementia ­conversion rates
(2%–8% per annum) (Tierney et al., 1996; Johanssen and
A number of concepts now exist to describe subclinical cog- Zarit, 1997; Kluger et al., 1999; Hogan and Ebly, 2000;
nitive disorder in the elderly. Application of these concepts Artero et al., 2001; Morris et al., 2001) than those observed
in general population studies gives quite different prevalence in ­clinical studies (12%–31%) (Bowen et al., 1997; Devanand
rates both between emerging concepts and even between et al., 1997; Flicker et al., 1991; Huang et al., 2000; Bozoki
studies conducted using the same concept (Table 41.1). et al., 2001; Stephan et al., 2008) as they include a wider
Clearly, such deficits have multiple causes. Longitudinal range of ­cognitive i­mpairments, which are probably less
population studies of subclinical cognitive deficits have severe than those presented in clinical s­ettings as well.
demonstrated multiple patterns of cognitive change with Although, persons labelled as MCI have a greater risk of
variable clinical outcomes including dementia, depression, dementia, many do not experience a f­urther cognitive

Table 41.1 Characteristics and estimated prevalence rates (where available) of nosological entities defining cognitive
impairment in elderly persons without dementia

Population
Concept Criteria prevalence %
BSF Kral (1962) Memory complaints, forgetting details, depression
AAMI Crook et al. (1986) Memory impairment with decrement on formal cognitive test 7–38
compared to young adults
LLF Blackford and La Rue (1989) AAMI with greater decrement on 50% of a test battery
AACD Levy et al. (1994) Impairment on any formal cognitive tests compared to peers 21–27
ARCD DSM-IV Objective decline in cognitive functioning due to old age 8
MCD ICD-10 Disorders of memory learning and concentration demonstrated by 4
testing due to disease
MNCD DSM-IV Difficulties in memory, learning, perceptual-motor, linguistic and
central executive functioning due to disease
CIND Graham et al. (1997) Circumscribed memory impairment and low MMSE score due to 11–80
disease or old age
MCI Petersen et al. (1997) Complaints of defective memory, demonstrated by deficit in 3–15
cognitive tests conducted with normal general intellectual
functioning compared to peers due to dementia
mNCD DSM-5 Difficulties in memory and learning, language, complex attention,
executive function, perceptual-motor function, social cognition
compared to peers due to dementia or other reasons
Abbreviations: 
BSF, benign senescent forgetfulness; AAMI, age-associated memory impairment; LLF, late-life forgetfulness; AACD,
­ageingassociated cognitive decline; ARCD, age-related cognitive decline; DSM-IV, Diagnostic and Statistical Manual of
Mental Disorders, Fourth Edition; MCD, mild cognitive disorder; ICD-10, International Classification of Diseases,
10th Revision; MNCD, mild ­neurocognitive disorder; CIND, cognitive impairment not dementia; MMSE, Mini–Mental State
Examination; MCI, mild ­cognitive impairment; mNCD, mild neurocognitive disorder.
Mild cognitive impairment (MCI): A historical perspective 423

decline and may even revert to normal status (Wada-Isoe close collaboration between specialist clinical units refining
et al., 2012; Ward et al., 2012). Population studies further clinical criteria and epidemiologists monitoring screening
indicate that current criteria have poor specificity in the efficiency within the general population, where MCI will
general ­population ­setting and may also miss many per- principally be detected.
sons who will develop dementia within a few years, but that
age-adjusted ­cognitive measures in conjunction with mea- REFERENCES
sures of d ­ isability, depression and anatomical changes on
cerebral imaging (volume loss in hippocampal and ento- American Psychiatric Association (1994). Diagnostic and
rhinal cortex) may ultimately p ­ rovide better dementia pre- Statistical Manual. IV. Washington, DC: American
dictions (Tierney et al., 1996; Touchon and Ritchie, 1999; Psychiatric Association.
Artero et al., 2001; Ritchie et al., 2001; Visser et al., 2002; American Psychiatric Association (2013). Diagnostic and
Artero et al., 2003; Palmer et al., 2003). Moreover, risk fac- Statistical Manual of Mental Disorders, Fifth Edition.
tors and clinical ­presentation of MCI may be different for Washington, DC: American Psychiatric Association.
men and women, such that greater specificity in detection Aretouli, E. and Brandt, J. (2010). Everyday functioning
may be achieved if sex-specific diagnostic algorithms are in mild cognitive impairment and its relationship with
proposed (Artero et al., 2008; Ritchie et al., 2010). executive cognition. International Journal of Geriatric
It has also been suggested that the addition of biological Psychiatry, 25: 224–233.
markers to definitions of MCI may also improve prediction. Artero, S., Ancelin, M.L., Portet, F. et al. (2008). Risk pro-
For example, longitudinal population studies have consis- files for mild cognitive impairment and progression to
tently shown that severe white matter lesions significantly dementia are gender specific. Journal of Neurology,
increase the risk of developing MCI and dementia, particu- Neurosurgery and Psychiatry, 79: 979–984.
larly, when located in temporal regions (Mortamais et al., Artero, S., Petersen, R., Touchon, J., and Ritchie, K.
2013; 2014). (2006). Revised criteria for mild cognitive impairment:
Markers more closely associated with onset of AD, nota- Validation within a longitudinal population study.
bly amyloid beta protein and tau ratios in cerebrospinal Dementia and Geriatric Cognitive Disorders, 22:
fluid also appear to improve MCI case identification and 465–470.
prognostic ability (Frankfort et al., 2008) along with less Artero, S., Tierney, M.C., Touchon, J., and Ritchie, K.
disease-specific markers such as acute-phase proteins, cyto- (2003). Prediction of transition from cognitive impair-
kines, isoprostanes, homocysteine and cholesterol (Song ment to senile dementia: A prospective longitudinal
et al., 2009). With regard to genetic markers, apart from the study. Acta Psychiatrica Scandinavica, 107: 390–393.
well-established association of MCI with the ApoE ε4 allele, Artero, S., Touchon, J., and Ritchie, K. (2001). Disability
epigenetic regulators including microRNA have also been and mild cognitive impairment: A longitudinal popu-
proposed as a means of better identification of MCI cases lation-based study. International Journal of Geriatric
with high risk of evolution towards dementia (Sheinerman Psychiatry, 16: 1092–1097.
et al., 2012). Many of these biomarkers, however, have high- Black, S.E. (1999). Can SPECT predict the future for
est discriminability at preclinical stages when it may be mild cognitive impairment? Canadian Journal of
difficult to demonstrate the cognitive deficit, which is cen- Neurological Sciences, 26 (1): 4–6.
tral to the notion of MCI. To this end, much finer tests will Blackford, R.C. and La Rue, A. (1989). Criteria for diagnos-
therefore need to be developed, focusing on the earliest sites ing age associated memory impairment: Proposed
of brain marker change, such as the posterior hippocampus improvements from the field. Developmental
and entorhinal cortex. Neuropsychology, 5: 295–306.
While the recognition of MCI as a potentially patho- Bowen, J., Teri, L., Kukull, W. et al. (1997). Progression
logical entity marks an early step towards the recognition to dementia in patients with isolated memory loss.
of non-dementia cognitive disorder as an important clini- Lancet, 349: 763–765.
cal problem with a potential treatment, it does not meet the Bozoki, A., Giordani, B., Heidebrink, J.L. et al. (2001). Mild
necessary validation criteria for a formal nosological entity cognitive impairments predict dementia in nonde-
(Ritchie et al., 2001). The feasibility of wide-scale treatment mented elderly patients with memory loss. Archives of
will very much depend on the development of precise diag- Neurology, 58 (3): 411–416.
nostic criteria. Premature application of MCI criteria for the Celsis, P., Agniel, A., Cardebat, D. et al. (1997). Age-
identification of subjects for clinical trials is likely to lead to related cognitive decline: A clinical entity? A longi-
either the inclusion of high numbers of non-cases (yielding tudinal study of cerebral blood flow and memory
high failure rates and subsequent discreditation of a treat- performance. Journal of Neurology, Neurosurgery and
ment, which may have been successful on sample subgroups) Psychiatry, 62 (6): 601–608.
or alternatively by restriction to subjects already manifest- Christensen, H., Henderson, A.S., Jorm, A.F. et al. (1995).
ing early dementia in reality, in which case it may be too ICD-10 mild cognitive disorder: Epidemiological
late to demonstrate potential preventive/delaying effects. evidence on its validity. Psychological Medicine, 25:
The future of MCI development will depend very much on 105–120.
424 Dementia

Crook, T., Bartus, R.T., Ferris, S.H. et al. (1986). Age associ- La Rue, A. and Jarvik, L.F. (1987). Cognitive function
ated memory impairment: Proposed diagnostic criteria and prediction of dementia in old age. International
and measures of clinical change – Report of a National Journal of Aging and Human Development, 25: 79–89.
Institute of Mental Health Work Group. Developmental Levy, R. on behalf of the Aging-Associated Cognitive
Neuropsychology, 2: 261–276. Decline Working Party of the International
Devanand, D.P., Folz, M., Gorlyn, M. et al. (1997). Psychogeriatric Association in collaboration with the
Questionable dementia: Clinical course and predictors World Health Organization (1994). Aging-associated
of outcome. Journal of the American Geriatrics Society, cognitive decline. International Psychogeriatrics, 6 (1):
45: 321–328. 63–68.
Di Carlo, A., Baldereschi, M., Amaducci, L. et al. (2000). Lopez, O.L., Jagust, W.J., Dulberg, C. et al. (2003).
Cognitive impairment without dementia in older people: Risk factors for mild cognitive impairment in the
Prevalence, vascular risk factors, impact on disability. Cardiovascular Health Study Cognition Study: Part 2.
Journal of the American Geriatrics Society, 48: 775–782. Archives of Neurology, 60 (10): 1394–1399.
Flicker, C., Ferris, F.H. and Reisberg, B. (1991). Mild cogni- Matthews, F.E., Stephan, B.C., McKeith, I.G. et al. (2008).
tive impairment in the elderly: Predictors of dementia. Two-year progression from mild cognitive impairment
Neurology, 41: 1006–1009. to dementia: To what extent do different definitions
Frankfort, S.V., Tulner, L.R., van Campen, J.P. et al. (2008). agree? Journal of the American Geriatrics Society, 56:
Amyloid beta protein and tau in cerebrospinal fluid 1424–1433.
and plasma as biomarkers for dementia: A review of McKelvey, R., Bergman, H. and Stern, J. (1999). Lack of
recent literature. Current Clinical Pharmacology, 3: prognostic significance of SPECT abnormalities in
123–131. elderly subjects with mild memory loss. The Canadian
Graham, J.E., Rockwood, K., Beattie, E.L. et al. (1997). Journal of Neurological Sciences, 26: 23–28.
Prevalence and severity of cognitive impairment with Morris, J.C., Storandt, M., Miller, J.P. et al. (2001). Mild
and without dementia in an elderly population. Lancet, cognitive impairment represents early-stage Alzheimer
349: 1793–1796. disease. Archives of Neurology, 58: 397–405.
Hogan, D.B. and Ebly, E.M. (2000). Predicting who will Mortamais, M., Portet, F., Brickman, A.M., et al. (2014).
develop dementia in a cohort of Canadian seniors. Education modulates the impact of white matter
Canadian Journal of Neurological Sciences, 27: 18–24. lesions on the risk of mild cognitive impairment and
Huang, C., Wahlund, L.O., Dierks, T. et al. (2000). dementia. American Journal of Geriatric Psychiatry,
Discrimination of Alzheimer’s disease and mild 22: 1336–1345.
cognitive impairment by equivalent EEG sources: Mortamais, M., Reynes, C., Portet, F. et al. (2013). Spatial
A cross-sectional and longitudinal study. Clinical distribution of cerebral white matter lesions predicts
Neurophysiology, 111: 1961–1967. progression to mild cognitive impairment and demen-
Johanssen, B. and Zarit, S.H. (1997). Early cognitive mark- tia. PLoS ONE, 8 (2): e56972. doi: 10.1371/journal.
ers of the incidence of dementia and mortality: A pone.0056972
longitudinal population-based study of the oldest old. Palmer, K., Fratiglioni, L. and Winblad, B. (2003). What is
International Journal of Geriatric Psychiatry, 12: 53–59. mild cognitive impairment? Variations in definitions
Johnson, K.A., Jones, K. and Holman, B.L. (1998). and evolution of nondemented persons with cognitive
Preclinical prediction of Alzheimer’s disease using impairment. Acta Neurologica Scandanavica, 179: 14–20.
SPECT. Neurology, 50: 1563–1572. Petersen, R., Doody, R., Kurz, A. et al. (2001). Current
Kluger, A., Ferris, S.H., Golomb, J. et al. (1999). concepts in mild cognitive impairment. Archives of
Neuropsychological prediction of decline to dementia Neurology, 58: 1985–1992.
in nondemented elderly. Journal of Geriatric Psychiatry Petersen, R.C., Smith, G.E., Waring, S.C. et al. (1997).
and Neurology, 12: 168–179. Aging, memory and mild cognitive impairment.
Kluger, A., Gianutsos, J.G., Golomb, J. et al. (1997). International Psychogeriatrics, 9: 65–69.
Motor/psychomotor dysfunction in normal aging, Petersen, R.C., Smith, G.E., Waring, S.C. et al. (1999). Mild
mild cognitive decline, and early Alzheimer’s dis- cognitive impairment: Clinical characterization and
ease: Diagnostic and differential diagnostic features. outcome. Archives of Neurology, 56: 303–308.
International Psychogeriatrics, 9: 307–316. Richards, M., Touchon, J., Ledésert, B. and Ritchie, K.
Kral, V.A. (1962). Senescent forgetfulness: Benign and (1999). Cognitive decline in ageing: Are AAMI and
malignant. Canadian Medical Association Journal, 86: AACD distinct entities? International Journal of
257–260. Geriatric Psychiatry, 14: 534–540.
Krasuki, J.S., Alexander, G.E., Horwitz, B. et al. (1998). Ritchie, K., Ancelin, M.L., Portet, F. et al. (2010).
Volumes of medial temporal lobe structures in patients Retrospective identification and characterization of
with Alzheimer’s disease and mild cognitive impair- mild cognitive impairment from a prospective popula-
ment (and in healthy controls). Biological Psychiatry, tion cohort. American Journal of Geriatric Psychiatry,
43: 60–68. 18 (8): 692–700.
Mild cognitive impairment (MCI): A historical perspective 425

Ritchie, K., Artero, S. and Touchon, J. (2001). Classification Touchon, J. and Ritchie, K. (1999). Prodromal cognitive
criteria for mild cognitive impairment: A population- disorder in Alzheimer’s disease. International Journal
based validation study. Neurology, 56: 37–42. of Geriatric Psychiatry, 14: 556–563.
Ritchie, K., Leibovici, D., Ledésert, B. and Touchon, J. Visser, P.J., Verhey, F.R., Hofman, P.A. et al. (2002).
(1996). A typology of sub-clinical senescent cogni- Medial temporal atrophy predicts Alzheimer’s dis-
tive disorder. British Journal of Psychiatry, 168 (4): ease in patients with minor cognitive impairment.
470–476. Journal of Neurology, Neurosurgery and Psychiatry,
Sheinerman, K.S., Tsivinsky, V.G., Crawford, F. et al. (2012). 72: 491–497.
Plasma microRNA biomarkers for detection of mild Wada-Isoe, K., Uemura, Y., Nakashita, S. et al. (2012).
cognitive impairment. Aging, 4 (9): 590–605. Prevalence of dementia and mild cognitive impairment
Schaie, K.W. and Willis, S.L. (1991). Adult personality in the rural island town of Ama-cho, Japan. Dementia
and psychomotor performance: Cross-sectional and and Geriatric Cognitive Disorders Extra, 2 (1): 190–199.
longitudinal analyses. Journals of Gerontology, 46: Ward, A., Arrighi, H.M., Michels, S. and Cedarbaum,
275–284. J.M. (2012). Mild cognitive impairment: Disparity of
Stephan, B.C., Brayne, C. and McKeith, I.G. (2008). Mild incidence and prevalence estimates. Alzheimer’s and
cognitive impairment in the older population: Who is Dementia: The Journal of the Alzheimer’s Association,
missed and does it matter? International Journal of 8: 14–22.
Geriatric Psychiatry, 23: 863–871. Wolf, H., Grunwald, M., Ecke, G.M. et al. (1998). The
Song, F., Poljak, A., Smythe, G.A. and Sachdev, P. (2009). prognosis of mild cognitive impairment in the elderly.
Plasma biomarkers for mild cognitive impairment Journal of Neural Transmission, 54: 31–50.
and Alzheimer’s disease. Brain Research Reviews, 61: World Health Organization (1993). The ICD-10
69–80. Classification of Mental and Behavioural Disorders.
Swift, J. (1726). Gulliver’s Travels. London, UK: Benjamin Diagnostic Criteria for Research. Geneva, Switzerland:
Motte, p. 223. World Health Organisation.
Tierney, M.C., Szalai, J.P., Snow, W.G. et al. (1996). Zaudig, M. (1992). A new systematic method of measure-
Prediction of probable Alzheimer’s disease in memory- ment and diagnosis of ‘mild cognitive impairment’ and
impaired patients: A prospective longitudinal study. dementia according to ICD-10 and DSM III-R criteria.
Neurology, 46: 661–665. International Psychogeriatrics, 4: 203–219.
42
Clinical characteristics of mild cognitive
impairment

YONAS E. GEDA, JANINA KRELL-ROESCH, ANNA PINK, KATHLEEN A. SPANGEHL AND


RONALD C. PETERSEN

Reisberg et al. were perhaps the first to coin the term MCI
42.1 INTRODUCTION (Reisberg et al., 1982; Flicker et al., 1991; Reisberg et al., 2008).
They defined MCI using the Global Deterioration Scale (GDS).
The promotion of health and prevention of disease is a time The GDS is an ordinal scale of 1 to 7 with 1 being normal and
honoured axiomatic truth (Geda et al., 2006). This is partic- 7 signifying severe dementia. A GDS score of 3 is defined as
ularly true of Alzheimer’s disease (AD), the most common MCI. In 1962, V.A. Kral used the term senescent forgetfulness
cause of dementia (Chong and Sahadevan, 2005). as the first attempt to characterize memory concerns with age-
It is crucial to delay the onset of dementia, one of the lead- ing. An initiative of the National Institute of Mental Health
ing causes of mortality and morbidity in late-life for compel- coined the term ‘age-associated memory impairment (AAMI)’
ling reasons (see Chapters 2, 3, 7 and 15). If it were possible (Crook et al., 1986). The concept of AAMI further accelerated
to delay the onset of dementia by about 2 years, the global the research work in the grey zone between ageing and demen-
prevalence of AD would decrease by 22.8 million cases. tia. However, AAMI was noted to have limitations, such as its
Effective prevention, as well as intervention, could dramati- ­restriction to memory domain only. Moreover, AAMI was
cally reduce disease onset and progression. The identification based on a comparison of memory function in older adults to
of mild cognitive impairment (MCI) contributes to the early that of young adults (at least one standard deviation below the
detection and prevention effort (Petersen et al., 1999b). mean for young adults). Such limitations led some to remark that,
MCI refers to an intermediate stage between the cognitive depending on the memory test selected, up to 90% of those older
changes of normal ageing and very early dementia (Petersen than 50 years would be labelled as impaired (Smith et al., 1991).
et al., 2005) (Figure 42.1). Individuals with MCI show cog- To address the weaknesses of AAMI, the International
nitive impairment greater than expected for their age, but Psychogeriatric Association defined the term ‘age-asso-
otherwise function independently and do not meet the com- ciated cognitive decline (AACD)’ (Levy, 1994). AACD
monly accepted criteria for dementia (Petersen et al., 1999b). addressed memory decline plus criteria for a variety of
cognitive domains presumed to decline in normal ageing.
Age and education adjusted normative values were also
42.2 HISTORICAL GENESIS OF MCI described. In addition to AAMI and AACD several other
terms and definitions were proposed (Blackford and La Rue,
In the past decades, a growing research effort has been 1989; Graham et al., 1997) including ‘questionable demen-
devoted to the identification of clinical and epidemiologi- tia’ (Devanand et al., 1997) as defined by a score of 0.5 on
cal characteristics as well as probable causes of dementia. the Clinical Dementia Rating Scale (CDR) (Morris, 1993).
During this progress, clinicians and investigators observed While both GDS and CDR are reliable scales when rating
some elderly persons who were neither demented nor cogni- severity of MCI along the continuum of cognitive impair-
tively normal for their age. This observation prompted them ment, they do not necessarily coincide with various stages
to investigate the intermediate stage between intact cogni- of impairment. Actually, patients with a GDS of 3 or CDR of
tive functioning and clinical dementia. 0.5 may in fact fulfil criteria for mild dementia or AD.

426
Clinical characteristics of mild cognitive impairment 427

Normal the combination of both clinical features and biomarker


measures for a diagnosis of MCI due to AD. Petersen et al.
noted that the NIA-AA criteria and especially the newly
added biomarker criteria are valid in most MCI subjects
Mild cognitive in community and clinical trial settings. However, a num-
impairment ber of subjects also showed conflicting biomarker results
inconsistent with the proposed AD model i.e. neurodegen-
eration without amyloid deposition (Petersen et al., 2013).
The reader is referred to the Section 42.5 of this chapter for
Alzheimer’s disease
more details.

Figure 42.1 Conceptual model of mild cognitive


42.2.4 MILD NEUROCOGNITIVE
­impairment (MCI). DISORDERS (THE NEW DSM-5
CRITERIA)
Out of the different constructs and terms proposed during
In the fifth edition of the Diagnostic and Statistical Manual
the past decades, MCI is probably the most empirically inves-
of Mental Disorders (DSM-5), the American Psychiatric
tigated construct as evidenced by the exponential increase
Association proposes a new construct of ‘Neurocognitive
in publications and citation numbers pertaining to MCI. In
Disorders (NCD)’, which covers the three entities: delir-
addition, the American Academy of Neurology (AAN) stated
ium, major NCD and mild NCD (American Psychiatric
that MCI is worthy of attention since individuals with MCI
Association and American Psychiatric Association DSM-5
have a high rate (10%–15% per year) of developing dementia
Task Force, 2013). The mild NCD criteria are similar to the
compared to the rate for the general population of 1%–2%
international criteria of MCI. In fact, the DSM-5 discusses
per year (Petersen et al., 2001). The criteria for amnestic MCI
the epidemiology and diagnostic markers of mild NCD
(aMCI) were first proposed in 1999 following an empirically
by drawing congruence between MCI and mild NCD. The
validated prospective and population-based cohort study
main difference between the two constructs is that mild
in Rochester, Minnesota (Petersen et al., 1999a). A num-
NCD is defined as acquired cognitive disorders pertinent
ber of factors facilitated the development of this construct.
to all age groups, whereas MCI is based on research done
Researchers noted: (1) the existence of an intermediate zone
in the context of geriatric populations, although age is not
of cognitive impairment not captured by a clinical definition;
a part of the MCI definition. Mild NCD may improve the
(2) a rising importance of dementia for public health; (3) an
reliability of diagnoses; however, it has yet to withstand the
emerging clinical need for a construct beyond the binary
empirical scrutiny needed to be considered as a valid con-
diagnosis of present/absent dementia (Petersen et al., 2014)
struct (Stokin et al., 2015).
and (4) a diagnosis of MCI allows both, patients and caregiv-
ers, to plan for future familial challenges while the patient is
still able to make conscious decisions. The Mayo Clinic core
criteria published in 1999 were mainly focused on memory 42.3 CLINICAL FEATURES
impairment. The core required the presence of self or infor-
mant reported memory complaints and objective memory
impairment with essentially preserved cognitive functioning. Physicians often encounter a clinical scenario in which an
In Stockholm, Sweden 2003, a Key Symposium of experts elderly person presents with memory concerns. The patient
in the field of cognitive ageing revised the Mayo Clinic crite- is often interested in finding out whether AD is a possible
ria. The newly developed International criteria of MCI were diagnosis. While such concerns could represent other states
broadened to include other areas of cognitive impairment such as late-life depression or the so-called ‘worried-well’
and were no longer restricted to memory. This revision led state, memory concerns may also be suggestive of an evolv-
to a new understanding of MCI, which became a clinical ing AD. Consider, for example, a typical clinical scenario
syndrome with multiple clinical profiles due to various aeti- described below.
ologies (Petersen et al., 2014).
42.3.1 CASE RECORD OF PATIENT A
42.2.3 MCI DUE TO AD (NATIONAL
INSTITUTE ON AGING [NIA]– A 73-year-old right-handed male patient presents with
ALZHEIMER’S ASSOCIATION [AA] forgetfulness for recent events and future engagements.
CRITERIA) Family members and close friends notice these changes
as well. The patient has difficulty identifying the onset
In 2011, a work group of investigators from the NIA and of these symptoms, but felt that they started insidi-
the AA published guidelines for the diagnosis of MCI due ously and progressed gradually over a period of 2–3
to AD (Albert et al., 2011). The work group recommended years. Otherwise, he is living independently and has
428 Dementia

no difficulty carrying out activities of daily living, such If these conditions are met, a diagnosis of MCI may
as handling his own finances, cooking and driving. He be assumed and the clinician can proceed to identify the
denies depression, stress or other complicating medi- number and types of cognitive domains impaired. A diag-
cal issues. He requests an appointment with a physician nosis of aMCI-single domain is assumed if the impairment
in order to determine if this memory problem should involves only memory domain. aMCI-multiple domain per-
be pursued further. The clinical evaluation i.e. meticu- tains to impairments in the memory domain plus at least
lous history and physical examination, including bed- one other cognitive domain, such as language, executive
side cognitive screening using the short test of mental function or visuospatial skills. Likewise, a diagnosis of non-
status, is suggestive of cognitive impairment but not amnestic MCI (naMCI)-single domain is assumed if there
severe enough to warrant the diagnosis of dementia, is impairment in a single non-memory domain, whereas
hence a clinical diagnosis of MCI is made. Investigations naMCI-multiple domain refers to impairments in multiple
including psychometric testing and magnetic resonance non-memory domains.
imaging (MRI) are ordered. The neuropsychological test- Figure 42.3 depicts the presumed aetiologies along
ing confirms the clinical diagnosis. It reveals memory with clinical subtypes of MCI. The single- and multiple-
impairment, particularly on measures of learning and domain aMCI subtypes with presumed degenerative aeti-
delayed recall beyond what is felt to be normal for age; ology likely represent a prodromal form of AD (Winblad
but other cognitive domains such as language and visuo- et al., 2004). The non-amnestic subtypes that emphasise
spatial skills are relatively intact. An MRI of the head impairments in other cognitive domains aside from mem-
revealed mild hippocampal atrophy. ory may have a higher likelihood of progressing to a non-
AD dementia, e.g. dementia with Lewy bodies (Winblad
et al., 2004).
42.3.2 COMMENT
This patient probably has aMCI. He is becoming slightly
more forgetful, and this is noticeable to his family and
friends. The most salient feature of the history concerns 42.4 EPIDEMIOLOGY OF MCI
forgetfulness of insidious onset that gradually progresses
over a year or so. All other cognitive domains i.e. language, Three epidemiological indices are derived from descriptive
comportment-executive function and visuospatial skills studies of MCI: the prevalence, the incidence and the con-
are intact. The individual does not have a decline in func- version rate of MCI to dementia. Numerous studies have
tion. This finding likely represents an early disease process been conducted to investigate both the prevalence and inci-
involving the medial temporal lobe since meaningful infor- dence of MCI.
mation can no longer be encoded in an efficient manner, nor The Cardiovascular Health Study (CHS) was perhaps the
recalled well. first population-based study to estimate the prevalence of
MCI subtypes. The study was originally designed to exam-
42.3.3 DIAGNOSTIC ALGORITHM FOR ine cardiovascular risk factors (Fried et al., 1991). The CHS
MCI SUBTYPES reported an overall prevalence of MCI to be 22% with aMCI
accounting for 6% and multi-domain MCI representing
The diagnosis of MCI is anchored upon clinical data, i.e. 16% (Lopez et al., 2003).
detailed history, thorough physical ­examination and bed- Researchers in Japan retrofitted the MCI criteria in a
side cognitive screening, e.g. Mini-Mental State Examination cohort of over 65-year-olds and estimated the prevalence of
(MMSE) (Folstein et al., 1975) or equivalent scales (Kokmen MCI to be in the range of 1.7%–16.6% (Sasaki et al., 2009).
et al., 1987) to objectively confirm self- or informant- Retrofitting the MCI criteria might have introduced mea-
reported cognitive complaints. Even though aMCI is the surement bias in the Japanese study. An Australian research
most widely studied and empirically validated construct, group found the prevalence of MCI to be 3.8% and AACD
additional subtypes are also the subject of intense investiga- to be 3.1% in subjects 60–64 years old (Kumar et al., 2005).
tion (Winblad et al., 2004). Although the Australian study design was optimal, the
Figure 42.2 depicts the diagnostic algorithm used to cohort involved a younger age group, thus the prevalence
identify a particular subtype of MCI. This procedure is rate was understandably low.
initiated when a patient or informant, usually the spouse, A population-based study utilizing the operational crite-
reports cognitive complaints such as forgetfulness for recent ria of MCI in the elderly is likely to minimize sampling and
events and future engagements. The clinician should first measurement bias, thus leading to more precise estimates
determine the patient to be neither demented nor of normal of prevalence and incidence rates. One such example is the
cognition for age and education. Next there should be no population-based Mayo Clinic Study of Aging (MCSA)
substantial functional decline or if present, the functional launched in Olmsted County, Minnesota on the prevalence
decline should not be of sufficient magnitude as to warrant date of 1 October 2004 (Roberts et al., 2008). The preva-
the diagnosis of very mild dementia. lence of MCI from this study is estimated at approximately
Clinical characteristics of mild cognitive impairment 429

Mild cognitive impairment


Cognitive complaint

Not normal for age


Not demented
Cognitive decline
Essentially normal functional activities

MCI

Yes Memory impaired? No

Amnestic MCI Non-amnestic MCI

Single non-memory
Memory
Yes No Yes cognitive domain No
impairment only?
impaired?

Amnestic MCI Amnestic MCI Non-amnestic MCI Non-amnestic MCI


single domain multiple domain single domain multiple domain

Figure 42.2 Flow chart of decision process for making diagnosis of subtypes of MCI.

Aetiology
Degenerative Vascular Psychiatric Trauma

Single
AD Depression
domain

Amnestic
MCI
Clinical classification

Multiple
AD VaD Depression
domain

Single
FTD
domain
Non-
amnestic
MCI
Multiple
DLB VaD
domain

Figure 42.3 Classification of clinical subtypes of MCI with presumed aetiology.

15%, with a 2:1 ratio of aMCI to naMCI (Petersen, 2009). 42.4.1 INCIDENCE OF MCI
Prevalence of MCI increased with age, was found to be
higher in men than women, in those who were not mar- The incidence of MCI ranges from 1% to 6% per year
ried and in carriers of the apoprotein E-ε4 (APOE-ε4) allele while prevalence estimates range from 3% to 22% per year
(Petersen et al., 2010). Prevalence of MCI decreased, how- (Bennett et al., 2002; Hanninen et al., 2002; Larrieu et al.,
ever, with increasing years of education (Petersen et al., 2002; DeCarli, 2003; Lopez et al., 2003; Ganguli et al., 2004).
2010). A recent review of the literature by experts in the field This variability can in part be explained by sampling and
indicated that the prevalence of MCI could be as high as measurement biases (Sackett, 1979). Sampling methods that
18.9%, as measured by the expanded Mayo Clinic criteria may have yielded variability include using advertisements to
(Petersen et al., 2014). recruit research participants (introducing a non-respondent/
430 Dementia

volunteer bias) (Sackett, 1979). Measurement bias may be the Benton Visual Retention Test (Benton, 1983). Despite
introduced when retrofitting MCI criteria into a cohort this, recent evidence indicates that an MCI diagnosis
assembled for non-MCI study. Furthermore, older research (with or without reversion to cognitively normal status)
mainly focused on the Mayo Clinic core criteria of MCI is associated with an increased risk of dementia (Roberts
and was restricted to memory impairment, whereas, recent et al., 2014).
investigations utilise the expanded MCI criteria to include
memory impairment plus other cognitive domains. Due to 42.4.2 NEUROPSYCHIATRIC FEATURES
the above heterogeneity it is not surprising that prevalence
and incidence rates can vary significantly. Although the neurological, psychometric and neuroimag-
Past incidence estimates of MCI were primarily based ing aspects of MCI have been researched (Petersen et al.,
on clinical samples. However, in the past few years, sev- 1999a; Jack et al., 2000; Jack et al., 2009), there has been
eral population-based studies have been conducted to little investigation in the neuropsychiatric aspect of MCI. It
estimate the incidence of MCI. For example, the MCSA is reasonable to address this issue because the medial tem-
conducted a population-based prospective cohort study poral lobe and its connections with the prefrontal cortex
in i­ndividuals aged 70 years and older and reported and other structures not only play a critical role in cognitive
an incidence rate of 63.6 per 1000 person-years, with a function but may also be involved in emotional behaviour
higher rate in men (72.4) than in women (57.3) (Roberts (Mesulam, 1998). The first population-based investigation
et al., 2012). of neuropsychiatric symptoms of MCI was reported in 2002
In addition to studies on the progression from normal (Lyketsos et al., 2002). Since then, there have been similar
ageing to incident MCI, investigators have also examined studies on these features involving samples from primary
several studies that have estimated the progression rate of or tertiary care settings (Feldman et al., 2004; Geda et al.,
MCI to dementia (Flicker et al., 1991; Tierney et al., 1996; 2004; Hwang et al., 2004; Beaulieu-Bonneau and Hudon,
Petersen et al., 1999a; Daly et al., 2000; Petersen et al., 2005; 2009; Edwards et al., 2009).
Fischer et al., 2007). The findings vary depending upon the The MCSA has reported the population-based preva-
study design and measurement instruments used. Researchers lence of neuropsychiatric symptoms in normal ageing and
from Harvard University recruited study participants via MCI. The investigators observed that the three most com-
advertisement. They prospectively followed a cohort of sub- mon neuropsychiatric symptoms in MCI were depression
jects with MCI and reported a conversion rate of 6% per (27%), apathy (18.5%) and irritability (19.4%) (Geda et al.,
year (Daly et al., 2000). Alternatively, a recent multicentre, 2008). Lyketsos et al. (2002) reported similar, but slightly
randomized, double-blind, placebo-controlled clinical trial smaller frequencies of depression (20%), apathy (15%) and
reported a conversion rate of 16% per year (Petersen et al., irritability (15%) in the CHS. The CHS group suggested that
2005). A further community-based birth cohort study exam- selection bias might have led to an underestimation of their
ining progression rates between amnestic and non-amnestic prevalence estimates. This bias may account for the differ-
subtypes of MCI to AD showed the annual rate of conver- ences in crude frequency rates across the two studies. The
sion to AD for aMCI to be 19%, compared to almost 11% for Cache County population-based study reported prevalence
naMCI subjects (Fischer et al., 2007). Researchers from the figures for depression (7.2%), apathy (3.2%) and irritabil-
Karolinska Institute in Sweden reported in 2008 that 33.3% ity of (4.6%) in cognitively normal persons (Lyketsos et al.,
of aMCI patients, 10% of naMCI-single domain patients and 2000). The Mayo Clinic group did observe slightly higher
75.0% of naMCI-multiple domain patients progressed to AD figures for depression (11.5%), apathy (4.8%) and irritability
after 3 years of follow-up (Palmer et al., 2008). Hence, the (7.6%) in normal ageing (Geda et al., 2008).
rather small rate reported by the Harvard group could be
attributed to non-respondent/volunteer bias (Sackett, 1979). 42.4.3 INCIDENCE OF MCI BY BASELINE
The higher percentages from the community-based cohort NEUROPSYCHIATRIC STATUS
study may be due to prevalence/incidence bias where in MCI
subjects with advanced cognitive impairment (prevalent There are limited data on the incidence of MCI as predi-
MCI) may have higher conversion rates to dementia (Fischer cated by baseline neuropsychiatric symptoms (Wilson
et al., 2007; Mitchell and Shiri-Feshki, 2008). The common et al., 2007; Brodaty et al., 2012; Geda et al., 2014). The
theme in all the studies is that MCI c­ onstitutes a high-risk Sydney Memory and Ageing Study reported that agitation
group for dementia. and anxiety predicted cognitive decline in 873 individuals
There is an ongoing controversy regarding the ‘insta- aged 70–90 years. (Brodaty et al., 2012). The Chicago Health
bility’ of the MCI construct (Ritchie et al., 2001; Larrieu and Ageing Project reported that the ‘distress-prone’ elderly
et al., 2002). Larrieu et al. from France reported a rever- subjects were more likely to develop incident MCI as com-
sion rate (i.e. from MCI back to normal cognitive ageing) pared to those who were less distress prone (Wilson et al.,
to be as high as 40% over a 2–3 years follow-up. However, 2007). Recently, the MCSA prospectively followed 1587 cog-
one major limitation of their study was how they defined nitively normal elderly (70 years and older) to the outcome
MCI, based on only one single memory measurement, of incident MCI, as predicted by baseline neuropsychiatric
Clinical characteristics of mild cognitive impairment 431

symptoms. The investigators observed that baseline psychi- the four theoretical models are not mutually exclusive
atric symptoms were of similar or greater magnitude as bio- and may interact. Furthermore, these proposed theoreti-
markers (genetic and structural MRI) in predicting the risk cal models remain to be hypothetical and speculative until
of incident MCI (Geda et al., 2014). confirmed by mechanistic research (Geda et al., 2013)
(Figure 42.4).
42.4.4 PROPOSED MECHANISMS LINKING
NEUROPSYCHIATRIC SYMPTOMS
WITH COGNITIVE OUTCOMES 42.5 BIOMARKERS
In the past, the following four possible theoretical models
that may explain the link between neuropsychiatric symp- The neurodegenerative process of AD likely precedes the
toms and MCI/AD have been proposed (Geda et al., 2013): development of clinical manifestations of AD by several
(1) neuropsychiatric symptoms as ‘risk factors’: a specific years if not decades (Braak and Braak, 1991; Shaw et al.,
neuropsychiatric symptom may have a direct deleterious 2007). About 30% of cognitively normal elderly subjects
effect on the brain and therefore lead to MCI/AD; (2) shared meet criteria for AD based on their pathology at autopsy
risk factor or confounding pathway: a biological risk factor (Price and Morris, 1999; Knopman et al., 2012). Biomarkers
for MCI/AD may lead to both cognitive outcome and neu- can measure pathological processes in vivo. The most
ropsychiatric symptoms; (3) a synergistic interaction: a neu- common underlying pathological process in MCI is AD.
ropsychiatric symptom and a biological factor may interact Biomarkers in AD can provide evidence of AD pathology
to have a synergistic effect of elevating the risk of MCI/AD; in MCI via markers of amyloid deposition and neurode-
(4) reverse causality: a subject may show reactive depression generation (Albert et al., 2011). The most widely used bio-
and/or other neuropsychiatric symptoms after the person markers are neuroimaging and cerebrospinal fluid (CSF)
starts noticing cognitive decline. It should be noted that derived measures. Biomarkers of amyloid positron emission

(a) Neuropsychiatric Unknown Brain DAT/MCI


symptoms mechanism damage

Neuropsychtric
(b) symptoms
Susceptibility
gene variant
or other
risk factor DAT/MCI

(c) Preclinical Neuropsychatric


DAT/MCI DAT/MCI
symptoms

(d)
Susceptibility
gene variant Interaction DAT/MCI
or other
risk factor

Neuropsychiatric
symptoms

Figure 42.4 Four hypotheses of the possible mechanisms linking depression to MCI: (a) Aetiologic pathway, (b) confound-
ing, (c) reverse casualty and (d) interaction.
432 Dementia

tomography (PET) imaging or amyloid β42 (Aβ42) mea- demonstrated by studies showing that positive amyloid scans
surement in CSF provide direct evidence of amyloid deposi- correlate with low CSF Aβ42 (Fagan et al., 2006; Grimmer
tion in the brain. In contrast, tau deposition in the form of et al., 2009; Jagust et al., 2009; Tolboom et al., 2009) Studies
neurofibrillary tangles (NFT) may reflect neurodegenera- have shown that low CSF Aβ42 has been associated with AD
tion. Other measures of neurodegeneration include brain diagnosis at autopsy (Clark et al., 2003, Strozyk et al., 2003).
atrophy as measured by MRI, and brain hypometabolism as In addition, low CSF Aβ42 has been shown to predict con-
measured by fluorodeoxyglucose-PET (FDG-PET). version of MCI to dementia (Riemenschneider et al., 2002;
Hansson et al., 2006, Mattsson et al., 2009).
42.5.1 BIOMARKERS OF AMYLOID AND
TAU DEPOSITION 42.5.2 BIOMARKERS OF
NEURODEGENERATION
The plaques that are formed during the pathologic process
of AD consist of extracellular deposits of amyloid β-protein
(Aβ), which primarily form filamentous fibrils. This process
42.5.2.1 Magnetic resonance imaging
can be detected via imaging (Clark et al., 2011) and CSF bio-
markers of amyloid deposition. Structural MRI can measure cerebral atrophy. Cerebral
atrophy is associated with neuronal loss (Bobinski et al.,
42.5.1.1 PET amyloid imaging 2000). This can be shown in volumetric MRI studies investi-
gating hippocampal atrophy, ventricular volume expansion,
The development of the amyloid-imaging PET tracer, whole brain atrophy or cortical thinning and may reflect
Pittsburgh Compound-B (PiB), is an important contribution neurodegeneration. Furthermore, MRI studies have been
to the study of ageing (Klunk et al., 2004). A few studies have shown to predict future progression to dementia in subjects
examined the serial properties of PiB over time (Klunk et al., with MCI (Jack et al., 1999; Bakkour et al., 2009).
2006; see Chapter 12. Current data suggest that 20%–30%
of cognitively normal subjects have positive PiB scans while
about 60% of MCI subjects have positive scans (Lopresti et al., 42.5.2.2 Fluorodeoxyglucose-PET
2005). Furthermore, another PET tracer, 18F-florbetapir, has
The metabolic activity of the brain is associated with synaptic
a relatively long radioactive half-life, and this in part has con-
activity (Schwartz et al., 1979; Attwell and Laughlin, 2001).
tributed for this agent to get Food and Drug Administration
Therefore, hypometabolism as measured by FDG-PET may
(FDA) approval (Wong et al., 2010).
indicate neurodegeneration. Subjects with AD show a charac-
AD patients show widespread cortical amyloid uptake
teristic pattern of hypometabolism in bilateral temporopari-
with the most prominent uptake in the frontal cortex and
etal regions, including the posterior cingulate and precuneus
precuneus followed by the parietal and temporal cortices
(Hoffman et al., 2000; Jagust et al., 2007). Hypometabolism
(Kemppainen et al., 2006). A positive PiB (Wolk et al., 2009;
in temporoparietal regions also predicts conversion from
Villemagne et al., 2011) or 18F-florbetapir (Doraiswamy
MCI to dementia (Chetelat et al., 2003; Landau et al., 2010).
et al., 2012; Landau et al., 2012) amyloid imaging scan is
associated with progression from MCI to AD. It should be
noted, however, that more research is needed on this area, to 42.5.2.3 CSF tau
develop precise prediction models.
Tau protein in healthy individuals is involved in the out-
growth of neuronal processes by promoting and stabiliz-
42.5.1.2 Tau imaging ing microtubules. Abnormal phosphorylation affects the
binding affinity of tau resulting in neuronal aggregations of
Recently, investigations of tauopathy, employing tau imaging NFT. High CSF phosphorylated tau (P-tau) correlates with
ligands, has begun. Studies have been conducted in tauopa- NFT (Tapiola et al., 1997; Clark et al., 2003). While total tau
thy mouse models (Maruyama et al., 2013) and preliminary is nonspecific and is increased in various neurologic condi-
studies have also been conducted involving human subjects tions, such as stroke, traumatic brain injury or prion dis-
(Chien et al., 2013). However, tau imaging is currently in its eases (Hesse et al., 2001; Otto et al., 2002; Ost et al., 2006),
rudimentary stage in the investigation of ageing and MCI. P-tau seems more specific to AD and may help in distin-
guishing AD from frontotemporal dementia (Sjogren et al.,
42.5.1.3 CSF amyloid β42 2001) or dementia with Lewy bodies (Parnetti et al., 2001).
Total tau levels have been more frequently studied than
Aβ42 is the primary protein in amyloid plaques. Aβ42 is a P-tau (Blennow and Hampel, 2003). In addition, elevated
42-amino acid peptide formed by mutations on the amyloid levels of CSF total tau and P-tau independently predict
precursor protein (APP) and processing pathways (Hardy conversion from MCI to dementia (Riemenschneider et al.,
and Selkoe, 2002) Amyloid deposition in the brain is asso- 2002; Herukka et al., 2005; Hansson et al., 2006; Mattsson
ciated with reduced levels of CSF amyloid. This has been et al., 2009).
Clinical characteristics of mild cognitive impairment 433

Table 42.1 Clinical trials in mild cognitive impairment

Sponsor Duration End point Treatments


ADCS a 3 years AD Vitamin E Donepezil
Merck 2–3 years AD Rofecoxib
Novartis 3 years AD Rivastigmine
Janssen 2 years Symptoms Galantamine
Pfizer 6 months Symptoms Donepezil
UCB 1 year Symptoms Piracetam
Elan, Myriad, Neurochem and 8 years (median AD Ginkgo biloba (not effective on
GlaxoSmithKline follow-up: 6.1 years) progression rate)
Viatris 1 year Symptoms Vitamin B6
Aerobic exercise
(walking helped improve memory
[men], attention [women])
Department of Veterans Affairs, the 21 days Symptoms Intranasal insulin (retained more verbal
Institute for the Study on Aging and information after a delay)
National Institute on Aging
Abbreviations: AD, Alzheimer’s disease; UCB, Union chimique belge.
a Alzheimer’s Disease Cooperative Study supported by National Institute on Aging, Pfizer, Inc., Eisai, Inc. and Roche Vitamins.

A meta-analysis that examined the diagnostic accuracy


of combining the three CSF biomarkers (i.e. Aβ42, total tau 42.6 TREATMENT
and P-tau) reported that the best predictor of conversion
from MCI to dementia was an abnormal ratio of Aβ42 to There is no standard treatment for MCI (see Chapter 43),
total tau or abnormal levels of both markers; however, the however, as the focus of AD research moves towards preven-
addition of P-tau did not increase diagnostic accuracy (Van tion, numerous clinical trials are being undertaken, since
Rossum et al., 2010). MCI is considered to be the optimum stage for interven-
The field of ageing increasingly emphasizes the use of tional studies to delay the onset of dementia (Chertkow,
biomarkers for research and potential long-term clinical 2002; Bhalla et al., 2009). Table 42.1 outlines the clinical trials
use. In 2011, the NIAand the AA published guidelines for targeting MCI currently under way or recently completed.
the identification of preclinical AD and MCI due to AD The reader is referred elsewhere for a detailed discussion
(NIA–AA criteria) and placed an unprecedented empha- of clinical trials targeting MCI (Petersen et al., 2001; Jelic et
sis on biomarkers of AD pathophysiology. The preclinical al., 2006; Petersen, 2007). One major clinical trial that targeted
AD criteria are the following: Stage 0, no abnormal amyloid MCI was the Alzheimer’s Disease Cooperative Study (ADCS)
(A) or neurodegeneration (N) biomarkers (A−/N−); Stage 1, funded by the NIA, with additional funding from Pfizer, Inc.,
abnormal amyloid only (A+/N−); Stage 2, abnormal amy- Eisai, Inc. and Roche Vitamins. It involved 70 North American
loid and neurodegeneration (A+/N+); and Stage 3, same as medical centres, and was a randomized, double-blind, pla-
Stage 2 plus subtle cognitive changes that do not meet MCI cebo-controlled study involving three arms to assess the safety
criteria (Albert et al., 2011; Jack et al., 2011; McKhann et al., and efficacy of high-dose vitamin E (2000 IU per day) and
2011; Sperling et al., 2011). donepezil (10 mg per day) (Petersen et al., 2005) and was pow-
The authors of a recent study showed that the NIA–AA ered to decrease the conversion rate from MCI to AD from an
criteria could be applied to most MCI subjects; however, a estimated 45% down to 30% over the course of 3 years.
considerable number of subjects had conflicting biomarker Seven hundred and sixty-nine subjects were random-
results inconsistent with the proposed AD model i.e. neu- ized and the annual conversion rate from MCI to AD was
rodegeneration without amyloid deposition. About 23% approximately 16% per year. Over the course of the study,
of cognitively normal subjects showed neurodegeneration donepezil reduced the risk of progressing to AD for the
without evidence of amyloid deposition, and the authors first 18 months of the trial while vitamin E had no thera-
have referred to this entity as suspected non-Alzheimer’s peutic effect. The secondary cognitive measures supported
pathophysiology (SNAP) (Jack et al., 2012; Petersen et al., the overall group progression rates. No unexpected adverse
2013). Imaging and chemical biomarkers are having an events were observed. This was the first therapeutic trial
increasing role in the field of ageing; however, they are far to demonstrate an ability to delay the clinical diagnosis of
removed from being used in clinical practice. Future trans- AD using donepezil at least over the 18 months of the trial.
lational research needs to be carried out to reliably and accu- Donepezil has also been investigated as a therapy in other
rately identify subjects in the presymptomatic stages of AD. trials (Doody et al., 2009; Lu et al., 2009; Petrella et al., 2009).
434 Dementia

Clinical trials have not been limited to pharmacological Blackford, R.C. and La Rue, A. (1989). Criteria for
agents and have examined exercise (Lautenschlager et al., 2008) diagnosing age-associated memory impairment.
and leisure activities/cognitive activities such as reading, writ- Developmental Neuropsychology, 5 (4): 295–306.
ing and playing board games (Verghese et al., 2003, 2006, 2009). Blennow, K. and Hampel, H. (2003). CSF markers for
Both forms of intervention appeared helpful (see Chapter 43). incipient Alzheimer’s disease. The Lancet Neurology,
Although there are several ongoing clinical trials, no 2: 605–613.
generally effective pharmacological interventions have Bobinski, M., De Leon, M.J., Wegiel, J. et al. (2000). The
been demonstrated in MCI. Nonetheless, as MCI is a rap- histological validation of post mortem magnetic reso-
idly evolving area of investigation, more effective treatment nance imaging-determined hippocampal volume in
options are likely to be forthcoming. Alzheimer’s disease. Neuroscience, 95: 721–725.
Braak, H. and Braak, E. (1991). Neuropathological
stageing of Alzheimer-related changes. Acta
Neuropathologica, 82: 239–259.
42.7 SUMMARY Brodaty, H., Heffernan, M., Draper, B. et al. (2012).
Neuropsychiatric symptoms in older people with and
The field of ageing and dementia has increasingly empha- without cognitive impairment. Journal of Alzheimer’s
sized the early detection and treatment of cognitive impair- Disease, 31: 411–420.
ment. The construct of MCI, defined as the intermediate Chertkow, H. (2002). Mild cognitive impairment. Current
stage between normal cognitive ageing and dementia, meets Opinion in Neurology, 15: 401–407.
this need. Currently various clinical trials are targeting MCI Chetelat, G., Desgranges, B., De La Sayette, V. et al.
or equivalent conditions. Furthermore, the development of (2003). Mild cognitive impairment: Can FDG-PET pre-
biomarkers has accelerated research work addressing both dict who is to rapidly convert to Alzheimer’s disease?
the presymptomatic and prodromal phases of dementia. Neurology, 60: 1374–1377.
Chien, D.T., Bahri, S., Szardenings, A.K. et al. (2013). Early
clinical PET imaging results with the novel PHF-tau
REFERENCES
radioligand [F-18]-T807. Journal of Alzheimer’s Disease,
Albert, M. S., DeKosky, S.T., Dickson, D. et al. (2011). 34 (2): 457–468.
The diagnosis of mild cognitive impairment due to Chong, M.S. and Sahadevan, S. (2005). Preclinical
Alzheimer’s disease: Recommendations from the Alzheimer’s disease: Diagnosis and prediction of pro-
National Institute on Aging-Alzheimer’s Association gression. TheLancet Neurology, 4 (9): 576–579.
workgroups on diagnostic guidelines for Alzheimer’s Clark, C.M., Schneider, J.A., Bedell, B.J. et al. (2011). Use
disease. Alzheimer’s & Dementia: Journal of the of florbetapir-PET for imaging beta-amyloid pathol-
Alzheimer’s Association, 7: 270–279. ogy. JAMA: The Journal of the American Medical
American Psychiatric Association and American Psychiatric Association, 305: 275–283.
Association DSM-5 Task Force (2013). Diagnostic Clark, C.M., Xie, S., Chittams, J. et al. (2003).
and Statistical Manual of Mental Disorders: DSM-5. Cerebrospinal fluid tau and beta-amyloid: How well do
Washington, DC: American Psychiatric Association. these biomarkers reflect autopsy-confirmed dementia
Attwell, D. and Laughlin, S.B. (2001). An energy budget diagnoses? Archives of Neurology, 60: 1696–1702.
for signaling in the grey matter of the brain. Journal of Crook, T., Bartus, R., Ferris, S. et al. (1986). Age-associated
Cerebral Blood Flow and Metabolism, 21: 1133–1145. memory impairment: Proposed diagnostic criteria and
Bakkour, A., Morris, J.C. and Dickerson, B.C. (2009). The measures of clinical change – Report of a National
cortical signature of prodromal AD: Regional thinning Institute of Mental Health Work Group. Developmental
predicts mild AD dementia. Neurology, 72: 1048–1055. Neuropsychology, 2 (4): 261–276.
Beaulieu-Bonneau, S. and Hudon, C. (2009). Sleep distur- Daly, E., Zaitchik, D., Copeland, M. et al. (2000). Predicting
bances in older adults with mild cognitive impairment. conversion to Alzheimer disease using standardized clini-
International Psychogeriatrics, 21: 654–666. cal information. Archives of Neurology, 57: 675–680.
Bennett, D.A., Wilson, R.S., Schneider, J.A. et al. (2002). DeCarli, C. (2003). Mild cognitive impairment: Prevalence,
Natural history of mild cognitive impairment in older prognosis, aetiology, and treatment. The Lancet
persons. Neurology, 59: 198–205. Neurology, 2: 15–21.
Benton, A., Hamsher, K., De, S. et al. (1983). Contributions Devanand, D.P., Folz, M., Gorlyn, M. et al. (1997).
to Neuropsychological Assessment. New York, NY: Questionable dementia: Clinical course and predictors
Oxford University Press. of outcome. Journal of American Geriatric Society, 45:
Bhalla, R.K., Butters, M.A., Becker, J.T. et al. (2009). 321–328.
Patterns of mild cognitive impairment after treat- Doody, R.S., Ferris, S.H., Salloway, S. et al. (2009). Donepezil
ment of depression in the elderly. American Journal of treatment of patients with MCI: A 48-week randomized,
Geriatric Psychiatry, 17: 308–316. placebo-controlled trial. Neurology, 72: 1555–1561.
Clinical characteristics of mild cognitive impairment 435

Doraiswamy, P.M., Sperling, R.A., Coleman, R.E. et al. Graham, J.E., Rockwood, K., Beattie, B.L. et al. (1997).
(2012). Amyloid-beta assessed by florbetapir F 18 PET Prevalence and severity of cognitive impairment with
and 18-month cognitive decline: A multicenter study. and without dementia in an elderly population. Lancet,
Neurology, 79: 1636–1644. 349: 1793–1796.
Edwards, E.R., Spira, A.P., Barnes, D.E. and Yaffe, K. Grimmer, T., Riemenschneider, M., Forstl, H. et al. (2009).
(2009). Neuropsychiatric symptoms in mild cognitive Beta amyloid in Alzheimer’s disease: Increased deposi-
impairment: Differences by subtype and progres- tion in brain is reflected in reduced concentration in
sion to dementia. International Journal of Geriatric cerebrospinal fluid. Biological Psychiatry, 65: 927–934.
Psychiatry, 24: 716–722. Hanninen, T., Hallikainen, M., Tuomainen, S. et al. (2002).
Fagan, A.M., Mintun, M.A., Mach, R.H. et al. (2006). Prevalence of mild cognitive impairment: A popula-
Inverse relation between in vivo amyloid imaging load tion-based study in elderly subjects. Acta Neurologica
and cerebrospinal fluid Abeta42 in humans. Annals of Scandinavica, 106: 148–154.
Neurology, 59: 512–519. Hansson, O., Zetterberg, H., Buchhave, P. et al. (2006).
Feldman, H., Scheltens, P., Scarpini, E. et al. (2004). Association between CSF biomarkers and incipient
Behavioral symptoms in mild cognitive impairment. Alzheimer’s disease in patients with mild cognitive
Neurology, 62: 1199–1201. impairment: A follow-up study. The Lancet Neurology,
Fischer, P., Jungwirth, S., Zehetmayer, S. et al. (2007). 5: 228–234.
Conversion from subtypes of mild cognitive impair- Hardy, J. and Selkoe, D.J. (2002). The amyloid hypothesis
ment to Alzheimer dementia. Neurology, 68: 288–291. of Alzheimer’s disease: Progress and problems on the
Flicker, C., Ferris, S.H. and Reisberg, B. (1991). Mild cogni- road to therapeutics. Science, 297: 353–356.
tive impairment in the elderly: Predictors of dementia. Herukka, S.K., Hallikainen, M., Soininen, H. and Pirttila, T.
Neurology, 41: 1006–1009. (2005). CSF Abeta42 and tau or phosphorylated tau
Folstein, M.F., Folstein, S.E. and McHugh, P.R. (1975). and prediction of progressive mild cognitive impair-
‘Mini-mental state’. A practical method for grading the ment. Neurology, 64: 1294–1297.
cognitive state of patients for the clinician. Journal of Hesse, C., Rosengren, L., Andreasen, N. et al. (2001).
Psychiatric Research, 12: 189–198. Transient increase in total tau but not phospho-
Fried, L.P., Borhani, N.O., Enright, P. et al. (1991). The tau in human cerebrospinal fluid after acute stroke.
Cardiovascular Health Study: Design and rationale. Neuroscience Letters, 297: 187–190.
Annals of Epidemiology, 1: 263–276. Hoffman, J.M., Welsh-Bohmer, K.A., Hanson, M. et al.
Ganguli, M., Dodge, H.H., Shen, C. and DeKosky, S.T. (2000). FDG PET imaging in patients with pathologi-
(2004). Mild cognitive impairment, amnestic type: An cally verified dementia. Journal of Nuclear Medicine,
epidemiologic study. Neurology, 63: 115–121. 41: 1920–1928.
Geda, Y.E., Knopman, D.S., Mrazek, D.A. et al. (2006). Hwang, T.J., Masterman, D.L., Ortiz, F. et al. (2004). Mild
Depression, apolipoprotein E genotype, and the cognitive impairment is associated with characteristic
incidence of mild cognitive impairment: A prospective neuropsychiatric symptoms. Alzheimer Disease and
cohort study. Archives of Neurology, 63: 435–440. Associated Disorders, 18: 17–21.
Geda, Y.E., Roberts, R.O., Knopman, D.S. et al. (2008). Jack, C.R., Albert, M., Knopman, D. et al. (2011).
Prevalence of neuropsychiatric symptoms in mild Introduction to the recommendations from the
cognitive impairment and normal cognitive aging: National Institute on Aging-Alzheimer’s Association
Population-based study. Archives of General workgroups on diagnostic guidelines for Alzheimer’s
Psychiatry, 65: 1193–1198. disease. Alzheimer’s & Dementia:Journal of the
Geda, Y.E., Roberts, R.O., Mielke, M.M. et al. (2014). Alzheimer’s Association, 7 (3): 257–262.
Baseline neuropsychiatric symptoms and the risk of Jack, C.R., Jr., Knopman, D.S., Weigand, S.D. et al. (2012).
incident mild cognitive impairment: A population-based An operational approach to National Institute on
study. American Journal of Psychiatry, 171: 572–581. Aging-Alzheimer’s Association criteria for preclinical
Geda, Y.E., Schneider, L.S., Gitlin, L.N. et al. (2013). Alzheimer disease. Annals of Neurology, 71: 765–775.
Neuropsychiatric symptoms in Alzheimer’s dis- Jack, C.R., Jr., Lowe, V.J., Weigand, S.D. et al. (2009).
ease: Past progress and anticipation of the future. Serial PIB and MRI in normal, mild cognitive impair-
Alzheimer’s & Dementia:Journal of the Alzheimer’s ment and Alzheimer’s disease: Implications for
Association, 9: 602–608. sequence of pathological events in Alzheimer’s dis-
Geda, Y.E., Smith, G.E., Knopman, D.S. et al. (2004). ease. Brain, 132: 1355–1365.
De novo genesis of neuropsychiatric symptoms Jack, C.R., Jr., Petersen, R.C., Xu, Y.C. et al. (1999).
in mild cognitive impairment (MCI). International Prediction of AD with MRI-based hippocampal volume in
Psychogeriatrics, 16: 51–60. mild cognitive impairment. Neurology, 52: 1397–1403.
436 Dementia

Jack, C.R., Jr., Petersen, R.C., Xu, Y. et al. (2000). Rates of Lopez, O.L., Jagust, W.J., Dekosky, S.T. et al. (2003).
hippocampal atrophy correlate with change in clinical Prevalence and classification of mild cognitive impair-
status in aging and AD. Neurology, 55: 484–489. ment in the Cardiovascular Health Study Cognition
Jagust, W.J., Landau, S.M., Shaw, L.M. et al. (2009). Study: Part 1. Archives of Neurology, 60: 1385–1389.
Relationships between biomarkers in aging and Lopresti, B.J., Klunk, W.E., Mathis, C.A. et al. (2005).
dementia. Neurology, 73: 1193–1199. Simplified quantification of Pittsburgh Compound B
Jagust, W., Reed, B., Mungas, D. et al. (2007). What does amyloid imaging PET studies: A comparative analysis.
fluorodeoxyglucose PET imaging add to a clinical diag- Journal of Nuclear Medicine, 46: 1959–1972.
nosis of dementia? Neurology, 69: 871–877. Lu, P.H., Edland, S.D., Teng, E. et al. (2009). Donepezil
Jelic, V., Kivipelto, M. and Winblad, B. (2006). Clinical trials in delays progression to AD in MCI subjects with depres-
mild cognitive impairment: Lessons for the future. Journal sive symptoms. Neurology, 72: 2115–2121.
of Neurology, Neurosurgery and Psychiatry, 77: 429–438. Lyketsos, C., Lopez, O., Jones, B. et al. (2002). Prevalence
Kemppainen, N.M., Aalto, S., Wilson, I.A. et al. (2006). of neuropsychiatric symptoms in dementia and mild
Voxel-based analysis of PET amyloid ligand [11C] cognitive impairment: Results from the cardiovascu-
PIB uptake in Alzheimer disease. Neurology, 67: lar health study. JAMA: The Journal of the American
1575–1580. Medical Association, 288: 1475–1483.
Klunk, W.E., Engler, H., Nordberg, A. et al. (2004). Imaging Lyketsos, C.G., Steinberg, M., Tschanz, J.T. et al. (2000).
brain amyloid in Alzheimer’s disease with Pittsburgh Mental and behavioral disturbances in dementia:
Compound-B. Annals of Neurology, 55: 306–319. Findings from the Cache County Study on Memory in
Klunk, W.E., Mathis, C.A., Price, J.C. et al. (2006). Two- Aging. American Journal of Psychiatry, 157: 708–714.
year follow-up of amyloid deposition in patients with Maruyama, M., Shimada, H., Suhara, T. et al. (2013).
Alzheimer’s disease. Brain, 129: 2805–2807. Imaging of tau pathology in a tauopathy mouse model
Knopman, D.S., Jack, C.R., Jr., Wiste, H.J. et al. (2012). and in Alzheimer patients compared to normal con-
Short-term clinical outcomes for stages of NIA-AA pre- trols. Neuron, 79: 1094–1108.
clinical Alzheimer disease. Neurology, 78: 1576–1582. Mattsson, N., Zetterberg, H., Hansson, O. et al. (2009).
Kokmen, E., Naessens, J.M. and Offord, K.P. (1987). A CSF biomarkers and incipient Alzheimer disease in
short test of mental status: Description and prelimi- patients with mild cognitive impairment. JAMA: The
nary results. Mayo Clinical Proceedings, 62: 281–288. Journal of the American Medical Association, 302:
Kumar, R., Dear, K.B., Christensen, H. et al. (2005). 385–393.
Prevalence of mild cognitive impairment in 60- to McKhann, G.M., Knopman, D., Chertkow, H. et al. (2011).
64-year-old community-dwelling individuals: The The diagnosis of dementia due to Alzheimer’s dis-
personality and total health through life 60+ Study. ease: Recommendations from the National Institute
Dementia and Geriatric Cognitive Disorders, 19: on Aging and the Alzheimer’s Assocation Workgroup.
67–74. Alzheimer’s & Dementia:Journal of the Alzheimer’s
Landau, S.M., Harvey, D., Madison, C.M. et al. (2010). Association, 7 (3): 269.
Comparing predictors of conversion and decline in Mesulam, M.M. (1998). From sensation to cognition. Brain,
mild cognitive impairment. Neurology, 75: 230–238. 121 (Pt 6): 1013–1052.
Landau, S.M., Mintun, M.A., Joshi, A.D. et al. (2012). Mitchell, A.J. and Shiri-Feshki, M. (2008). Temporal trends
Amyloid deposition, hypometabolism, and longitudinal in the long term risk of progression of mild cognitive
cognitive decline. Annals of Neurology, 72: 578–586. impairment: A pooled analysis. Journal of Neurology,
Larrieu, S., Letenneur, L., Orgogozo, J.M. et al. (2002). Neurosurgery and Psychiatry, 79: 1386–1391.
Incidence and outcome of mild cognitive impairment Morris, J.C. (1993). The Clinical Dementia Rating (CDR):
in a population-based prospective cohort. Neurology, Current version and scoring rules. Neurology, 43:
59: 1594–1599. 2412–2414.
Lautenschlager, N.T., Cox, K.L., Flicker, L. et al. (2008). Ost, M., Nylen, K., Csajbok, L. et al. (2006). Initial CSF
Effect of physical activity on cognitive function in total tau correlates with 1-year outcome in patients
older adults at risk for Alzheimer disease: A random- with traumatic brain injury. Neurology, 67: 1600–1604.
ized trial. JAMA: The Journal of the American Medical Otto, M., Wiltfang, J., Cepek, L. et al. (2002). Tau protein
Association, 300: 1027–1037. and 14-3-3 protein in the differential diagnosis of
Levy, R. (1994). Aging-associated cognitive decline. Creutzfeldt–Jakob disease. Neurology, 58: 192–197.
Working Party of the International Psychogeriatric Palmer, K., Backman, L., Winblad, B. and Fratiglioni, L.
Association in collaboration with the World Health (2008). Mild cognitive impairment in the general popula-
Organization. International Psychogeriatrics, 6: tion: Occurrence and progression to Alzheimer disease.
63–68. American Journal of Geriatric Psychiatry, 16 (7): 603–611.
Clinical characteristics of mild cognitive impairment 437

Parnetti, L., Lanari, A., Amici, S. et al. (2001). CSF phos- Ritchie, K., Artero, S. and Touchon, J. (2001). Classification
phorylated tau is a possible marker for discriminating criteria for mild cognitive impairment: A population-
Alzheimer’s disease from dementia with Lewy bodies. based validation study. Neurology, 56: 37–42.
Phospho-Tau International Study Group. Neurology Roberts, R.O., Geda, Y.E. and Knopman, D.S. (2012). The
Science, 22: 77–78. incidence of MCI differs by subtype and is higher in
Petersen, R.C. (2007). Mild cognitive impairment: men: The Mayo Clinic Study of Aging. Neurology, 78:
Current research and clinical implications. Seminars in 342–351.
Neurology, 27: 22–31. Roberts, R.O., Geda, Y.E., Knopman, D.S. et al. (2008).
Petersen, R.C. (2009). Early diagnosis of Alzheimer’s disease: The Mayo Clinic Study of Aging: Design and sampling,
Is MCI too late? Current Alzheimer Research, 6: 324–330. participation, baseline measures and sample charac-
Petersen, R.C., Aisen, P., Boeve, B.F. et al. (2013). Mild teristics. Neuroepidemiology, 30: 58–69.
cognitive impairment due to Alzheimer disease in the Roberts, R.O., Knopman, D.S., Mielke, M.M. et al. (2014).
community. Annals of Neurology, 74: 199–208. Higher risk of progression to dementia in mild
Petersen, R.C., Caracciolo, B., Brayne, C. et al. (2014). Mild cognitive impairment cases who revert to normal.
cognitive impairment: A concept in evolution. Journal Neurology, 82: 317–325.
of Internal Medicine, 275: 214–228. Sackett, D.L. (1979). Bias in analytic research. Journal of
Petersen, R.C., Roberts, R.O., Knopman, D.S. et al. (2010). Chronic Disease, 32: 51–63.
Prevalence of mild cognitive impairment is higher in men. Sasaki, M., Kodama, C., Hidaka, S. et al. (2009). Prevalence
The Mayo Clinic Study of Aging. Neurology, 75: 889–897. of four subtypes of mild cognitive impairment and
Petersen, R.C., Smith, G.E., Waring, S.C. et al. (1999a). APOE in a Japanese community. International Journal
Mild cognitive impairment: Clinical characterization of Geriatric Psychiatry, 24: 1119–1126.
and outcome. Archives of Neurology, 56: 303–308. Schwartz, W.J., Smith, C.B., Davidsen, L. et al. (1979).
Petersen, R.C., Smith, G.E., Waring, S.C. et al. (1999b). Metabolic mapping of functional activity in the hypo-
Mild cognitive impairment: Clinical characterization thalamo-neurohypophysial system of the rat. Science,
and outcome. Archives of Neurology, 56: 303–308. 205: 723–725.
[Published erratum appears in Archives of Neurology Shaw, L.M., Korecka, M., Clark, C.M. et al. (2007).
1999 June; 56 (6): 760.] Biomarkers of neurodegeneration for diagnosis
Petersen, R.C., Stevens, J.C., Ganguli, M. et al. (2001). and monitoring therapeutics. Nature Reviews Drug
Practice parameter: Early detection of dementia: Mild Discovery, 6: 295–303.
cognitive impairment (an evidence-based review). Report Sjogren, M., Davidsson, P., Tullberg, M. et al. (2001).
of the Quality Standards Subcommittee of the American Both total and phosphorylated tau are increased
Academy of Neurology. Neurology, 56: 1133–1142. in Alzheimer’s disease. Journal of Neurology,
Petersen, R.C., Thomas, R.G., Grundman, M. et al. (2005). Neurosurgery and Psychiatry, 70: 624–630.
Vitamin E and donepezil for the treatment of mild cog- Smith, G., Ivnik, R.J., Petersen, R.C. et al. (1991). Age-
nitive impairment. New England Journal of Medicne, associated memory impairment diagnoses: Problems
352: 2379–2388. of reliability and concerns for terminology. Psychology
Petrella, J.R., Prince, S.E., Krishnan, S. et al. (2009). Effects and Aging, 6: 551–558.
of donepezil on cortical activation in mild cognitive Sperling, R.A., Aisen, P.S., Beckett, L.A. et al. (2011).
impairment: A pilot double-blind placebo-controlled Toward defining the preclinical stages of Alzheimer’s
trial using functional MR imaging. American Journal of disease: Recommendations from the National Institute
Neuroradiology, 30: 411–416. on Aging-Alzheimer’s Association workgroups
Price, J.L. and Morris, J.C. (1999). Tangles and plaques on diagnostic guidelines for Alzheimer’s disease.
in nondemented aging and ‘preclinical’ Alzheimer’s Alzheimer’s & Dementia:Journal of the Alzheimer’s
disease. Annals of Neurology, 45: 358–368. Association, 7: 280–292.
Reisberg, B., Ferris, S.H., De Leon, M.J. and Crook, T. Stokin, G.B., Popovic, M., Gelpi, E. et al. (2015).
(1982). The Global Deterioration Scale for assessment Neuropathologic features of anti-dipeptidyl-pepti-
of primary degenerative dementia. American Journal dase-like protein-6 antibody encephalitis. Neurology,
of Psychiatry, 139: 1136–1139. 84: 430–432.
Reisberg, B., Ferris, S.H., Kluger, A. et al. (2008). Mild Strozyk, D., Blennow, K., White, L.R. and Launer, L.J.
cognitive impairment (MCI): A historical perspective. (2003). CSF Abeta 42 levels correlate with amyloid-
International Psychogeriatrics, 20: 18–31. neuropathology in a population-based autopsy study.
Riemenschneider, M., Lautenschlager, N., Wagenpfeil, S. Neurology, 60: 652–656.
et al. (2002). Cerebrospinal fluid tau and beta-amyloid Tapiola, T., Overmyer, M., Lehtovirta, M. et al. (1997). The
42 proteins identify Alzheimer disease in subjects with level of cerebrospinal fluid tau correlates with neurofi-
mild cognitive impairment. Archives of Neurology, 59: brillary tangles in Alzheimer’s disease. Neuroreport, 8:
1729–1734. 3961–3963.
438 Dementia

Tierney, M.C., Szalai, J.P., Snow, W.G. et al. (1996). Verghese, J., Lipton, R.B., Katz, M.J. et al. (2003). Leisure
A ­prospective study of the clinical utility of activities and the risk of dementia in the elderly. New
ApoE ­genotype in the prediction of outcome in England Journal of Medicine, 348: 2508–2516.
patients with memory impairment. Neurology, 46: Villemagne, V.L., Pike, K.E., Chetelat, G. et al. (2011).
149–154. Longitudinal assessment of Abeta and cognition in
Tolboom, N., van der Flier, W.M., Yaqub, M. et al. (2009). aging and Alzheimer disease. Annals of Neurology, 69:
Relationship of cerebrospinal fluid markers to 11C-PiB 181–192.
and 18F-FDDNP binding. Journal of Nuclear Medicine, Wilson, R.S., Schneider, J.A., Boyle, P.A. et al. (2007).
50: 1464–1470. Chronic distress and incidence of mild cognitive
Van Rossum, I.A., Vos, S., Handels, R. and Visser, P.J. impairment. Neurology, 68: 2085–2092.
(2010). Biomarkers as predictors for conversion from Winblad, B., Palmer, K., Kivipelto, M. et al. (2004). Mild
mild cognitive impairment to Alzheimer-type demen- cognitive impairment – Beyond controversies, towards
tia: Implications for trial design. Journal of Alzheimer’s a consensus: Report of the International Working
Disease, 20: 881–891. Group on Mild Cognitive Impairment. Journal of
Verghese, J., Cuiling, W., Katz, M.J. et al. (2009). Leisure Internal Medicine, 256: 240–246.
activities and risk of vascular cognitive impairment in Wolk, D.A., Price, J.C., Saxton, J.A. et al. (2009). Amyloid
older adults. Journal of Geriatric Psychiatry Neurology, imaging in mild cognitive impairment subtypes. Annals
22: 110–118. of Neurology, 65: 557–568.
Verghese, J., Levalley, A., Derby, C. et al. (2006). Wong, D.F., Rosenberg, P.B., Zhou, Y. et al. (2010). In vivo
Leisure activities and the risk of amnestic mild imaging of amyloid deposition in Alzheimer disease
­cognitive impairment in the elderly. Neurology, 66: using the radioligand 18F-AV-45 (florbetapir [corrected]
821–827. F 18). Journal of Nuclear Medicine, 51: 913–920.
43
Managing the patient with mild cognitive
impairment

NICOLA T. LAUTENSCHLAGER AND ALEXANDER F. KURZ

Usually the diagnostic ‘label’ of MCI is given by a special-


43.1 INTRODUCTION ist such as a neurologist, geriatrician or psychiatrist (e.g. in
a memory clinic) to whom the family doctor might have
There is an abundance of publications focusing on cognitive referred the patient. While initially the term MCI was very
changes during old age, including mild cognitive impair- much restricted to research studies, it is now used more
ment (MCI). The majority of these articles focus on diagnostic frequently in clinical settings (Petersen, 2007), but the cri-
issues and prognostic prediction for future cognitive decline. teria used for diagnosing MCI can vary widely. However,
More recently, the topic of prevention of dementia or dementia the consensus criteria of the International Working Group
risk reduction has also had an impact on research and clinical on MCI in 2004 are often used now (Winblad et al., 2004).
management of MCI. In addition to pharmacological trials, More recent diagnostic concepts such as ‘mild neurocog-
non-pharmacological interventions investigating the role of nitive disorder’ in the Diagnostic and Statistical Manual
modifiable and potentially protective factors are being trialled of Mental Disorders, 5th Edition (DSM-V) (2013) or the
among populations with MCI. However, publications discuss- National Institute on Aging-Alzheimer’s Association
ing how to best manage the patient with MCI outside research (NIA-AA) criteria of ‘MCI due to AD’ (Albert et al., 2011)
studies are less common. With increasing awareness and or ‘prodromal AD’ (Dubois et al., 2010) will no doubt be
knowledge among the population in relation to Alzheimer’s explored in the future for their potential usefulness in the
disease (AD), specialists and memory clinics receive more clinical context. What the newer criteria have in common is
referrals of younger patients, who, often still in the workforce, that current knowledge is used to not only define the criteria
worry about their cognitive function. Managing strategies for for MCI or mild neurocognitive disorder but also to make a
adults with subjective memory complaints (SMC) and MCI statement in relation to the most likely underlying pathol-
need to be individually tailored as they target individuals ogy as to why the patient is experiencing MCI. As such,
from midlife to old age. This chapter focuses on the current these criteria, if validated for the clinical environment, will
practical issues of managing a patient with MCI. help to make the heterogeneity of the term MCI more trans-
parent and therefore give better guidance for management
and prognosis. Receiving a diagnosis of MCI means auto-
43.2 DISCLOSURE AND THE IMPACT matically being at a statistically increased risk of developing
a dementia syndrome later on, however, due to the aetio-
OF DIAGNOSIS
logical heterogeneity of the condition, different outcomes
are possible. Broadly speaking, patients might progress to
The process from first raising concerns in a clinical set- a dementia syndrome with passage of time or rather pres-
ting regarding the cognitive performance of a person later ent quite stable cognitive impairment or even revert back to
diagnosed with MCI to giving information on the diagnos- normal cognition (Gauthier et al., 2006). However, in popu-
tic process and outcome, management options and regular lations visiting the memory clinic, most patients with MCI
follow-up appointments is a journey that can last for several will deteriorate eventually due to referral related enrichment
years and often ultimately results in a dementia diagnosis. with neurodegenerative or cerebrovascular pathologies. In

439
440 Dementia

clinical settings, such as memory clinics, various patholo- also with the next of kin who might have already moved
gies have been reported in autopsy studies of progressive into a carer role without necessarily having processed this
MCI (Bennett et al., 2005; see Chapters 41 and 42 for more gradual change consciously. It has been shown that family
details). members of patients with MCI already experience feelings
Obviously, the individual clinical presentation, medi- of loss and carer burden typically observed in carers of peo-
cal history and available evidence on the underlying cause ple with AD (Artero et al., 2001; Garand et al., 2005; Lu and
will determine how the diagnosis should be disclosed to Haase, 2009) and twice the general population risk depres-
the patient and family and what prognostic advice is to be sion (Guerriero et al., 2004). However, it is important to use
given. MCI as a diagnosis often has a different meaning for the term ‘carer’ carefully when communicating with MCI
a patient from a family with familial AD as compared to patients and their families. Since the essence of the diag-
someone who has no family history of cognitive impair- nostic category of MCI is that the level of independence of
ment. Many specialists inform the patient and next of kin the patient does not justify a diagnosis of dementia or major
about the many unknown elements surrounding the MCI neurocognitive disorder, the term ‘carer’ could be confusing
‘label’ and the fact that research in this area is still a work in this context. A more neutral term like ‘family member’ or
in progress (Petersen, 2007) and this openness is usually ‘friend’ is preferable (Austrom and Lu, 2009). This does not
appreciated. Where available and appropriate, the question contradict recent findings showing that patients with MCI
of prognosis might make it relevant to educate patients and do experience some impairment in very complex activities
family members about the potential usefulness of biomark- of daily living (ADLs) (Perneczky et al., 2006) with family
ers, which can help with estimating the risk for clinical dete- members needing to provide ‘caregiving’ for more complex
rioration. Structural magnetic resonance imaging (MRI) is ADLs (McIlvane et al., 2008).
recommended when considering a clinical diagnosis of MCI Ethical concerns have been raised about disclosing the
and can assist with estimating the underlying pathologies ‘label’ MCI to patients and family members. These concerns
contributing to the clinical presentation as well as the risk include opening the door to distress, helplessness and hope-
for clinical deterioration in the near future. However, more lessness, with potential catastrophic outcomes like suicide
standardization and international guidelines are needed for (Werner and Korczyn, 2008) in light of the unavailability of
the clinical context (Petersen et al., 2001; Jack et al., 2009; effective treatment and looming AD. There is controversy
Jack et al., 2010; Jack et al., 2011; Petersen, 2011). Functional about whether patients with MCI already experience lim-
imaging, such as fludeoxyglucose or more recently, ited awareness of their symptoms, but, overall lack of insight
carbon-11-labelled Pittsburgh compound B or florbetapir is considered less common than in AD patients (Vogel et al.,
positron emission tomography (PET) can give an indica- 2004; Kalbe et al., 2005). However, in the majority of cases,
tion as to what extent AD pathology is present and might the patient and family member already know that some-
impact on prognosis (Small et al., 2008; Johnson et al., 2013; thing is wrong and therefore the diagnosis does not surprise
Villemagne et al., 2013). Another biomarker of relevance is them with ‘bad news’ but confirms symptoms observed,
low cerebrospinal fluid (CSF) Aβ42, which has been shown often for many months. In this context, the term ‘mild’ in
to have some predictive potential for future progression MCI might be seen as trivializing symptoms, which causes
towards dementia; however, a routine lumbar puncture as great concern. It is helpful to prevent this sentiment by
part of diagnostic and management procedures for MCI is explaining that ‘mild’ is used in comparison to severe cog-
currently not recommended for many countries (Mattsson nitive impairment such as dementia or major neurocog-
et al., 2009; Petersen, 2009; see Chapter 42 for more infor- nitive disorder, but not in relation to the individual, often
mation on biomarkers and MCI). upsetting experience. Some clinicians, therefore, prefer to
Educating the patient and family in detail about the rel- describe the specific clinical findings without using the tem
evance of clinical symptoms, assessment process and results MCI. On the other hand, similar to the disclosure process
of tests, for example, showing and explaining the relevant of a dementia diagnosis, it should not be overlooked that
imaging scans, is usually helpful since it validates the con- patients also have the right ‘not to know’ and, therefore, it is
cerns of the patient and family and frames the symptoms important to adjust the level of detail of information given
of MCI in a medical context. This should include the expla- accordingly.
nation that a thorough search for ‘reversible’ causes of the
symptoms was part of the investigations and did not reveal
any relevant potential causes. All this should help fam- 43.3 PHARMACOLOGICAL AND
ily members to accept the medical diagnosis and therefore
NON-PHARMACOLOGICAL
alleviate anger, denial and guilt (Austrom and Lu, 2009).
This is especially crucial in patients where the observed
APPROACHES
impairment fluctuates and the next of kin wonders whether
the patient is ‘putting the symptoms on’. In patients where No pharmacological approaches have yet shown significant
the clinical findings strongly suggest a pre-dementia syn- improvement of symptoms or delay of progression in MCI
drome, the possible emergence of psychological stress and and consequently there are currently no approved medi-
depression should not only be raised with the patient but cations specifically for the treatment of MCI (Russ and
Managing the patient with mild cognitive impairment 441

Morling, 2012). Randomized controlled trials (RCTs) with Yaffe, 2014; Astey et al., 2015) but also in individuals
negative results for the primary outcome have been con- with SMC and MCI (Lautenschlager et al., 2014; see also
ducted with cholinesterase inhibitors, vitamin E, piracetam, Chapter 55).
rofecoxib, ginkgo biloba and testosterone supplementa- Evidence is still limited, and findings are not consis-
tion (Salloway et al., 2004; Petersen et al., 2005; Thal et al., tent, but increasing evidence suggests that especially cog-
2005; Jelic et al., 2006; Feldman et al., 2007; DeKosky et al., nitive and physical activity are activities, which should
2008; Emmelot-Vonk et al., 2008; Winblad et al., 2008; Snitz also be encouraged in individuals with SMC and MCI
et al., 2009). However, there is some debate on whether the (Lautenschlager et al., 2014). Several theories have been
RCT with donepezil and vitamin E showed some promis- put forward to explain how cognitive activity could ben-
ing results by demonstrating a reduced risk for the first 12 efit cognition in old age and, therefore, potentially help
months in the donepezil group, which grew even after 24 patients with MCI. These theories include ‘use it or lose
months for apolipoprotein E (ApoE) ε4–positive patients. it’, ‘environmental enrichment’, ‘the cognitive reserve
Off-label use of cholinesterase inhibitors continues to be hypothesis’ and the ‘scaffolding theory of cognitive aging’
discussed in the literature (Diniz et al., 2009) and often in approach (Fratiglioni et al., 2004; Salthouse, 2006; Stern,
combination with memantine, seems to be used by many 2006; Park and Reuter-Lorenz, 2009). Often cognitive
clinicians. A survey in the United States showed that one activity is grouped into further subtypes, such as cogni-
quarter of patients with MCI received anti-AD medications tive stimulation, cognitive training and cognitive rehabili-
(Weinstein et al., 2009). A more recent 48-week RCT with tation, and significant benefits for participants with MCI
donepezil in MCI showed small but significant changes have been reported with all three subtypes, however, only
on the Alzheimer’s Disease Assessment Scale-Cognitive few investigated whether these benefits translated into
Subscale (ADAS-Cog), but no significant changes in the clinical and functional improvement (Kurz et al., 2011).
primary outcome ­measure of global function (Doody et al., A recent Cochrane review highlighted non-consistent find-
2009) was observed. RCTs with galantamine were overall ings dependent on whether the control group was passive
negative, but again showed interesting results, with a non- or active which called for high-quality trials (Martin et al.,
significant reduction of progression rates and a reduced rate 2011), but especially more recent studies have reported
of progression of brain atrophy in the galantamine group. positive findings (Hampstead et al., 2014).
However, there was also an increased mortality rate reported As with cognitive activity, there are promising find-
in ­galantamine-treated participants (Gold et al., 2004) and ings for the outcome cognition such as exposing non-
a recent Cochrane review indicated that patients with MCI demented older adults to regular ideally moderate physical
might be at greater risk of experiencing an increase of activity of at least 150 minutes per week (Sofi et al., 2010;
adverse effects when treated with cholinesterase inhibitors Lautenschlager et al., 2012; Norton et al., 2014). As with
(Russ et al., 2012). A small trial with memantine in patients cognitive activity, RCTs in SMC and MCI are still sparse
with age-associated memory impairment (AAMI) was with often inconsistent findings (Lautenschlager et al.,
negative in relation to measures of memory performance 2013), but a recent RCT with 170 subjective memory
(Ferris et al., 2007). Pharmacological trial experiences complainers and MCI patients aged 50 and above found
to date highlight the ongoing challenges in choosing the significant differences between the intervention and con-
most appropriate methodology and d ­ iagnostic criteria for trol groups (Lautenschlager, 2008; Cox et al., 2013). The
MCI (Knopman, 2009) and targeted trials with individuals 24-week intervention was individually tailored and home-
who qualify for the ­narrow diagnosis of ­prodromal AD via based, focusing on walking, but allowing combinations
routine use of biomarkers might improve the trial results with other forms of physical activity depending on pref-
record in the future (see also Chapter 42). erence and medical history. Participants in the interven-
There is increasing evidence that good general medical tion group were asked to exercise for at least 150 minutes
management is particularly important for patients with per week. To enhance adherence, a modified behavioural
MCI. This includes adhering to current recommendation to intervention based on social cognitive theory (Cox et al.,
prevent and manage hypertension, heart disease, diabetes 2003) was implemented via workshop, manual, newslet-
and stroke, especially, in cases where vascular risk factors ters and regular phone calls. Participants were reassessed
or cerebrovascular changes are present (Langa and Levine, after 24 weeks, 12 and 18 months and the primary out-
2014). come measure, the ADAS-Cog showed significant differ-
In the absence of effective pharmacological treatment, ences between the groups at all three time points. While
considering non-pharmacological approaches for an indi- this encouraging finding needs to be confirmed in larger
vidualized management plan becomes crucial. Reducing RCTs, it demonstrates that patients with MCI can be moti-
modifiable risk factors for dementia and AD and enhanc- vated to participate in a physical activity programme and
ing potential protective factors are of particular interest cognitively benefit from it. Currently, several international
here as evidence is mounting that this approach could groups are trying to combine potentially successful inter-
delay the onset of dementia and AD not only in cognitively ventions in very large multicentre trials with individuals
healthy middle aged and older populations as a dementia at risk of dementia, aiming to maximize the protective
risk reduction strategy (Norton et al., 2014; Smith and effect. To date a number of trials reported benefits in
442 Dementia

neuropsychological outcomes, including attention, execu-


tive functions and memory. Evidence is increasing that BOX 43.1: Important topics to consider in
next to aerobic exercise additional resistance training as the management of MCI
well as physical activity with cognitive components such as
tai chi, can be of benefit (Lam et al., 2011; Nagamatsu et al., Disclosure of diagnosis
2012). More research needs to be done to elicit specific Information on assessment results
information on the optimal combination of types of physi- Individualized information on prognosis
cal activity, their duration and intensity, as well as how to Pharmacological management options
best motivate individuals with SMC and MCI to modify Non-phamarmacological approaches
their behaviour. Despite these limitations in knowledge, Information on research trials in the area
recent reviews on MCI promote the importance to encour- Monitoring health and vascular risk factors
age patients with MCI to consider regular cognitive and Neuropsychiatric symptoms
physical activity (Knopman and Petersen, 2014). Discuss ADLs (including driving)
Other potentially protective factors include social activi- Planning the future (e.g. finances, travelling, etc.)
ties (Karp et al., 2006) and a healthy diet with a focus on Information on AD treatment in case of progression
specifically a ‘Mediterranean diet’ (Bhat, 2009). This seems ‘Carer’ burden
logical since a healthy diet positively affects the vascular risk Information on useful contacts (e.g. Alzheimer
factors mentioned above. Nutrients such as fruit, vegetables, association)
fish, nuts, cereals and olive oil have been identified as poten- Regular follow-up appointments
tially lowering the risk of dementia (Scarmeas et al., 2006).
The effect of these nutrients is largely attributed to antioxi-
dants and polyunsaturated fatty acids; however, more trials and using standard instruments like the Neuropsychiatric
are needed (Bhat, 2009; Fotuhi et al., 2009). Inventory can be useful (also see Chapter 42).
Only recently evidence is emerging that offering a multi- After considering pharmacological and non-pharmacological
domain approach where several of these protective factors strategies, a regular follow-up visit with using standardized
are combined can lead in older adults at risk of cognitive instruments is the best approach for ­identifying cogni-
decline and dementia to cognitive benefits (Ngandu et al., tive decline as early as possible. This approach can help to
2015). Long-term follow-up of these studies will help to reassure patients and family members, that, while there is
determine whether these approaches can also impact limited effective treatment for MCI, significant progression
dementia incidence. of symptoms will not be overlooked and therefore, anti-
dementia treatment can be started as soon as it has been
proved helpful. Furthermore, management of potential
impairment of complex ADLs should be discussed, cover-
43.4 PRACTICAL CONSIDERATIONS ing important areas such as work, finances, caring for sig-
AND CONCLUSIONS nificant others, driving, etc. Recent findings demonstrate
that patients with MCI perform worse on driving tests
The above findings and clinical experience suggests that than cognitively healthy older adults (Frittelli et al., 2009;
after disclosure of the diagnosis of MCI to the patient and Wadley et al., 2009) and deteriorated in their financial skills
family, individually suitable management strategies should when progressing towards dementia (Triebel et al., 2009).
be considered. Box 43.1 gives some guidance on relevant Also, medical decision-making capacity can deteriorate
topics, which could be raised during a discussion about with progression of symptoms (Okonkwo et al., 2008) of
management options. dementia. Additional individual counselling for patient
An increasing body of evidence emphasizes neuro- and family might be helpful (Ellison, 2008) if high levels
psychiatric symptoms (Edwards et al., 2009; Simard of distress are detected and especially, if next of kin start
et al., 2009) as well as sleep disturbance in MCI (Beaulieu- experiencing carer burden.
Bonneau and Hudon, 2009). Between 35% and 75% of To summarize, the best possible management of patients
patients with MCI experience neuropsychiatric symptoms with MCI is of increasing importance in light of the rising
with the most common being depression, apathy, anxiety uptake of the diagnosis in clinical settings. The manage-
and irritability. These patients have been reported to have a ment approach needs to be individualized depending on the
higher risk of progression to AD than those without these medical history, results of assessments and investigations
symptoms (Apostolova and Cummings, 2008; Edwards as well as the personal situation of the patient and family
et al., 2009). Patients with MCI should be screened for involved. If available, a multidisciplinary team approach
symptoms of depression and treated if pharmacological might be best to address the complex needs of the patient
treatment is indicated. Due to cognitive impairment, the and family (Ellison, 2008). The approach should not be
antidepressant should be chosen carefully, avoiding anti- nihilistic. A proactive management plan with regular fol-
cholinergic medication. Clinicians should screen for and low-up visits is crucial to help patients and family members
treat neuropsychiatric symptoms in patients with MCI cope in this difficult period of uncertainty.
Managing the patient with mild cognitive impairment 443

REFERENCES delaying progression to Alzheimer’s disease. European


Archives of Psychiatry and Clinical Neuroscience, 259
Albert, M.S., DeKosky, S.T., Dickson, D. et al. (2011). (4): 248–256.
The diagnosis of mild cognitive impairment due to Doody, R.S., Ferris, S.H., Salloway, S. et al. (2009).
Alzheimer’s disease: Recommendations from the Donepezil treatment of patients with MCI: A 48-week
National Institute on Aging-Alzheimer’s Association randomized, placebo-controlled trial. Neurology, 72:
workgroups on diagnostic guidelines for Alzheimer’s 1555–1561.
disease. Alzheimer’s & Dementia: The Journal of the Dubois, B., Feldman, H.H., Jacova, C. et al. (2010).
Alzheimer’s Association, 7 (3): 270–279. Revising the definition of Alzheimer’s disease. Lancet
Apostolova, L.G. and Cummings, J.L. (2008). Neurology, 9: 1118–1127.
Neuropsychiatric manifestations in mild cognitive Edwards, E.R., Spira, A.P., Barnes, D.E. and
impairment: A systematic review of the literature. Yaffe, E. (2009). Neuropsychiatric symptoms in mild
Dementia and Geriatric Cognitive Disorders, 25: cognitive impairment: Differences by subtype and pro-
115–126. gression to dementia. International Journal of Geriatric
Artero, S., Touchon, J. and Ritchie, K. (2001). Disability Psychiatry, 24: 716–722.
and mild cognitive impairment: A longitudinal popu- Ellison, J.M. (2008). A 60-year-old woman with mild mem-
lation-based study. International Journal of Geriatric ory impairment: Review of mild cognitive impairment.
Psychiatry, 16: 1092–1097. The Journal of the American Medical Association, 300:
Astey, K.J., Eramudugolla, D.E., Lautenschlager, N.T. and 1566–1574.
Dixon, R.A. (2015). Bridging the translation gap: From Emmelot-Vonk, M.H., Verhaar, H.J., Nakhai Pour H.R. et al.
dementia risk assessment to advice on risk reduction. (2008). Effect of testosterone supplementation on
Journal of Prevention of Alzheimer’s Disease, 2 (3): functional mobility, cognition, and other parameters
189–198. in older men. The Journal of the American Medical
Austrom, M.G. and Lu, Y. (2009). Long term caregiving: Association, 299: 39–52.
Helping families of persons with mild cognitive impair- Feldman, H.H., Ferris, S., Winblad, B. et al. (2007). Effect
ment cope. Current Alzheimer Research, 6: 392–398. of rivastigmine on delay to diagnosis of Alzheimer’s
Beaulieu-Bonneau, S. and Hudon, C. (2009). Sleep distur- disease from mild cognitive impairment: The InDDEx
bances in older adults with mild cognitive impairment. study. Lancet Neurology, 6 (6): 501–512.
International Psychogeriatrics, 21: 654–666. Ferris, S., Schneider, L., Farmer, M. et al. (2007). A dou-
Bennett D.A., Schneider J.A., Bienias J.L. et al. (2005). ble-blind, placebo-controlled trial of memantine in
Mild cognitive impairment is related to Alzheimer age-associated memory impairment (memantine in
disease pathology and cerebral infractions. Neurology, AAMI). International Journal of Geriatric Psychiatry, 22:
64: 843–841. 448–455.
Bhat, R.S. (2009). You are what you eat: Of fish, fat and Fotuhi, M., Mohassel, P. and Yaffe, K. (2009). Fish con-
folate in late-life psychiatric disorders. Current Opinion sumption, long-chain omega-3 fatty acids and risk of
in Psychiatry, 22: 541–545. cognitive decline or Alzheimer disease: A complex
Cox, K.L., Burke, V., Gorely, T.J. et al. (2003). Controlled association. Nature Clinical Practice Neurology, 5:
comparison of retention and adherence in home- vs 140–152.
center-initiated exercise interventions in women ages Fratiglioni, L., Paillard-Borg, S. and Winblad, B. (2004). An
40–65 years: The S.W.E.A.T. Study (Sedentary Women active and socially integrated lifestyle in late life might
Exercise Adherence Trial). Preventive Medicine, 36 (1): protect against dementia. The Lancet Neurology, 3:
17–29. 343–353.
Cox, K.L., Flicker, L., Almeida, O.P. et al. (2013). The Frittelli, C., Borghetti, D., Iudice, G. et al. (2009). Effects
FABS trial: A randomized control trial of the effects of Alzheimer’s disease and mild cognitive impairment
of a 6-month physical activity intervention on adher- on driving ability: A controlled clinical study by simu-
ence and long-term physical activity and self-efficacy lated driving test. International Journal of Geriatric
in older adults with memory complaints. Preventive Psychiatry, 24: 232–238.
Medicine, 57: 824–830. Garand, L., Dew, M.A., Eazo, L.R. et al. (2005). Caregiving
DeKosky, S.T., Williamson, J.D., Fitzpatrick, A.L. et al.; burden and psychiatric morbidity in spouses of per-
Ginkgo Evaluation of Memory (GEM) Study sons with mild cognitive impairment. International
Investigators.(2008). Ginkgo biloba for prevention Journal of Geriatric Psychiatry, 20: 512–522.
of dementia: A randomized controlled trial. The Gauthier, S., Reisberg, B., Zaudig, M. et al.; International
Journal of the American Medical Association, 300 (19): Psychogeriatric Association Expert Conference on
2253–2262. Mild Cognitive Impairment.(2006). Mild cognitive
Diniz, B.S., Pinto, J.A. Jr., Gongaza, M.L.C. et al. (2009). To impairment. Lancet, 367: 1262–1270.
treat or not to treat? A meta-analysis of the use of cho- Gold, M., Francke, S., Nye, J.S. et al. (2004). Impact of
linesterase inhibitors in mild cognitive impairment for APOE genotype on the efficacy of galantamine for the
444 Dementia

treatment of mild cognitive impairment. Neurobiology Lam, L.C., Chau, R.C., Wong, B.M. et al. (2011). Interim
of Ageing, 25: S521. follow-up of a randomized controlled trial compar-
Guerriero, A.M., Damush, T.M., Hartwell, C.W. et al. (2004). ing Chinese style mind body (tai chi) and stretching
The development and implementation of non-phar- exercises on cognitive function in subjects at risk of
macological protocols for the management of patients progressive cognitive decline. International Journal of
with Alzheimer disease and their families in a multi- Geriatric Psychiatry, 26: 733–740.
racial primary care setting. Gerontologist, 44: 548–553. Langa, K.M. and Levine, D.A. (2014). The diagnosis
Hampstead, B.M., Gillis, M.M. and Stringer, A.Y. (2014). and management of mild cognitive impairment. A
Cognitive rehabilitation of memory for mild cogni- clinical review. The Journal of the American Medical
tive impairment: A methodological review and model Association, 312 (23): 2551–2561.
for future research. Journal of the International Lautenschlager, N.T. (2013). Physical activity and cogni-
Neuropsychol Society, 20 (2): 135–151. tion in older adults with mild cognitive impairment and
Jack C.R., Jr., Barkhof, F., Bernstein, M.A. et al. (2011). dementia. Neurodegenerative Disease Management,
Steps to standardization and validation of hippocam- 3: 211–218.
pal volumetry as a biomarker in clinical trials and diag- Lautenschlager, N.T., Anstey, K.J. and Kurz, A.F. (2014).
nostic criterion for Alzheimer’s disease. Alzheimer’s & Non-pharmacological strategies to delay decline.
Dementia: The Journal of the Alzheimer’s Association, Maturitas, 79: 170–173.
7 (4): 474–485. Lautenschlager, N.T., Cox, K. and Cyarto, E.V. (2012). The
Jack, C.R., Jr., Lowe, V.J., Weigand, S.D. et al. (2009). influence of exercise on brain ageing and dementia.
Serial PIB and MRI in normal, mild cognitive impair- Biochimica et Biophysica Acta, 1822: 474–481.
ment and Alzheimer’s disease: Implications for Lautenschlager, N.T., Cox, K.L., Flicker, L. et al. (2008).
sequence of pathological events in Alzheimer’s dis- Effect of physical activity on cognitive function in older
ease. Brain, 132: 1355–1365. adults at risk for Alzheimer disease: A randomized
Jack, C.R., Jr., Wiste H.J., Vemuri P. et al.; Alzheimer’s trial. The Journal of the American Medical Association,
Disease Neuroimaging Intiative. (2010). Brain beta- 300: 1027–1037.
amyloid measures and magnetic resonance imaging Lu, Y.-F.Y. and Haase, J.E. (2009). Experience and per-
atrophy both predict time-to-progression from mild spectives of caregivers of spouse with mild cognitive
cognitive impairment to Alzheimer’s disease. Brain, impairment. Current Alzheimer Research, 6: 384–391.
133 (11): 3336–3348. Martin, M., Clare, L., Altgassen, A.M. et al. (2011).
Jelic, V., Kivipelto, M. and Wilblad, B. (2006). Clinical trials in Cognition-based interventions for healthy older
mild cognitive impairment: Lessons for the future. Journal people and people with mild cognitive impairment.
of Neurology Neurosurgery and Psychiatry, 77: 429–438. Cochrane Database Systematic Reviews, 19 (1): 1–51.
Johnson, K.A., Sperling, R.A., Gidiscin, C.M. et al.; Mattsson, N., Zetterberg, H., Hansson, O. et al. (2009).
AV45-A11 study group. (2013). Florpetapir (F18-AV-45) CSF biomarkers and incipient Alzheimer disease
PET to assess amyloid burden in Alzheimer’s disease in patients with mild cognitive impairment. The
dementia, mild cognitive impairment, and normal Journal of the American Medical Association, 302:
aging. Alzheimer’s & Dementia: The Journal of 385–393.
the Alzheimer’s Association, 9 (Suppl. 5): S72–S83. McIlvane, J.M., Popa, M.A., Robinson, B. et al. (2008).
Kalbe, E., Salmon, E., Perani, D. et al. (2005). Anosognosia Perceptions of illness, coping, and well-being in
in very mild Alzheimer’s disease but not in mild cogni- persons with mild cognitive impairment and their care
tive impairment. Dementia and Geriatric Cognitive partners. Alzheimer Disease and Associated Disorders,
Disorders, 19: 349–356. 22: 284–292.
Karp, A., Paillard-Borg, S., Wang, H.X. et al. (2006). Mental, Nagamatsu, L.S., Handy, T.C., Hsu, C.L. et al. (2012).
physical and social components in leisure activities Resistance training promotes cognitive and functional
equally contribute to decrease dementia risk. Dementia brain plasticity in seniors with probable mild cogni-
and Geriatric Cognitive Disorders, 21: 65–73. tive impairment. Archives of Internal Medicine, 172:
Knopman, D.S. (2009). Cracking the therapeutic nut in 666–668.
mild cognitive impairment: Better nuts and better Ngandu, T., Lehtisalo, J., Solomon, A. et al. (2015). A
nutcrackers. Neurology, 72: 1542–1543. 2 year multidomain intervention of diet, exercise,
Knopman, D.S. and Petersen, R.C. (2014). Mild cognitive cognitive training, and vascular risk monitoring versus
impairment and mild dementia: A clinical perspective. control to prevent cognitive decline in at-risk elderly
Mayo Clinic Proceedings, 89: 1452–1459. people (FINGER): A randomized controlled trial. The
Kurz, A.F., Leucht, S. and Lautenschlager, N.T. (2011). The Lancet, 385 (9984): 2255–2263.
clinical significance of cognition-focused interventions Norton, S., Matthews, F.E., Barnes, D.E. et al. (2014).
for cognitively impaired older adults. A systematic Potential for primary prevention of Alzheimer’s dis-
review of randomized controlled trials. International ease: An analysis of population-based data. Lancet
Psychoagriatrics, 23: 1364–1375. Neurology, 13: 788–794.
Managing the patient with mild cognitive impairment 445

Okonkwo, O.C., Griffith, H.R., Copeland, K. et al. (2008). supported by international experts. Journal of
Medical decision-making capacity in mild cognitive Alzheimer’s Disease, 38: 699–703.
impairment: A 3-year longitudinal study. Neurology, Snitz B.E., O’Meara, E.S., Calrson, M.C. et al. (2009).
71: 1474–1480. Ginkgo biloba for preventing cognitive decline in
Park, D.C. and Reuter-Lorenz, P. (2009). The adaptive older adults. The Journal of the American Medical
brain: Aging and neurocognitive scaffolding. Annual Association, 302: 2663–2670.
Review of Psychology, 60: 173–196. Sofi, F., Valecchi, D., Bacci, D. et al. (2010). Physical activ-
Perneczky, R., Pohl, C., Sorg, C. et al. (2006). Complex ity and risk of cognitive decline: A meta-analysis of
activities of daily living in MCI: Conceptual and diag- prospective studies. Journal of Internal Medicine, 269:
nostic issues. Age and Ageing, 35: 240–245. 107–117.
Petersen, R.C. (2007). Mild cognitive impairment: Stern, Y. (2006). Cognitive reserve and Alzheimer dis-
Current research and clinical implications. Seminars in ease. Alzheimer Disease and Associated Disorders, 20
Neurology, 27 (1): 22–31. (Suppl. 2): S69–S74.
Petersen, R.C. (2009). Early diagnosis of Alzheimer’s Thal, L.J., Ferris, S.H., Kirby, L. et al. (2005). A randomized,
disease: Is MCI too late? Current Alzheimer Research, double-blind, study of Rofecoxib inpatients with mild
6: 324–330. cognitive impairment. Neuropsychopharmacology, 30:
Petersen, R.C. (2011). Clinical practice: Mild cognitive 1204–1215.
impairment. The New England Journal of Medicine, Triebel, K.L., Martin, R., Griffith, H.R. et al. (2009).
364: 2227–2234. Declining financial capacity in mild cognitive impair-
Petersen, R.C., Stevens, J.C., Ganguli, M. et al. (2001). ment: A 1-year longitudinal study. Neurology, 73:
Practice parameter: Early detection of dementia: Mild 928–934.
cognitive impairment (an evidence-based review). Villemagne, V.L., Burnham, S., Bourgeat, P. et al. (2013).
Report of the Quality Standards Subcommittee of Amyloid β deposition, neurodegeneration, and cogni-
the American Academy of Neurology. Neurology, 56: tive decline in sporadic Alzheimer’s disease: A pro-
1133–1142. spective cohort study. Lancet Neurology, 12: 357–367.
Petersen, R.C., Thomas, R.G., Grundman, M. et al. (2005). Vogel, A., Stockholm, J., Gade, A. et al. (2004). Awareness
Vitamin E and donepezil for the treatment of mild of deficits in mild cognitive impairment in Alzheimer
cognitive impairment. The New England Journal of disease: Do persons with MCI have impaired insight?
Medicine, 352: 2379–2388. Dementia and Geriatric Cognitive Disorders, 17:
Russ, T.C. and Morling, J.R. (2012). Cholinesterase inhibi- 181–187.
tors for mild cognitive impairment. Cochrane Database Wadley, V.G., Okonkwo, O., Crowe, M. et al. (2009). Mild
Systematic Reviews, (9): CD009132. cognitive impairment and everyday function: An inves-
Salloway, S., Ferris, S., Kluger, A. et al. (2004). Efficacy of tigation of driving performance. Journal of Geriatric
donepezil in mild cognitive impairment: A randomized Psychiatry Neurology, 22: 87–94.
placebo-controlled trial. Neurology, 63: 651–657. Weinstein, A.M., Barton, C., Ross, L. et al. (2009).
Salthouse, T.A. (2006). Mental exercise and mental aging: Treatment practices of mild cognitive impairment in
Evaluating the validity of the “use it or lose it” hypoth- California Alzheimer’s Disease Centers. Journal of the
esis. Perspectives on Psychological Science, 1: 68–87. American Geriatrics Society, 57 (4): 686–690.
Scarmeas, N., Stern, Y., Tang, M. et al. (2006). Werner, P. and Korczyn, A.D. (2008). Mild cognitive
Mediterranean diet and risk for Alzheimer disease. impairment: Conceptual, assessment, ethical, and
Annals of Neurology, 63: 1709–1717. social issues. Clinical Interventions in Aging, 3:
Simard, M. and Van Reekum, R. (2009). Psychological dis- 413–420.
tress and risk for dementia. Current Psychiatry Reports, Winblad, B. Gauthier, S., Scinto, L. et al. (2008). Safety and
11: 41–47. efficacy of galantamine in subjects with mild cognitive
Small, G.W., Bookheimer, S.Y., Thompson, P.M. et al. impairment. Neurology, 70: 2024–2035.
(2008). Current and future uses of neuroimaging for Winblad, B., Palmer, K., Kivipelto, M. et al. (2004). Mild
cognitively impaired patients. Lancet Neurology, 7: cognitive impairment – Beyond controversies, towards
161–172. a consensus: Report of the International Working
Smith, D.A. and Yaffe, K. (2014). Dementia (including Group on Mild Cognitive Impairment. Journal of
Alzheimer’s disease) can be prevented: Statement Internal Medicine, 256: 240–246.
3
Part    

Alzheimer’s Disease: Clinical aspects

44 Risk factors for Alzheimer’s disease 447


Patricia A. Boyle and Robert S. Wilson
45 The natural history of Alzheimer’s disease 453
Jody Corey-Bloom and Michael S. Rafii
46 The neuropathology of Alzheimer’s disease 470
Colin L. Masters
47 Neurochemistry of Alzheimer’s disease 486
Paul T. Francis
48 The central role of Aβ amyloid and tau in the pathogenesis of Alzheimer’s disease 492
Craig W. Ritchie and Colin L. Masters
49 Inflammation in Alzheimer’s disease 508
Clive Holmes
50 Genetics of Alzheimer’s disease 519
Margie Smith
51 Blood and cerebrospinal fluid biomarkers for Alzheimer’s disease 528
Simone Lista, Henrik Zetterberg, Sid E. O’Bryant, Kaj Blennow and Harald Hampel
52 Established treatments for Alzheimer’s disease 539
Alexander Kurz and Nicola T. Lautenschlager
53 Drug treatments in development for Alzheimer’s disease 554
Paul Yates and Michael Woodward
54 Vitamins and complementary therapies used to treat Alzheimer’s disease 587
Dennis Chang, Genevieve Z. Steiner, Dilip Ghosh and Alan Bensoussan
55 Prevention of Alzheimer’s disease and Alzheimer’s dementia 598
Tom C. Russ, Karen Ritchie and Craig W. Ritchie
56 Trial designs 613
Serge Gauthier
44
Risk factors for Alzheimer’s disease

PATRICIA A. BOYLE AND ROBERT S. WILSON

Additionally, in this chapter, we discuss the possible


44.1 INTRODUCTION neurobiological mechanisms by which various risk fac-
tors influence the development of AD, with the caveat that
Alzheimer’s disease (AD) is one of the top 10 leading not all risk factors have been well-studied in this regard.
causes of death for which there is no cure or disease Moreover, the relationships among risk factors, common
modifying treatment, making it one of the most feared age-related neuropathologies, such as the pathology of
conditions of ageing (Alzheimer’s Association, 2015). By AD, and cognitive impairment are complex. We focus on
the time a clinical diagnosis of AD is made, the pathologi- three primary ways in which risk factors can be associ-
cal hallmarks of the disease are widespread in the brain, ated with AD: (1) they can initiate pathological processes
making it very difficult to minimize damage and alter that lead to cognitive impairment and AD, (2) they can
disease progression. Prevention of AD, therefore, ranks modify or alter the relation of pathology to cognition and
among the most significant public health challenges of the (3) they can be related to the clinical expression of AD
twenty-first century. Identification of risk factors for AD but unrelated to its pathology. We discuss neurobiological
is important and will facilitate novel strategies to prevent mechanisms whenever possible because an understand-
or delay onset of disease. ing of how risk factors lead to AD is essential to develop
strategies for disease prevention and/or delaying onset of
dementia. Thus, we present findings from clinical–patho-
logic studies and focus in particular on the role of AD
44.2 RISK FACTORS FOR AD pathological hallmarks (i.e. amyloid plaques and neurofi-
brillary tangles) and cerebral infarcts, the most common
causes of cognitive impairment in old age.
In this chapter, we review the major factors associated with
the risk of AD, with an emphasis on those supported by
data from prospective studies of incident disease. Although
much of the initial information regarding risk factors came 44.3 GENETIC RISK FACTORS
from studies conducted on persons with prevalent disease,
such studies are susceptible to bias because they involve 44.3.1 APOLIPOPROTEIN E ε4 ALLELE
only a limited, non-random fraction of persons with AD.
Further, because disease prevalence is highly related to sur- Amyloid precursor protein, the presenilins and apolipo-
vival, population-based studies of prevalent disease can lead protein E: Several studies have examined the role of genetic
to the identification of factors that are associated with sur- factors as determinants of AD and mutations in three
vival after the onset of the disease rather than actual risk of genes are now known to cause AD (i.e. amyloid precursor
the disease. Studies conducted on incident disease, in which protein, presenilin 1 and presenilin 2 – see Chapter 50).
risk factors are identified prior to disease onset, are better Notably, however, these mutations account for only about
positioned for identifying true risk factors. Considerable 1% of AD cases, particularly early onset of AD. In contrast
data from such studies are now available and several factors to mutations, several polymorphisms are associated with an
have been identified, including genetic, experiential and increased risk of AD, but the apolipoprotein E gene is the
medical factors, with some increasing and others decreas- only consistently replicated risk factor for the most common
ing risk and many of which are modifiable. expression of AD, that which begins after 65 years of age.
447
448 Dementia

Persons with one or more copies of the ε4 allele have about 44.4.2 FEMALE SEX
a two- to three-fold greater risk of developing AD com-
pared with those without the ε4 allele (Evans et al., 1997a; Female sex is often considered a risk factor for AD, as the dis-
Jellinger, 2006; Murrell et al., 2006) and some data suggest ease is more common among women than men, particularly
that this association may be stronger among whites than in those over the age of 80 (Alzheimer’s Association, 2015).
African Americans. AD pathology plays an important role This sex difference appears to be applicable to increased
in the association of the ε4 allele with AD risk; ε4 is associ- longevity among women. Overall, studies of persons under
ated with the accumulation of AD pathology and findings 85 years of age do not suggest strong sex differences in AD
suggest that the ε4 allele works through amyloid deposition incidence and the situation after the age of 85 is uncertain
and subsequent tangle formation to cause cognitive impair- because of the paucity of men in this age group. It has been
ment (Bennett et al., 2003, 2005a). However, the ε4 allele hypothesized that women may be more vulnerable to AD
also increases the likelihood of cerebral infarcts, which also due to hormonal changes that follow menopause, but this
contributes to AD (Schneider et al., 2005). Altogether, these remains unclear. One clinical–pathologic study suggested
findings suggest that the ε4 allele leads to accumulation of that AD pathology was more likely to be expressed as a clin-
AD pathology but also interacts with other risk factors to ical dementia syndrome in women than in men and these
cause cognitive impairment and AD. findings may be due to possible sex differences in amyloid
precursor protein processing (Barnes et al., 2005).

44.3.2 APOLIPOPROTEIN E ε2 ALLELE 44.4.3 EDUCATION


In contrast to the ε4 allele, which increases the risk of AD, Many studies have shown that the risk of AD is reduced
the presence of the ε2 allele is associated with a reduced risk among persons with higher levels of education (Evans et al.,
of AD (Wilson et al., 2002c; Jellinger, 2006). Although data 1997b; Karp et al., 2004; Caamano-Isorna et al., 2006).
are limited because ε2 allele is uncommon, persons with Notably, however, a review study concluded that education
ε2 allele have about a 40% lower risk of AD compared with is not related to the rate of cognitive decline (Wilson et al.,
those without the allele (Farrer et al., 1997). Although vari- 2009b), the primary clinical manifestation of AD, suggest-
ous mechanisms have been proposed, clinical–pathologic ing that the association of education with AD is largely due
findings suggest that the ApoE genotype affects the risk of to its robust association with the level of cognitive function.
AD primarily by affecting the accumulation of AD pathol- Nevertheless, it has been hypothesized that education con-
ogy (Bennett et al., 2003, 2005a). tributes to neural reserve by somehow reducing the adverse
Other genetic variants: recent studies have identified sev- clinical impact of mild to moderate pathologic burden.
eral other genetic variants including complement receptor 1 Consistent with this idea, clinical–pathologic studies have
(CR1) and phosphatidylinositol-binding clathrin assembly shown that education reduces the association of AD patho-
protein (PICALM) associated with AD risk (Chibnik et al., logic burden, particularly amyloid, with the level of cogni-
2011). These may also work via their effect on AD pathology, tion proximate to death (Bennett et al., 2005b).
particularly amyloid, but additional studies are needed to
fully elucidate mechanisms. 44.4.4 RACE/ETHNICITY
African Americans and Hispanics may have a substantially
increased risk of AD compared with non-Hispanic whites
44.4 EXPERIENTIAL FACTORS (Tang et al., 2001; Evans et al., 2003); however, it is difficult
to interpret from research conducted earlier, because race
44.4.1 AGE and ethnicity are entangled with socio-economic and cul-
tural variables that strongly influence cognitive test perfor-
Old age is often touted as the strongest risk factor for AD. mance and there are disparities in risk factors and clinical
Indeed, AD is fairly rare among younger persons, with presentations of disease (Barnes et al., 2014). Thus, the avail-
about 200,000 persons under the age of 65 currently affected able data on the association of race/ethnicity and AD should
in the United States, but the risk of AD increases dramati- be interpreted with caution.
cally after the age of 65 (Alzheimer’s Association, 2015).
Currently, in the United States, about 5.1 million persons 44.4.5 DEPRESSIVE SYMPTOMS AND
over the age of 65 have AD and the increase in risk rises con- LONELINESS
siderably with each decade of increase in age, with nearly
half of those over the age of 85 affected (Barnes et al., 2003; Data from several studies support a connection between
Alzheimer’s Association, 2015). Importantly, however, age depressive symptoms and the risk of AD (Wilson et al.,
is not the actual cause of AD, but rather a proxy for other 2002, 2003; Andersen et al., 2005), with one prospective
causes as yet unknown in the disease processes that lead to study reporting that the risk increased by about 20% for
the development of AD. each depressive symptom endorsed (Wilson et al., 2002a).
Risk factors for Alzheimer’s disease 449

In addition, a related psychological construct, referred to, 44.4.8 COGNITIVE ACTIVITY


as loneliness (i.e. the subjective sense of connectedness to
others) also is related to AD risk, even after controlling Considerable evidence suggests that cognitive activity and
depressive symptoms (Wilson et al., 2007c). Although, cognitive experiences in life are associated with a reduced
some have suggested that depressive symptoms and risk of AD in old age (Hall et al., 2000; Wilson et al., 2002b;
loneliness are a consequence of incipient dementia or its Boyle et al., 2012). In one study, a cognitively inactive per-
underlying pathology rather than a true risk factor, these son was 2.6 times more likely to develop AD than a cogni-
psychological factors do not change considerably with tively active person (Wilson et al., 2007d). Again, cognitive
advancing age and are not strongly related to AD pathol- activity is not related to AD pathology or infarcts (Wilson
ogy or other common dementia-related pathologies (i.e. et al., 2007d, 2013), suggesting that it must work via another
infarcts) (Wilson et al., 2003, 2008, 2015). Depressive mechanism to help maintain function even in the face of
symptoms and loneliness, therefore, are not merely early accumulating disease pathology.
symptoms of the disease and are likely to work via some
other mechanism to affect AD risk. 44.4.9 SOCIAL ENGAGEMENT AND
SOCIAL NETWORKS
44.4.6 NEUROTICISM Several studies have examined the association of social
engagement and AD (Fratiglioni et al., 2000; Barnes et al.,
The personality trait referred to as neuroticism (i.e. prone-
2004) and findings generally suggest that people with an
ness to psychological distress) is associated with an increased
extensive social network have a decreased risk of developing
risk of AD, such that, in one study a person with a high level
AD. Relatively, little is known about the potential neuro-
of neuroticism was more than twice as likely to develop
biological basis of this association, although one clinical–
AD compared to someone with a low level (Wilson et al.,
pathologic study reported that large social networks reduced
2006). Notably, however, distress proneness is unrelated to
the impact of AD pathology on the level of cognitive func-
measures of Alzheimer’s pathology and does not modify the
tion in older persons (Bennett et al., 2006). We suspect that
association of cognition with common neuropathologies
cognitive processing skills that allow people to develop and
(Wilson et al., 2005, 2007A). These results suggest that, as in
maintain large social networks help provide some form of
the case of depressive symptoms and loneliness, neurobio-
neural reserve, such that socially connected persons can tol-
logical mechanisms other than AD pathology must be the
erate greater amounts of AD pathology prior to developing
underlying cause for the association of this personality trait
clinical AD, but more research is necessary in this area.
with dementia. Chronic stress has been associated with a
spectrum of neurobiological changes in brain regions criti-
cal for memory and may damage selected neural systems in 44.4.10 PHYSICAL ACTIVITY
such a way that less dementia-related pathology is needed
Physical activity is associated with a reduced risk of AD in
to produce clinical dementia, but additional research is
some but not all prospective research conducted supports
warranted.
this (Podewils et al., 2005; Scarmeas et al., 2009; Buchman
et al., 2012). The mechanisms linking physical activity to
44.4.7 CONSCIENTIOUSNESS AND AD are not well-understood, but physical activity contrib-
PURPOSE IN LIFE utes to cardiovascular health and may partially reduce the
risk of AD via vascular mechanisms. However, physical
The personality trait of conscientiousness (i.e. the ten- activity also has been related to decreased production of
dency to be hard-working and focused) has been shown to AD pathology in transgenic animal models and additional
be associated with a considerable reduction in the risk of research needs to be conducted to establish the neurobiol-
AD, such that in one study, a person with a high score was ogy underlying the association of physical activity and AD.
89% was less likely to develop AD compared to someone
with a low score (Wilson et al., 2007e). Similarly, a related 44.4.11 VITAMIN INTAKE AND DIET
construct referred to as purpose in life (i.e. the tendency
to derive a sense of meaning in life and be intentional and Several population-based studies have examined the asso-
goal-focused) is associated with a substantially reduced ciation between vitamin intake, particularly E, C and other
risk of AD (Boyle et al., 2010). Both conscientiousness and antioxidants and diet with the risk of AD, but results are
purpose in life are unrelated to AD pathology but modify mixed (Kawas, 2006; Morris, 2009). Perhaps, more com-
the association of AD pathology with the level of cogni- pelling evidence suggests that high saturated or trans-fatty
tion proximate to death (Wilson et al., 2002d, 2007e). acids increase the risk of AD and high polyunsaturated
This ­suggests that conscientiousness and purpose in life or monounsaturated fatty acids decrease risk (Morris and
provide a buffer against AD and help maintain cogni- Tagney, 2014). Towards that end, recent data suggests that
tive function even in the face of accumulating disease Mediterranean-style diets, which generally include olive
pathology. oil as the primary fat, high fish consumption and moderate
450 Dementia

consumption of wine with meals, as well as diets aimed to 44.5.3 OBESITY AND THE METABOLIC
combat hypertension, which include low consumption of SYNDROME
saturated fat and sweets and relatively higher consumption
of dairy, are associated with a reduced risk of AD (Morris Obesity and the metabolic syndrome (based on several com-
et al., 2015). Vitamins and diet are generally thought to mon disorders including obesity, hypertriglyceridemia, low
work via their antioxidant effects in the brain, in addition high-density lipoprotein levels, hypertension and hypergly-
to potential vascular benefits, but clinical–pathologic data caemia) appear to be associated with the risk of AD (Yaffe
are greatly needed. Moreover, data on vitamins and diet are et al., 2004; Kivipelto et al., 2005). The mechanisms under-
difficult to interpret, partially because vitamin users and lying these associations are unclear, but it is well established
careful eaters tend to differ in important ways compared that obesity has numerous adverse health effects and both
to those who do not use vitamins in their diet and precise obesity and the metabolic syndrome are associated with
measurement of vitamin intake is very difficult to ascertain other vascular risk factors and an increased risk of cerebro-
in large-scale trials. vascular disease. These conditions may increase the risk of
AD via their association with cerebrovascular disease or via
another pathologic process, but clinical–pathologic data are
44.5 MEDICAL RISK FACTORS needed to elucidate these mechanisms.

44.5.1 CEREBROVASCULAR DISEASE/ 44.5.4 OLFACTORY DYSFUNCTION


STROKE Prospective studies have shown that olfactory impairment
(e.g. difficulty identifying familiar odours) is associated with
Although cerebrovascular disease is often considered a
the development of AD and its precursor, mild cognitive
pathologically distinct entity (i.e. vascular dementia) or a
impairment (Wilson et al., 2007b, 2007f, 2009a). Clinical–
contributing cause of dementia (i.e. mixed dementia), cere-
pathologic studies have shown that impaired odour recogni-
brovascular disease itself is associated with a substantially
tion is robustly related to level of AD pathologic changes in the
increased risk of AD (Honig et al., 2003). Consistent with
brain, particularly, density of neurofibrillary tangles in central
these findings, clinical–pathologic data suggest that cerebral
olfactory regions (Wilson et al., 2007b) and this association is
infarcts work additively with AD pathology and indepen-
seen even in older people without apparent cognitive dysfunc-
dently contribute to likelihood of clinical AD (Arvanitakis
tion and after using a controlling apparatus for other behav-
et al., 2006). Additional research is needed to further clar-
ioural and genetic risk factors (Wilson et al., 2009), suggesting
ify exactly how cerebrovascular disease and AD pathology
that olfactory dysfunction is an early sign of impending AD.
work together to increase the risk of AD.

44.5.5 INFLAMMATORY MARKERS
44.5.2 OTHER VASCULAR FACTORS
Increasing evidence suggests that inflammation may play an
High blood pressure, diabetes, heart disease and hypercho- important role in the pathogenesis of AD and findings from
lesterolemia are associated with an increased risk of AD some prospective studies suggest that the level of systemic
(Kivipelto et al., 2005; Arvanitakis et al., 2006). Blood pres- markers of inflammation, particularly C-reactive protein
sure rates in AD is the most well-studied and high blood and interleukin-6 are associated with an increased risk of AD
pressure in midlife is predictive of an increased risk of AD, (Barnes et al., 2014). Several potential mechanisms form the
but the role of late-life blood pressure in relation to AD is underlying causes for the association between inflammation
less clear. Diabetes also has been consistently associated and AD. Inflammation can occur as a consequence of neuro-
with an increased risk of AD and cholesterol studies give degeneration or, alternatively, it may be a causal part of a cas-
mixed results. The mechanisms underlying these associa- cade of events that lead to the accumulation of AD pathology,
tions are likely to be complex. For example, as noted above particularly, amyloid deposition. Inflammation is also associ-
in connection to stroke, vascular risk factors are associated ated with vascular disease and may be related to dementia via
with the development of cerebrovascular disease and likely cerebrovascular disease. Additional research is needed to fur-
to increase AD risk via their association with cerebrovas- ther clarify the role of inflammation in the development of AD.
cular disease. Notably, however, clinical–pathologic studies
have reported mixed findings with regard to the association
of vascular risk factors with pathologic changes in the brain;
for example, diabetes has been inconsistently related to AD
44.6 CONCLUSION
pathology (Arvanitakis et al., 2006). Further, it is likely that
the associations of vascular factors with AD are multifacto- Using data from prospective studies of incident disease, sev-
rial, such that, both vascular factors add to and interact with eral factors associated with the risk of AD have been iden-
each other and potentially other risk factors (e.g. ApoE) to tified, including genetic, experiential and medical factors,
cause AD. many of which are modifiable (particularly, experiential
Risk factors for Alzheimer’s disease 451

factors). The relationships among risk factors, common Boyle, P.A., Buchman, A.S., Barnes, L.L. and Bennett, D.A.
age-related neuropathologies and cognitive impairment are (2010). Purpose in life is associated with a reduced
complex and in many cases unclear. However, studies have risk of Alzheimer’s disease and mild cognitive
shown that some risk factors work directly via their influence impairment among community based older persons.
on the accumulation of AD and other common age-related Archives of General Psychiatry, 67: 304–310.
pathologies (i.e. infarcts) and others modify the association Boyle, P.A., Buchman, A.S., Wilson, R.S. et al. (2012).
of cognition with pathology, such that they ­benefit cogni- Effect of purpose in life on the relation between
tion in the face of accumulating neuropathology. Risk fac- Alzheimer’s disease changes on cognitive function
tors that work via these mechanisms offer specific targets for in advanced age. Archives of General Psychiatry, 69
future interventions to prevent, delay, or reduce the burden (5): 499–505.
posed by AD and warrant particular focus in behavioural Buchman, A.S., Boyle, P.A., Yu, L. et al. (2012). Association
intervention and pharmacological studies. of total daily physical activity with the risk of AD and
rate of cognitive decline in older adults. Neurology,
78: 1323–1329.
REFERENCES Caamano-Isorna, F., Corral, M., Montes-Martinez, A. et al.
(2006). Education and dementia: A meta-analytic
Alzheimer’s Association. (2015). Alzheimer’s Disease Facts study. Neuroepidemiology, 26 (4): 226–232.
and Figures. Available at www.alz.org. Chibnik, L.B., Shulman, J.M., Leurgans, S.E. et al. (2011).
Andersen, K., Lolk, A., Kragh-Sorensen, P. et al. (2005). The Alzheimer’s susceptibility locus CR1 is associated
Depression and the risk of Alzheimer disease. with increased amyloid plaque burden and age-
Epidemiology, 16 (2): 233–238. related cognitive decline. Annals of Neurology, 69:
Arvanitakis, Z., Schneider, J.A., Wilson, R.S. et al. (2006). 560–569.
Diabetes is related to cerebral infarction but not Evans, D., Beckett, L., Field, T. et al. (1997a).
to AD pathology in older persons. Neurology, 67: Apolipoprotein E e4 and incidence of Alzheimer’s
1960–1965. disease in a community population of older persons.
Barnes, L.L. and Bennett, D.A. (2014). Alzheimer’s disease JAMA, 277: 822–824.
in African Americans: Risk factors and challenges for Evans, D.A., Bennett, D.A., Wilson, R.S. et al. (2003).
the future. Health Affairs, 33: 580–586. Incidence of Alzheimer disease in a biracial urban
Barnes, L.L., Mendes de Leon, C.F., Wilson, R.S. et al. community: Relation to apolipoprotein E allele status.
(2004). Social resources and cognitive decline in a Archives of Neurology, 60 (2): 185–189.
population of older African Americans and whites. Evans, D., Hebert, L., Beckett, L. et al. (1997b). Education
Neurology, 63: 2322–2326. and other measures of socioeconomic status and risk
Barnes, L.L., Wilson, R.S., Bienias, J.L. et al. (2005). Sex of incident Alzheimer disease in a defined popula-
differences in the clinical manifestations of Alzheimer tion of older persons. Archives of Neurology, 54 (11):
disease pathology. Archives of General Psychiatry, 62 1399–1405.
(6): 685–691. Farrer, L.A., Cupples, L.A., Haines, J.L. et al. (1997). Effects
Barnes, L.L., Wilson, R.S., Schneider, J.A. et al. (2003). of age, sex, and ethnicity on the association between
Gender, cognitive decline, and risk of AD in older apolipoprotein E genotype and Alzheimer disease.
persons. Neurology, 60: 1777–1781. A meta-analysis. APOE and Alzheimer Disease Meta
Bennett, D.A., Schneider, J.A., Tang, Y. et al. (2006). The Analysis Consortium. JAMA, 278 (16): 1349–1356.
effect of social networks on the relation between AD Fratiglioni, L., Wang, H.X., Ericsson, K. et al. (2000).
pathology and level of cognitive function in old peo- Influence of social network on occurrence of demen-
ple: A longitudinal cohort study. Lancet Neurology, 5 tia: A community-based longitudinal study. The
(5): 406–412. Lancet, 355: 1315–1319.
Bennett, D.A., Schneider, J.A., Wilson, R.S. et al. (2005a). Hall, K.S., Gao, S., Unverzagt, F.W. et al. (2000). Low educa-
Amyloid mediates the association of apolipoprotein tion and childhood rural residence: Risk for AD in
E e4 allele to cognitive function in older people. African Americans. Neurology, 54: 95-99.
Journal of Neurology Neurosurgery and Psychiatry, Honig, L.S., Tang, M.X., Albert, S. et al. (2003). Stroke and
76 (9): 1194–1199. the risk of Alzheimer disease. Archives of Neurology,
Bennett, D.A., Schneider, J.A., Wilson, R.S. et al. (2005b). 60 (12): 1707–1712.
Education modifies the association of amyloid but not Jellinger, K.A. (2006). Alzheimer 100 – Highlights in the
tangles with cognitive function. Neurology, 65 (6): history of Alzheimer research. Journal of Neural
953–955. Transmission, 113 (11): 1603–1623.
Bennett, D.A., Wilson, R.S., Schneider, J.A. et al. (2003). Karp, A., Kareholt, I. and Oui, C. (2004). Relation of educa-
Apolipoprotein E epsilon4 allele, AD pathology, tion and occupation-based socioeconomic status to
and the clinical expression of AD. Neurology, 60 (2): incident AD. American Journal of Epidemiology, 159
246–255. (2): 175–183.
452 Dementia

Kawas, C.H. (2006). Medications and diet: Protective pathology and odour identification in old age.
factors for AD? Alzheimer Disease and Associated Journal of Neurology Neurosurgery and Psychiatry,
Disorder, 20 (3 Suppl. 2): S89–S96. 78: 30–35.
Kivipelto, M., Ngandu, T., Fratiglioni, L. et al. (2005). Wilson, R.S., Barnes, L.L., Bennett, D.A. et al. (2005).
Obesity and vascular risk factors at midlife and the Proneness to psychological distress and risk of
risk of dementia and Alzheimer disease. Archives of Alzheimer disease in a biracial community. Neurology,
Neurology, 62 (10): 1556–1560. 64 (2): 380–382.
Morris, M.C. (2009). The role of nutrition in Alzheimer’s Wilson, R.S., Barnes, L.L., Mendes de Leon, C.F. et al.
disease: Epidemiological evidence. European Journal (2002a). Depressive symptoms, cognitive decline,
of Neurology, 16 (Suppl. 1): 1–7. and risk of AD in older persons. Neurology, 59 (3):
Morris, M.C. and Tagney, C.C. (2014). Dietary fat composi- 364–370.
tion and dementia risk. Neurobiological Aging, 35 Wilson, R.S., Bennett, D.A., Bienias, J.L. et al. (2002b).
(Suppl. 2): S59–S64. Cognitive activity and incident AD in a population-
Morris, M.C., Tangney, C.C., Wang, Y. et al. (2015). MIND based sample of older persons. Neurology, 59 (12):
diet associated with reduced incidence of Alzheimer’s 1910–1914.
disease. Alzheimer’s Dementia, 11 (9): 1007–1014. Wilson, R.S., Bienias, J.L., Berry-Kravis, E. et al. (2002c).
Murrell, J.R., Price, B., Lane, K.A. et al. (2006). Association The apolipoprotein E epsilon 2 allele and decline
of apolipoprotein E genotyoe and Alzheimer disease in episodic memory. Journal of Neurology,
in African Americans. Archives of Neurology, 63 (3): Neurosurgery and Psychiatry, 73 (6): 672–677.
431–434. Wilson, R.S., Boyle, P.A., Capuano, A.W. et al. (2015).
Podewils, L.J., Guallar, E., Kuller, L.H. et al. (2005). Physical Late-life depression is not associated with demen-
activity, APOE genotype, and dementia risk: Findings tia-related pathology. Neuropsychology, 30 (2):
from the Cardiovascular Health Cognition Study. 135–142.
American Journal of Epidemiology, 161 (7): 639–651. Wilson, R.S., Hebert, L.E., Scherr, P.A. et al. (2009b).
Scarmeas, N., Luchsinger, J.A., Schupf, N. et al. (2009). Educational attainment and cognitive decline in old
Physical activity, diet, and risk of Alzheimer disease. age. Neurology, 72 (5): 460–465.
JAMA, 302 (6): 627–637. Wilson, R.S., Krueger, K.R., Arnold, S.E. et al. (2007c).
Schmidt, R., Schmidt, H., Curb, J.D. et al. (2002). Early Loneliness and risk of AD. Archives of General
inflammation and dementia: A 25-year follow-up of Psychiatry, 64: 234–240.
the Honolulu-Asia Aging Study. Annals of Neurology, Wilson, R.S., Mendes De Leon, C.F., Barnes, L.L. et al.
52 (2): 168–174. (2002d). Participation in cognitively stimulating activi-
Schneider, J.A., Bienias, J.L., Wilson, R.S. et al. (2005). The ties and risk of incident Alzheimer disease. JAMA,
apolipoprotein E epsilon4 allele increases the odds 287 (6): 742–748.
of chronic cerebral infarction detected at autopsy in Wilson, R.S., Scherr, P.A., Schneider, J.A. et al. (2007d).
older persons. Stroke, 36 (5): 954–959. Relation of cognitive activity to risk of developing
Tang, M.X., Cross, P., Andrews, H. et al. (2001). Incidence Alzheimer disease. Neurology, 69 (20): 1911–1920.
of AD in African-Americans, Caribbean Hispanics, and Wilson, R.S., Schneider, J.A., Arnold, S.E. et al. (2007e).
Caucasians in northern Manhattan. Neurology, 56 (1): Conscientiousness and the incidence of Alzheimer
49–56. disease and mild cognitive impairment. Archives of
Wilson, R.S., Arnold, S.E., Beck, T.L. et al. (2008). Change in General Psychiatry, 64 (10): 1204–1208.
depressive symptoms during the prodromal phase of Wilson, R.S., Schneider, J.A., Arnold, S.E. et al. (2007f).
Alzheimer disease. Archives of General Psychiatry, 65 Olfactory identification and incidence of mild cogni-
(4): 439–445. tive impairment in older age. Archives of General
Wilson, R.S., Arnold, S.E., Schneider, J.A. et al. (2006). Psychiatry, 64 (7): 802–808.
Chronic psychological distress and risk of AD in old Wilson, R.S., Schneider, J.A., Bienias, J.L. et al. (2003).
age. Neuroepidemiology, 27 (3): 143–153. Depressive symptoms, clinical AD, and cortical
Wilson, R.S., Arnold, S.E., Schneider, J.A. et al. (2009a). plaques and tangles in older persons. Neurology, 61
Olfactory impairment in presymptomatic Alzheimer’s (8): 1102–1107.
Disease. Annals of the New York Academy of Wilson, R.S., Segawa, E., Boyle, P.A. and Bennett, D.A.
Sciences, 1170: 730–735. (2013). Lifespan cognitive activity, neuropathologic
Wilson, R.S., Arnold, S.E., Schneider, J.A. et al. (2007a). burden, and cognitive aging. Neurology, 81 (4):
Chronic distress, age-related neuropathology, and late- 314–321.
life dementia. Psychosomatic Medicine, 69 (1): 47–53. Yaffe, K., Kanaya, A., Lindquist, K. et al. (2004). The meta-
Wilson, R.S., Arnold, S.E., Schneider, J.A. et al. (2007b). bolic syndrome, inflammation, and risk of cognitive
The relation between cerebral Alzheimer’s disease decline. JAMA, 292 (18): 2237–2242.
45
The natural history of Alzheimer’s disease

JODY COREY-BLOOM AND MICHAEL S. RAFII

methods in prodromal form of AD and, recently, in pre-


45.1 INTRODUCTION clinical AD. Nonetheless, a firm understanding of the
clinical manifestations and natural progression of AD
Even if new disease-modifying treatments are developed, dementia are required to approach patients practically,
Alzheimer’s disease (AD) will continue to be an enormous thoughtfully and comprehensively.
public health problem. Knowledge of the clinical features Memory loss is the cardinal and commonest present-
and natural history of AD will facilitate planning in response ing complaint in AD. Initially, the patient has difficulty
to the increased demands of medical and social resources. ­recalling new information such as names or details of con-
In addition, this information will enable clinicians to pro- versation, while remote memories are relatively preserved.
vide patients and families with a more reliable prediction The pattern of memory impairment in AD is quite distinc-
of the course of the disease for treatment options, as well tive. Declarative memory (facts [semantic] and events [epi-
as financial and family planning. Furthermore, monitor- sodic]), which depends on medial temporal structures such
ing the stages of AD is important in recruiting patients for as the entorhinal cortex and hippocampus, is profoundly
clinical studies at definable levels of severity and for under- affected in AD, while subcortical systems supporting pro-
standing the expected course of decline related to putative cedural memory are relatively spared until quite late in the
treatment effects. disease (Carlesimo et al., 1992).
Semantic and episodic memory are both profoundly
impaired in early AD. Within episodic memory, there
is a distinction between immediate recall (e.g. mental
45.2 CLINICAL FEATURES rehearsal of a phone number), memory for recent events
(within the past 2 weeks) and memory of remote events.
The diverse spectrum of symptoms of AD reflects dys- Memory for recent events, served by the hippocampus
function of widespread regions of the cerebral cortex. and entorhinal cortex in the medial temporal lobe, is
Symptoms begin insidiously, making it difficult to date prominently impaired in early AD. In contrast, immedi-
the onset of cognitive and functional decline precisely. ate memory (encoded in the prefrontal cortices) is spared
Progression is generally gradual, yet can be interleaved early on, as are memories that are consolidated for long
with occasional plateaus; however, reliable measurement periods (years), which can be recalled in the absence of
of clinical disease progression in AD is difficult because hippocampal functioning (Zola-Morgan et al., 1986).
of variability between and within subjects. Converging The most precise way to describe the early memory defi-
evidence from longitudinal studies of clinically normal cit in AD is therefore an anterograde episodic amnesia.
elderly and familial AD cohorts strongly suggests that the The symptomatic manifestations of this type of memory
AD pathophysiological process begins decades before the failure include: forgetting important dates or events, ask-
manifestation of clinical dementia. Hypothetical models ing for the same information over and over again, relying
have been proposed in which biomarkers of AD become heavily on memory aides, trouble following a familiar rec-
increasingly abnormal in an ordered manner as the disease ipe or keeping track of monthly bills, trouble remember-
progresses and symptoms emerge (Jack et al., 2010). The ing the rules of a favourite game, forgetting where things
field of AD has, therefore, steadily marched earlier in the are or how they got there, losing objects and being unable
disease continuum, evaluating diagnostic and therapeutic to go back the steps to find them again. With progression,

453
454 Dementia

the memory loss worsens to include remote memory gain, loss of libido, lack of interest and reduced motor ­activity
(Wilson et al., 1983; Sullivan et al., 1986; Cummings and are common to both AD and depression, may account for
Cole, 2002). the high rate of ‘depression’ found in some studies (Lazarus
Early in AD, judgement and abstraction are often et al., 1987; Becker et al., 1994; Jost and Grossberg, 1996). In
impaired, suggesting involvement of the frontal lobes. patients with severe dementia, there is an apparent paucity
Social comportment and interpersonal skills are often strik- of depressive symptoms. A likely explanation is that certain
ingly preserved and may remain relatively intact long after features of depression (guilt, hopelessness, anxiety) are less
memory and insight are lost. The ability to conduct one’s discernible in subjects with severe dementia. But, symptoms
self in brief social interactions despite significant underly- such as crying and diminished sleep can be expressed even
ing impairment, however, is often superficial and can be at later stages of AD.
identified by the astute clinician as somewhat of a façade. Psychosis is frequent in people with AD (Schneider et al.,
Language is frequently normal in the early stages of AD, 2004). One observational study of 329 patients who did not
although, reduced conversational output (logopaenia) may have psychosis at initial evaluation demonstrated a cumu-
be noted. When asked to generate word lists in 1 minute, lative incidence of hallucinations and delusions of 20% in
patients with AD dementia perform significantly worse on a 1 year, 36% in 2 years and 50% in 3 years (Paulsen et al.,
category fluency test (e.g. lists of animals) than on a letter flu- 2000). The prevalence of psychotic symptoms in AD, the
ency test (e.g. lists of words beginning with F). This reflects relationship of hallucinations and delusions to the severity of
the specific deficit in semantic memory (Canning et al., cognitive impairment and the influence of psychosis on the
2004). As dementia progresses, many patients become more rate of cognitive decline are controversial. Other estimates
recognizably aphasic. Initially, this manifests as dysnomia of the prevalence of psychotic symptoms in AD range from
and mild loss of fluency with paraphasic errors and relative 10% to 73%, with most in the range of 28%–38% (Wragg and
preservation of repetition, a pattern resembling transcorti- Jeste, 1989; Mega et al., 1999). Delusions have been reported
cal sensory aphasia (Cummings et al., 1985; Cummings and in 13%–75% of AD patients and hallucinations, more com-
Cole, 2002). At a later stage, language is obviously dysfluent monly visual than auditory, in 3%–50% (Mayeux et al.,
and repetition impaired; terminally, a state of near-mutism 1985; Cummings et al., 1987; Reisberg et al., 1987; Drevets
may occur (Murdoch et al., 1987). and Rubin, 1989; Reisberg et al., 1989; Becker et al., 1994;
Visuospatial impairment in AD results in symptoms Jost and Grossberg, 1996; Cook et al., 2003). This substantial
such as misplacing objects or getting lost and difficulty variation in frequencies is probably due to different method-
with tasks such as recognizing and drawing complex figures ologic approaches, especially in defining delusions and hal-
(Brouwers et al.,1984; Kavcic and Duffy, 2003). Difficulty lucinations and due to differences in the range of severity of
with calculation (affecting skills such as handling money), AD in each series. Although psychotic symptoms may occur
apraxia and agnosia are further problems that develop in throughout the course of AD, they have their highest fre-
AD. Apraxia may impair activities such as operating appli- quency at a moderate level of dementia (Drevets and Rubin,
ances or dressing; as might be expected, more complex 1989). The manifestation of psychosis in dementia can result
skills tend to break down first, while highly overlearned in significant distress for patients with AD, as well as for
motor tasks (e.g. playing a musical instrument, using tools) caregivers (Steele et al., 1990). These symptoms constitute
may be retained until, relatively, later years (Rapcsak et al., the primary reason for transfer of patients with dementia
1989; Helmes and Ostbye, 2002). Agnosia develops in any- from general hospitals to psychiatric hospitals and for their
time between the middle to late stages of AD and includes institutionalization (Hebert et al., 2001).
features such as failing to recognize family members. In addition to depression and psychotic symptoms, AD
Behavioural or psychiatric symptoms occur f­requently patients show a wide range of behavioural abnormalities
in AD (see Chapters 7 and 8). Over and above, a gen- including agitation, wandering, sleep disturbances and dis-
eral decline of activity and anhedonia in virtually all inhibition (Becker et al., 1994; Jost and Grossberg, 1996;
AD patients, depressive symptoms occur in about 25%, Lyketsos et al., 2002). These often impose a significant burden
although severe depression is uncommon (Cummings et al., on caregivers and may precipitate nursing home placement.
1987; Rapcsak et al., 1989; Rosen and Zubenko, 1991; Becker In contrast to psychotic symptoms, behavioural disturbances
et al., 1994; Cummings and Cole, 2002; Helmes and Ostbye, are more clearly associated with the degree of dementia. For
2002; Starkstein et al., 2008; Snowden et al., 2015). The fre- example, a study of 127 AD patients found that the overall
quency of significantly depressed mood in patients with AD number of behavioural problems increased significantly with
ranged from 0% to 87% in 30 studies reviewed by Wragg worsening cognitive function (Teri et al., 1988). Agitation
and Jeste (1989) with higher rates reported from acute care includes physical aggression, verbal a­ggression and non-
facilities than in studies conducted on outpatient research aggressive behaviours. Physically non-aggressive behaviours,
sites. While many AD patients have depressive symptoms, such as motor restlessness and pacing, are the most common
most do not display enough features to meet Diagnostic forms of agitation in outpatients with AD. However, physi-
and Statistical Manual of Mental Disorders, Fourth Edition cal aggression is the most distressing behaviour to custo-
(DSM-IV) criteria for major depressive disorders. The fact dians and occurs in about 20% of patients (Reisberg et al.,
that vegetative symptoms, such as sleep disturbance, weight 1987; Burns et al., 1990; Hooker et al., 2002). Various factors
The natural history of Alzheimer’s disease 455

appear to be important predictors of aggressive behaviour, visual field defects. This constellation of findings implicates
including premorbid history of aggression, a troubled pre- the dorsal visual stream in the lateral occipital and parieto-
morbid relationship between the caregiver and patient and a occipital regions (Aharon-Peretz et al., 1999).
greater number of social and medical problems. Wandering Another variant of AD is the so-called Lewy body vari-
behaviour affects 3%–26% of AD outpatients. Insomnia and ant (LBV) of AD (see Chapters 61 through 63) with promi-
sleep disturbances are inconstant features of AD and sexual nent deficits in visuospatial function relative to amnesia
disinhibition is reported in less than 10% of patients (Burns (Katzman et al., 1995; Hansen, 1997; Connor et al., 1998).
et al., 1990; Becker et al., 1994). Thus, behavioural symptoms Compared to patients with AD alone, those with LBV have
are a pervasive and poorly understood concomitant symp- a greater frequency of extrapyramidal manifestations and
tom of cognitive deterioration in AD. subcortical deficits, somewhat better recall selected mem-
The incidence of seizures in patients with AD is greater ory tasks and a significantly lower frequency of neocortical
than that expected in the elderly population, with an inci- neurofibrillary tangles at autopsy.
dence rate of 484 per 100,000 person-years (95% confidence As neuronal degeneration progresses in AD, all symp-
interval [CI] = 287–784) (Irizarry et al., 2012). Independent toms worsen and eventually patients become uncommuni-
risk factors for new-onset seizures in AD include younger cative and unable to care for themselves, walk, or maintain
age, greater degree of cognitive impairment and history of continence. They require total care, including feeding and
antipsychotic usage. are often institutionalized. During end-stage AD, death
Excluding mental status testing, the neurologic exami- usually results from the complications of being bed-bound,
nation is usually normal in AD and the presence of sig- such as aspiration pneumonia, urinary tract infections, sep-
nificant or lateralizing abnormalities often suggests other sis or pulmonary embolism (Molsa et al., 1986).
diagnoses. However, primitive reflexes (snout, glabellar,
grasp), impaired graphesthaesia and an abnormal response
to double simultaneous stimulation (face-hand test) are
frequently encountered in AD (Galasko et al., 1990; Becker
45.3 CLINICAL UTILITY OF ASSESSING
et al., 1994). Variable features, including extrapyramidal
CHANGE
signs (EPS – rigidity and bradykinesia), gait disturbances
and myoclonus may occasionally be seen early in the course The course of AD and the combination of symptoms that
of AD and increase in prevalence with the severity of AD manifest in each patient are markedly heterogeneous; how-
(Stern et al., 1987). The frequency of EPS in patients with ever, there are several direct clinical applications of track-
AD was as low as 12% to 14% in some series (Galasko et al., ing change. First, in early cases of AD, where impairment
1990; Burns et al., 1991; Becker et al., 1994) and as high is mild, documenting progression helps to confirm or
as 28%–92% in others (Mayeux et al., 1985; Molsa et al., establish the diagnosis. Clearly, progressive cognitive dete-
1986; Bakchine et al., 1989; Lopez et al., 2000), depending rioration is incompatible with normal ageing, depression,
on the severity of dementia in patients who were studied. delirium, or a static encephalopathy. The clinical follow-up
In intermediate or advanced stages of dementia, patients of patients who are initially classified as having AD greatly
often develop non-specific impairment of gait and balance, increases diagnostic accuracy.
leading to an increased risk of falls. Myoclonus develops Second, progression to a greater degree than expected
late in the course of AD and longitudinal studies report an may prompt the physician to look for a superimposed factor
increase in its frequency during follow-up (Mayeux et al., that may be treatable, such as hypothyroidism, intercurrent
1985; Tschampa et al., 2001). Its prevalence in AD has varied infection, medication toxicity or depression. Improvement
from 0% to 68%, although the usual reported rate is about or stabilization of psychometric scores indicates a favour-
10% (Hauser et al., 1986). able response to treating a remediable factor contributing
A minority of AD cases do not present the classic fashion to dementia.
with progressive amnestic dementia. One variant of AD is Third, defining the anticipated course of AD in detail
posterior cortical atrophy (PCA) (see Chapter 77). This syn- enables clinicians to provide patients and families with real-
drome manifests with progressive cortical visual impair- istic expectations regarding the disease and aids in making
ment. Patients are often first evaluated by optometrists or decisions for day care or institutionalization.
ophthalmologists for visual complaints such as difficulty in Longitudinal cognitive change may be fruitfully applied
reading and driving. Detailed neurologic evaluation may to several problems in clinical research. Change over time
elicit elements of the Balint syndrome: simultanagnosia (the on mental status scores may be used to construct models
inability to integrate a visual scene despite adequate acuity of the clinical course of ‘typical’ AD. This methodological
to resolve individual elements), optic ataxia (the inability to approach has ramifications, such as testing whether genetic/
reach accurately under visual guidance) and ocular apraxia environmental factors or experimental treatment influences
(the inability to gaze directly and accurately at a new tar- the rate of progression of dementia.
get, frequently leading to difficulty in reading). Other fea- Attempts to refine the natural history of AD have uti-
tures include visual agnosia, prosopagnosia and visual field lized two basic methods: global staging systems and serial
neglect. Less common are visual hallucinations and frank changes on psychometric test scores.
456 Dementia

45.3.1 MEASURING CHANGE WITH The GDS divides AD into a succession of major clinically
GLOBAL STAGING SYSTEMS distinguishable stages (further subdivided into 16) ranging
from normal cognition (GDS 1) to severe dementia (GDS 7)
Staging systems for AD comprise information on intellec- (Reisberg et al., 1982). Criteria for many of the stages combine
tual, social and community functioning obtained by narra- history (initially of memory decline, later of functional impair-
tion of history rather than formal rating scales. Two widely ment, progressing to loss of self-care, language and gait), and
used schemes are the Washington University’s Clinical mental status examination. Stages defined by this integration
Dementia Rating (CDR) (Hughes et al., 1982; Morris, 1997) of history, ADL and cognition should show high correlations
and the Global Deterioration Scale (GDS) (Reisberg et al., with mental state screening tests: indeed, the GDS correlates
1982, 1986), which divides AD into a succession of stages. well with the Mini-Mental State Examination (MMSE) (r =
These scales reflect an overall indirect evaluation of cog- 0.9, p < .001) and the BIMC (r = 0.8, p < .001) (Reisberg et al.,
nition, primarily obtained from caregivers, rather than a 1989). GDS stages 1 and 2 correspond to CDR 0 representing
direct assessment of the patient, but are able to assess the normal ageing, and GDS 3 is equivalent to CDR 0.5. In a lon-
influence of cognitive loss on the ability to conduct every- gitudinal study, only 5% of GDS 2 subjects became demented
day activities. They provide information about clinically over 3–4 years, whereas 15% of GDS 3 subjects progressed to
­meaningful function and behaviour and are less affected more advanced stages of dementia (Reisberg et al., 1986).
by ‘floor’ and ‘ceiling’ effects commonly associated with The GDS has similar virtues and drawbacks in compari-
­psychometric tests. son to the CDR, namely, difficulty in defining a natural his-
The CDR incorporates information from patients and tory over periods shorter than a year or two. The primary
informants concerning 6 areas of mental function: memory, use of these staging systems may be research in progress and
orientation, judgment and problem-solving, community in following patients observed longitudinally over extended
affairs, home and hobbies and personal care. Each area is periods, since the transitions from one stage to the next
scored as 0 = normal, 0.5 = questionable, 1 = mild, 2 = mod- occur over relatively long intervals.
erate, or 3 = severe impairment and integrated to obtain an
overall stage of dementia, using similar definitions of 0, 0.5
and 1 to 3. Inter-rater reliability is high, with a correlation of 45.3.2 MEASURING CHANGE BY RATE OF
0.91 and the CDR has been standardized for multicentre use DECLINE ON COGNITIVE TESTS
(Morris et al., 1997). Since the CDR includes cognitive items
and activities of daily living (ADL), it is not surprising that As a more flexible longitudinal index of brain function, some
it correlates well with the cognitive Blessed Information- studies have measured the annual rate of change (ARC) of
Memory-Concentration (BIMC) scale (r = 0.55) and with the mental status test scores in AD. Longitudinal data exist for
ADL-based Blessed Dementia Scale (BDS) (r = 0.53) (Berg et ARC on five mental status tests in common use: the BIMC
al., 1988). These authors followed a cohort of 43 initially mild test, the MMSE, the Dementia Rating Scale (DRS), the
AD patients (CDR 1) for 90 months. Over 67% of patients Cambridge Cognitive Examination (CAMCOG) (Roth et al.,
progressed to CDR 3 (severe dementia) in 50 months and 1986) and the Alzheimer’s Disease Assessment Scale (ADAS)
over 80% in 66 months. Many patients in this CDR 1 group (Rosen et al., 1984). Unfortunately, although these rates of
reached end points: 50% of patients were institutionalized in decline are available, their generalizability is uncertain.
40 months and 80% in 60 months. Thirty per cent of patients Katzman et al. (1988) compared four groups of subjects
died in 40 months and 40% in 72 months. Although, the with AD on the BIMC, with follow-up over 1 year (Reisberg
CDR is a useful staging instrument and has become widely et al., 1989). These cohorts differed substantially with regard
accepted in the clinical setting as a reliable and valid global to age, education, sex, severity of dementia and place of resi-
assessment measure for AD, movement from one stage to the dence and included community-dwelling and institution-
next may take several years (Storandt et al., 2002). Focus has, alized subjects seen in private practice or public hospital
therefore, shifted to the CDR sum of boxes score (CDR-SB), settings. The mean rate of change on the BIMC did not differ
which sums the ratings for the six cognitive and functional significantly among the four groups for subjects who ini-
domains and permits more sensitive measurement of annual tially made 24 or fewer errors (out of a possible 33). In those
or short-term rate of change in AD. Using data from 792 patients, the ARC on the BIMC was 4.4 errors per year and
participants enrolled in longitudinal studies at an academic did not vary with age, education or place of residence. Among
AD research centre, one recent study (Williams et al., 2013) demented subjects with an initial BIMC score of more than
found that the annual rate of change on CDR-SB scores was 24 errors, the rate of change was lower, presumably, because
1.43 (standard error [SE = 0.05]) in the CDR 0.5 sample and the test had reached a floor. A similar overall rate of pro-
1.91 (SE = 0.07) in the CDR 1 sample. Two other studies have gression for the BIMC was found in three further studies in
demonstrated annualized rates of change on the CDR-SB of community-dwelling AD patients (Thal et al., 1988; Ortof
1.0–1.13 and 2.28 in the Consortium to Establish a Registry and Crystal, 1989; Salmon et al., 1990). Significant variation
for Alzheimer’s Disease (CERAD) registry database (Rossetti in the mean ARC on the BIMC has been observed, however,
et al., 2010) and Cache County Dementia Progression Study by other investigators. For example, Lucca et al. (1993) noted
(Tschanz et al., 2011), respectively. a change of only 2.6 (± 4.9) points on the BIMC over 1 year
The natural history of Alzheimer’s disease 457

in a group of 56 patients with AD comprising outpatients ‘floor’ on initial evaluation and declined further over 1 year.
and inpatients at geriatric institutions. Using data from the The initial mean MMSE for this group was 10 (± 5.9), lower
California Alzheimer’s Disease Diagnostic and Treatment than in studies cited above and changed by an average of 3.5
Centers, we observed an annual rate of change of 2.8 points points (Burns et al., 1991). In a study of 95 less-demented
on the BIMC (Corey-Bloom et al., 1993a). subjects (mean MMSE of 19.6 ± 3.8), Swanwick et al. (1998)
For the MMSE, a number of studies have reported a mean reported an annual decline on the CAMCOG of 9.42 points
ARC in AD ranging from 1.8 to 4.2 points. Of these studies, (± 9.52). However, as reflected by the large standard devia-
the one with the lowest ARC entered patients at a very mild tion, the ARC was normally distributed with a high degree
stage of AD (Becker et al., 1988). Studies with the highest of individual variability. Additionally, the rate of decline in
ARC had entered severely impaired subjects (Yesavage et al., the first year did not predict subsequent ARC.
1988; Burns et al., 1991), while the remaining studies showed The ADAS includes a cognitive portion, with a maximum
an intermediate ARC (about 2.5 points per year) in patients of 70 points and a non-cognitive subscale scored out of 40.
with an intermediate initial level of impairment (Salmon et Kramer-Ginsberg et al. (1988; R.C. Mohs, personal commu-
al., 1990; Teri et al., 1990; Corey-Bloom et al., 1993b; Aguero- nication) found an average ARC on the ADAS of 9.3 points
Torres et al., 1998; Swanwick et al., 1998; Thal et al., 2000; in 60 AD patients. Yesavage et al. (1988) reported a similar
Winblad et al., 2001). This illustrates that rate of change of ARC on the ADAS of 8.3 points in 30 patients with AD and
the MMSE is affected by the severity of cognitive impairment additionally noted a high correlation between selected sub-
even before the test reaches a floor. The MMSE tests a wide scales of the ADAS and the MMSE. In a larger group of 102
range of cognitive skills and individuals may be expected to AD patients participating in the acetyl-l-carnitine (ALCAR)
decline at varying rates and over different cognitive tasks dur- clinical trial, Thal et al. (2000) reported an ARC of 7.5 points.
ing the progression of AD. For example, patients lose points The Alzheimer’s Disease Assessment Scale-Cognitive sub-
on the orientation, recall and construction items before lan- scale (ADAS-cog) is currently the most common cognitive
guage or praxis becomes abnormal. The degree of variability outcome measurement used in randomized clinical treat-
of ARC for the BIMC (reflected by the standard deviations in ment trials for AD (Wolfson et al., 2002; Jones et al., 2009); a
ARC studies) is significantly smaller than that of the MMSE recent meta-analysis model representing data from approxi-
(Schneider, 1992), suggesting that the BIMC has less noise mately 19,972 AD patients in 52 clinical trials reported a rate
controls and may be more reliable as a change measure. of change of 5.5 points per year (Ito et al., 2010). The rates of
Few studies of longitudinal cognitive change in AD have decline in clinical trials may be less than those seen in natural
used extended follow-up periods. In a study distinguished by history studies because of inherent selection biases. Clinical
7 years of follow-up, Aguero-Torres et al. (1998) found a mean trial volunteers, who tend to have better overall healthcare,
ARC of 2.8 points on the MMSE during their first period are selected to reduce co-morbidities and typically have
of evaluation (entry to 3 years) and 3.0 points thereafter more involved caregivers than the general population.
(3–7 years). Salmon et al. (1990) compared the BIMC, MMSE Unfortunately, while mean ARC for each of these cogni-
and DRS in 55 community-dwelling patients with AD over tive scales was reasonably consistent between studies, the
2 years. Each subject’s scores on the three tests were highly standard deviations were fairly large, roughly equal to the
correlated when entered and after 1 and 2 years later. During means, indicating substantial individual variability. Some
the study period, patients lost a mean of 3.2 points on the patients’ scores did not decline and even improved over 1 to
BIMC, 2.8 points on the MMSE and 11.4 points on the DRS 2 years of follow-up. This variability has several possible
per year. In general, the group declined to a greater degree explanations. First, AD may not progress uniformly, since
on all three scales in the second year than in the first, but patients with early dementia may enter a ‘plateau’ phase with
this was statistically significant only for the DRS. For indi- relatively slow deterioration. Second, performance on these
viduals, however, the ARC of mental status scores in the first cognitive tests probably does not decline in a linear fashion.
year did not predict the next year’s rate of decline. Unlike the Third, test–retest variability is influenced by patients’ mood,
BIMC and the MMSE, the ARC of the DRS did not decrease attention and motivation and many other factors that have
with advanced dementia, indicating that the DRS has a wider an impact on day-to-day mental performance, adding a fur-
range for detecting change than these other tests. ther element of uncertainty to ARC calculations. A fourth
Two somewhat longer instruments, similar to the DRS, possibility is that biological and clinical factors may modify
have been used for serial assessment: the CAMCOG and the rate of progression of AD. These include demographic
the ADAS. The CAMCOG is the cognitive component of factors such as age of onset of the disease, gender and edu-
the Cambridge Mental Disorders of the Elderly Examination cation; co-morbid disease such as cerebral vascular disease,
(CAMDEX), an instrument designed for assessing demen- diabetes and hypertension; historical features such as lipid-
tia. It has sections testing memory, language, praxis, orienta- lowering agents, vitamin and non-steroidal anti-inflam-
tion, attention, calculation, abstraction and perception, with matory drug use; genetic factors such as apolipoprotein E
a maximum total score of 107. In a survey of 110 patients, status and clinical features such as psychosis and EPS.
spanning a wide spectrum of severity of AD, Burns et al. Perhaps the most widely used cognitive screening instru-
found a significant decline over 1 year in the overall score ment is the Montreal Cognitive Assessment (MOCA)
(12.3 points) and in virtually every abstraction were close to (Nasreddine et al., 2005). The MOCA is superior to the MMSE
458 Dementia

in its sensitivity and specificity for MCI and AD (Costa et al., hypothesis suggests that higher education delays the onset
2014; Dong et al., 2012). Longitudinal studies of the MOCA of ­accelerated decline but that, once it begins, it is more
have demonstrated its clinical utility in screening and follow rapid in persons with higher education. This is supported
up of patients with MCI and AD dementia (Gluhm et al., 2013). by Hall et al. (2007) who examined the Buschke Selective
Reminding Test (SRT) in the Bronx Aging Study and fur-
ther supported by Musicco et al. (2009), who reported faster
cognitive progression in more educated mild-to-moderate
45.4 CLINICAL PREDICTORS OF RATE AD subjects with a mean education of 8 (±4.4) years.
OF PROGRESSION The apolipoprotein E (APOE) ε4 allele is associated with
both a high likelihood of developing AD and an earlier age
As noted earlier, a number of factors have been identified of onset; nonetheless, there have been conflicting results with
which appear to affect the rate of decline in AD; however, regard to the influence of APOE genotype on the course of cog-
this is a thorny issue since variables thought to have inde- nitive decline in AD. Gomez-Isla et al. (1996), in a prospective
pendent prognostic significance may simply be markers of longitudinal study of 359 AD patients, found that the APOE ε4
the level of disease severity (Drachman et al., 1990). allele was not associated with any change in rate of progression
For example, using a large well-characterized sample of of dementia. Likewise, several others have reported that rate of
CERAD subjects, Morris et al. (1993) found that dementia decline in AD did not vary significantly across APOE geno-
progression may be non-linear, that rate-of-change determi- type on various cognitive assessments (Growdon et al., 1996;
nations were less reliable when the observation period was Kurz et al., 1996). Furthermore, Frisoni et al. (1995) reported
1 year or less and that rate of progression in AD was deter- a slower rate of progression with increasing ε4 gene dose in a
mined by the severity of cognitive impairment: the less severe retrospective examination of 62 sporadic AD patients. These
the dementia, the slower the rate of decline. Similarly, Teri latter findings were similar to those of Stern et al. who noted a
et al. (1995), using a community-based sample of 156 patients slower rate of decline on the modified MMSE among patients
diagnosed with probable AD followed for up to 5 years, found with ApoE ε4 alleles compared to patients with other geno-
that average rate of decline in cognitive function, measured types (Stern et al., 1997). This is not supported, however, by
by the MMSE and Mattis DRS, became more rapid as the dis- Craft et al. (1998) who reported an accelerated rate of decline
ease progressed. This was confirmed by Storandt et al. (2002) on the DRS in APOE ε4 homozygotes with probable AD and
who, in a large cohort of mildly demented subjects, found Hirono et al. (2003), who reported a similar accelerated rate of
that initial dementia severity was the most significant predic- decline on the ADAS-cog, even after using controlling appara-
tor of annual rate of progression on composite psychometric tus for age, gender, education, test interval and baseline scores.
testing, with increasingly steeper slopes as severity increased. The presence of at least one ε4 allele was associated with faster
In addition, these authors found that the rates of decline were cognitive decline in an incident population-based AD group;
highly variable from person to person and between groups also in two prevalent AD samples when adjusted for disease
as well. Most recently, rate of AD progression on the ADAS- severity (Cosentino et al., 2008). In a recent examination of
cog has been shown to increase with baseline severity in data this subject, using the Religious Orders Study (ROS) and the
from 817 patients in the Alzheimer’s Disease Neuroimaging Memory and Aging Project (MAP), Yu et al. (2013) found
Initiative (ADNI) database (Ito et al., 2011). that presence of the ε4 allele, working primarily through AD
Several studies investigating age at onset as a predic- pathology, was associated with an earlier onset of terminal
tor of deterioration, have reported a significant association decline and faster rates of cognitive decline before and after
between earlier age at onset and greater rate of cognitive or its onset.
functional worsening (Lucca et al., 1993; Jacobs et al., 1994; Concomitant cerebrovascular disease has not been
Teri et al., 1995; Mungas et al., 2001; Backman et al., 2003). shown to increase rate of cognitive decline in AD signifi-
This is not supported by two large prospective studies of cantly (Lee et al., 2000; Storandt et al., 2002; Bhargava et
AD subjects attending memory clinics (Bowler et al., 1998; al., 2006; Lo et al., 2012). However, one longitudinal study
Swanwick et al., 1998). with autopsy follow-up of 50 patients with AD and strokes,
Gender was not seen as an influence on cognitive decline compared to 218 AD patients without strokes, showed a
in several studies of AD (Reisberg et al., 1986; Drachman slight increase in the rate of decline on MMSE scores in the
et al., 1990; Burns et al., 1991; Mortimer et al., 1992; Jacobs co-morbid stroke group for subjects over the age of eighty
et al., 1994; Swanwick et al., 1998; Storandt et al., 2002). (Mungas et al., 2001). In addition, several studies have now
However, this was not the case for others who reported reported that vascular factors, such as myocardial infarc-
slower (Heston et al., 1981; Lucca et al., 1993; Aguero-Torres tion, carotid atherosclerosis and hypertension affect the rate
et al., 1998) and faster (Swanwick et al., 1998) rates of pro- of dementia progression after a diagnosis of AD (Mielke et
gression in men as compared to women. al., 2007; Helzner et al., 2009).
Level of education has also been examined as a predic- Although it has been suggested that patients who develop
tor of decline in several studies (Filley et al., 1985; Berg EPS such as tremor, bradykinesia (slowness of movement),
et al., 1988; Katzman et al., 1988; Drachman et al., 1990; rigid tone, or masked facies have a faster rate of decline, many
Teri et al., 1995; Storandt et al., 2002). The cognitive reserve of these studies may have included patients on neuroleptics
The natural history of Alzheimer’s disease 459

and those with LBV and diffuse Lewy body disease (DLBD) Whether vitamin use, oestrogen, lipid-lowering agents,
(Miller et al., 1991; Mortimer et al., 1992; Chui et al., 1994; or non-steroidal anti-inflammatory drugs (NSAIDs) affect
Stern et al., 1994; Portet et al., 2009). Furthermore, it is now progression of AD, it has not been investigated. High dose
clear that there is a pathological subset of AD patients with vitamin E appeared to slow disease progression in a dou-
alpha-synuclein positive cortical Lewy Bodies in addition to ble-blind, placebo-controlled trial in patients with AD of
typical AD pathology (Fleisher and Olichney, 2005; Weisman moderate severity (Sano et al., 1997). Reduction of serum
et al., 2007). A retrospective study by Lopez et al. (2000) homocysteine with folate, B6 and B12 has shown no benefit
compared clinical characteristics of 185 autopsy-proven AD in slowing AD progression (Aisen et al., 2008). Likewise,
and 60 autopsy-proven AD plus dementia with Lewy bodies a recent meta-analysis of the effect of vitamin B supple-
(DLB) subjects. These authors found no differences in rate of mentation failed to show any significant cognitive (ADAS-
cognitive decline between the two, although DLB subjects cog, MMSE), functional, behavioural or global benefit in
developed EPS earlier. A recent meta-analysis of 18 stud- patients with AD (Li et al., 2014). An omega-3 fatty acid,
ies, using the MMSE, also did not support the hypothesis docosahexaenoic acid, was recently proved in a large pla-
of a faster rate of cognitive decline in DLB compared to AD cebo controlled, randomized clinical trial to not be effec-
(Breitve et al., 2014). However, a study of 56 patients with DLB tive in slowing cognitive decline in AD (Quinn et al., 2009).
compared to 111 patients with AD, who were examined for up Although, several observational studies have suggested that
to 5 years on end points of hospitalization, institutionaliza- oestrogen replacement therapy may have a beneficial effect
tion and death (Hanyu et al., 2009), found that DLB patients on cognitive performance in women with AD, many have
had a significantly shorter time to clinical end points than substantial methodologic problems (Haskell et al., 1997;
those with AD (median time; 40 months versus 52 months, Yaffe et al., 1998). Furthermore, a study of the association
p < .0001). The proportion of hospitalizations (and death) was between serum oestradiol and oestrone levels in 120 hys-
significantly higher in DLB than in AD patients (30% versus terectomized women undergoing replacement found no
14%, p < .05), but differences with regard to nursing home significant association between hormone levels and cogni-
placement did not reach statistical significance (25% versus tive functioning after either 2 or 12 months of treatment
17%). Rates of decline on the MMSE were equivalent for both (Thal et al., 2003). Additional evidence that oestrogen may
groups. Although, there is evidence that the addition of Lewy not be beneficial in AD comes from the Women’s Health
bodies to AD pathology expedites clinical decline, it is not Initiative Memory Study (WHIMS), a randomized, double-
clear that the EPS of typical AD, free from Lewy Body pathol- blind, placebo-controlled clinical trial of 4532 postmeno-
ogy, predicts rapid decline. pausal women, comparing oestrogen and progesterone
Whether neuropsychiatric symptoms superimposed on versus placebo effects on incidence of dementia over 4 years.
AD influence cognitive deterioration or not, has been exam- The authors found that oestrogen plus progestin therapy
ined by several authors. Initial reports suggested that AD increased the risk of probable dementia in postmenopausal
patients reached end points on a modified MMSE or BDS women aged 65 years or above (Shumaker et al., 2003).
score more rapidly when psychotic symptoms were present Several large cohort analyses have demonstrated decreased
(Mayeux et al., 1985; Drevets and Rubin, 1989; Stern et al., prevalence of AD with use of various lipid-lowering agents
1990). However, these investigations did not match patients (Rockwood et al., 2002). A large multicentre placebo con-
according to their level of dementia. Controlling for severity trolled trial investigating the effect of simvastatin on pro-
of AD, other studies were unable to replicate these findings gression of cognitive decline in AD failed to meet its end
(Reisberg et al., 1986; Chen et al., 1991), although two more points (Sano et al., 2011). Analysis of longitudinal changes
have confirmed psychosis as a robust predictor of cogni- over 1 year revealed fewer declines among NSAID patients
tive decline (Chui et al., 1994; Stern et al., 1994). Lopez et than among non-NSAID patients on measures of verbal flu-
al. (1999) examined the effect of psychiatric symptoms and ency, spatial recognition and orientation, in a study consist-
medications on progression in 179 mild-to-moderate prob- ing of 210 patients with AD (Rich et al., 1995). On the other
able AD patients, followed for a mean of 49.5 ± 27.4 months. hand, a randomized placebo-controlled trial of naproxen
These authors found that the presence of psychiatric symp- and rofecoxib showed no effect on cognitive decline in
toms and the use of psychiatric medications were associated 351 patients with mild-to-moderate AD (Aisen et al., 2003).
with increased rate of functional (BDRS) but not cognitive
(MMSE) decline. One cross-sectional study reported that
AD patients with depression had more ADL impairment
45.5 PREDICTING NURSING HOME
than those without (Pearson et al., 1989), however, other
longitudinal studies found no effect of depression on the
PLACEMENT IN AD
rate of decline (Lopez et al., 1990; Storandt et al., 2002).
Recent data from the International Citicoline Trial on Acute Most patients with AD reside in nursing homes or related
Stroke (ICTUS) study demonstrated that the rate of cogni- institutions late in the course of the illness. The interval
tive decline in 1375 subjects with AD was not substantially between the diagnosis of AD and the need for institutional
influenced by the presence of specific neuropsychiatric phe- placement has practical and clinical importance, but is not
notypes (Canevelli et al., 2013). easy to predict. Behavioural symptoms such as agitation,
460 Dementia

wandering or aggression may pre-empt nursing home In a 5-year study of 92 patients who developed AD before
placement, regardless of the level of cognitive impairment. the age of 70, lower mental status scores or aphasia were
Other variables that greatly influence the decision for insti- more common in those who were institutionalized (Hutton
tutional placement of a demented patient may include social et al., 1985; Heyman et al., 1987). In most instances, nursing
support and caregiver burden. Many studies have evaluated home placement occurred because the patients had become
such predictors of nursing home placement. In a system- almost completely helpless and required 24-hour supervi-
atic review, Gaugler et al., (2009) examined 782 studies on sion; for 10 patients the decision was prompted by death or
nursing home placement of dementia patients, performing serious illness of their caregivers.
analysis on the 80 most relevant and high quality studies. A prospective study of 101 initially community-dwelling
They found that the most consistent predictors of nursing AD patients found that 12% with ‘mild AD’ were in nursing
home admission in dementia patients included severity of homes after 1 year and 35% after 2 years (Knopman et al.,
cognitive impairment, basic activity of daily living depen- 1988). For ‘advanced AD’, the figures were 39% at 1 year and
dencies, behavioural symptoms and depression. Caregivers 62% at 2 years. Total activity of daily living (ADL) scores
who reported greater emotional stress also increased risk were significantly related to institutionalization, while
of placement in the nursing home. Demographic variables, incontinence, irritability, inability to walk, wandering,
incontinence and use of available services did not predict hyperactivity and nocturnal behavioural problems were the
nursing home placement. leading reasons for placement cited by caregivers.
A large population based historical cohort study sug- A CERAD analysis, comprising 20 university medical
gests, unsurprisingly, that the mere presence of dementia centres, found that the median time from enrolment as a
increases the overall risk of nursing home admission (Eaker patient to nursing home placement was 3.1 years, with sig-
et al., 2002). In this study, the adjusted hazard ratio for insti- nificantly reduced times (2.1 years) for unmarried males
tutionalization, compared to non-demented controls, was (Heyman et al., 1997). The major predictors of time to
5.44 (95% CI = 3.68–8.05) for AD cases and 5.08 (95% CI = admission were measures of dementia severity upon entry
3.38–7.63) for non-AD dementia cases, independent of co- into the study, including the degree of cognitive impair-
morbid conditions. ment, functional disability and overall stage progression in
As noted, several studies have examined predictors of the disease. Age at entry and marital status (men only) also
institutionalization in AD. And, as expected, the predic- predicted time of nursing home admission. Similar findings
tive factors are complex and vary greatly among studies. with regard to marital status (Haupt and Kurz, 1993) and
One investigation of 14 married men with AD found that rate of functional decline (Wattmo et al., 2011) have been
urinary or faecal incontinence, inability to speak coher- reported by others (Figure 45.1).
ently and loss of bathing and grooming skills were major Finally, a large multiregional, prospective study
determinants of institutionalization (Hutton et al., 1985). ­distinguished by an extremely large number of patients

1.0
CDR = 0

0.8
Cumulative survival (%)

0.6 CDR = 0.5


CDR = 1 Incipient

0.4

0.2

0.0
CDR = 0.5
DAT

0 2 4 6 8 10 12 14 16 18 20 22
Years

Figure 45.1 Progression to nursing home placement in non-demented (Clinical Dementia Rating [CDR] of 0) and demented
(CDR 0.5 incipient, 0.5 dementia of the Alzheimer type and CDR 1) groups. (From Storandt, M. et al., Neurology, 59,
1034–1041, 2002.)
The natural history of Alzheimer’s disease 461

(n = 3944), found that specific caregiver traits (including Bowen et al., 1996; Heyman et al., 1996; Reisberg et al., 1996;
age ≥ 80 years, low income, poor health and high perceived Moritz et al., 1997; Claus et al., 1998; Gambassi et al., 1999;
burden as a result of caregiving) predicted earlier placement Lapane et al., 2001; Ueki et al., 2001), although this has not
for care recipients (Gaugler et al., 2003). been the case for all studies (Hier et al., 1989; Walsh et al.,
1990; Becker et al., 1994). Furthermore, it has been sug-
gested that predictors may differ according to gender. For
example, Moritz et al. (1997) found that, among men, but
45.6 SURVIVAL IN AD not women, survival times were negatively associated with
selected neurological symptoms, particularly, aphasia and
Length of survival in AD is highly variable, although an apraxia. Among women, but not men, a history of cardio-
excess mortality has consistently been reported (Katzman, vascular conditions was associated with poorer survival.
1976; Jorm et al., 1987; Aevarsson et al., 1998). Although, Lapane et al. (2001), on the other hand, found that the most
mean survival after symptom onset may range from 2 to 16 important predictors of mortality for men were severity of
years, the observed survival rate among AD population is dementia and the occurrence of delirium. For women, death
significantly less than the expected rate, based on life expec- was associated with impairment of ADLs, presence of pres-
tancy tables (Walsh et al., 1990). Depending on the study, sure sores, malnutrition and co-morbidity.
survival at 5 years ranges from 10% to 40% in different AD Patients with severe dementia at the beginning of the
populations. The most favourable survival rates are seen in study have shorter survival durations (Walsh et al., 1990;
mild cohorts followed up in outpatient settings, with low- Burns et al., 1991; Evans et al., 1991; Jagger et al., 1995;
est survival rates reported in relatively severely demented Heyman et al., 1996; Moritz et al., 1997; Claus et al., 1998;
subjects from hospital series. The median duration of sur- Ueki et al., 2001; Storandt et al., 2002) (Figure 45.2). In addi-
vival among CERAD patients from the time of study of tion, functional disability decreases survival time in AD
their entry in hospitals was 5.9 years (Heyman et al., 1996), (Martin et al., 1987; Bianchetti et al., 1995; Heyman et al.,
which compares favourably the 5.3 years reported by Walsh 1996; Ueki et al., 2001).
et al. (1990), the 5.7 years noted by Molsa et al. (1986) and Additional variables that may influence survival time
the 5.8 years described by Jost and Grossberg (1995). Three in patients with AD have not been well established, includ-
other studies found shorter median periods of survival of ing level of education (Becker et al., 1994; Stern et al., 1995;
3.4 years (Schoenberg et al., 1987), 3.5 years (Barclay et al., Geerlings et al., 1997), medical co-morbidity (Moritz et al.,
1985) and 4.3 years (Claus et al., 1998), possibly due to inclu- 1997; Aguero-Torres et al., 1998) and psychiatric or behav-
sion of severe subjects. ioural symptoms (Barclay et al., 1985; Molsa et al., 1986;
The shortened survival of AD patients results from com- Walsh et al., 1990; Stern et al., 1994; Samson et al., 1996);
plications due to severe mental decline. Malnutrition, dehy- however, exercise, diet and statin therapy have all been asso-
dration, pneumonia and other infections occur frequently ciated with lower risk of AD mortality in several studies
in the terminal stages when patients are bed-bound, incon- (Scarmeas et al., 2007; Scarmeas et al., 2011; Williams, 2015).
tinent and unable to communicate with others or feed them- Survival in AD appeared to be unrelated to ApoE ε4 gene
selves. Compared to other elderly individuals, AD patients dosage in one study (Corder et al., 1995). However, Tilvis
are not especially predisposed to cancer, cerebrovascular or et al. (1998) noted increased mortality in elderly demented
cardiovascular disease (Molsa et al., 1986). patients with the ε4 allele, a finding supported, at least for
Unremarkably, the most consistent predictors of mor- men, in a study consisting of 92 autopsy-confirmed AD
tality in AD patients are age, gender and dementia sever- cases (Dal Forno et al., 2002). On the other hand, Stern et al.
ity. Age was significantly associated with survival time in a (1997) found that the presence of at least one ε4 allele was
large sample of AD patients, drawn from California (Moritz associated with a less aggressive form of AD and a decreased
et al., 1997). Increasing age has also been related to shorter risk of mortality in 99 patients with probable AD followed
survival in several additional studies (Hier et al., 1989; for up to 6 years.
Walsh et al., 1990; van Dijk et al., 1991; Heyman et al., 1996; EPS appeared to be an important predictor of mortality
Reisberg et al., 1996; Aguero-Torres et al., 1998; Claus et al., in a population-based study of 198 patients with probable
1998; Gambassi et al., 1999; Ueki et al., 2001). Although two early-onset AD (Samson et al., 1996). EPS at entry were also
reports have indicated that survival rates of patients with associated with a higher relative risk of death in 236 patients
early-onset AD are considerably less than those of late-onset with mild AD followed up semi-annually (Stern et al., 1994).
AD (Heyman et al., 1987; McGonigal et al., 1992), this find- A study of 379 AD patients found similar heightened mor-
ing has not been confirmed by others (Walsh et al., 1990; tality due to EPS and demonstrated that individuals with
Bracco et al., 1994; Corder et al., 1995; Bowen et al., 1996; EPS were 3 times more likely to have Lewy bodies (LBs) at
Heyman et al., 1996). autopsy (Haan et al., 2002) (Figure 45.3). Both EPS and LBs
Most investigators have reported shorter survival times were associated with an earlier age of death. This was not
for men than women (Schoenberg et al., 1987; Berg et al., the case for a study by Lopez et al. (2000) who reported no
1988; Burns et al., 1991; van Dijk et al., 1991; Corder et al., differences between AD and AD+LB (LBV) with regard to
1995; Jagger et al., 1995; Molsa et al., 1995; Stern et al., 1995; survival.
462 Dementia

1.0

CDR = 0
0.8

Cumulative survival (%)


0.6
CDR = 0.5
CDR = 1 Incipient
0.4

0.2

0.0
CDR = 0.5
DAT

0 2 4 6 8 10 12 14 16 18 20 22
Years

Figure 45.2 Progression to the end point of death in non-demented (CDR of 0) and demented (CDR 0.5 incipient, 0.5
dementia of the Alzheimer type and CDR 1) groups. (From Storandt, M. et al., Neurology, 59, 1034–1041, 2002.)

1.00

0.90

0.80

0.70

0.60
Neither
Survival function

EPSs
LBs
0.50 Both

0.40

0.30

0.20

0.10

0
52 55 60 62 64 66 68 70 72 74 76 78 80 82 84 86 88 90 92 94 96 102
Age at death, years

Figure 45.3 Survival differences by the presence of extrapyramidal signs (EPSs) and Lewy bodies (LBs) from a life table
analysis. (From Haan, M.N. et al., Archives of Neurology, 59, 588–593, 2002.)
The natural history of Alzheimer’s disease 463

Becker, J.T., Huff, F.J., Nebes, R.D. et al. (1988).


45.7 CONCLUSIONS Neuropsychological function in Alzheimer’s disease.
Pattern of impairment and rates of progression.
Despite an abundance of observations about the natural Archives of Neurology, 45: 263–268.
history of AD, the variability of its clinical features and Berg, L., Miller, J.P., Storandt, M. et al. (1988). Mild senile
rate of progression has not been explained and the valid- dementia of the Alzheimer type: 2. Longitudinal
ity of ‘subtypes’ of AD has not yet been proved. The most assessment. Annals of Neurology, 23: 477–484.
consistent predictor of rapid deterioration and death in AD Bhargava, D., Weiner, M.F., Hynan, L.S. et al. (2006).
patients appears to be the degree of cognitive and functional Vascular disease and risk factors, rate of progression,
disability. Because of the relatively low prevalence of clini- and survival in Alzheimer’s disease. Journal of Geriatric
cal features such as EPS, depression and psychosis at early Psychiatry and Neurology, 19 (2): 78–82.
stages of the disease, larger studies conducted carefully con- Bianchetti, A., Scuratti, A., Zanetti, O. et al. (1995).
trolling the potential confounding effect of dementia sever- Predictors of mortality and institutionalization in
ity are needed to clarify their effect on prognosis. Alzheimer disease patients 1 year after discharge from
an Alzheimer dementia unit. Dementia, 6: 108–112.
Bowen, J.D., Malter, A.D., Sheppard, L. et al. (1996).
REFERENCES
Predictors of mortality in patients diagnosed with
Aevarsson, O., Svanborg, A. and Skoog, I. (1998). Seven- probable Alzheimer’s disease. Neurology, 47: 433–439.
year survival rate after age 85 years: Relation to Bowler, J.V., Munoz, D.G., Merskey, H. and Hachinski,
Alzheimer disease and vascular dementia. Archives of V. (1998). Factors affecting the age of onset and
Neurology, 55: 1226–1232. rate of progression of Alzheimer’s disease. Journal
Aguero-Torres, H., Fratiglioni, L., Guo, Z. et al. (1998). of Neurology, Neurosurgery and Psychiatry, 65 (2):
Prognostic factors in very old demented adults: A 184–190.
seven-year follow-up from a population-based survey Bracco, L., Gallato, R., Grigoletto, F. et al. (1994). Factors
in Stockholm. Journal of American Geriatrics Society, affecting course and survival in Alzheimer’s disease. A
46: 444–452. 9-year longitudinal study. Archives of Neurology, 51:
Aharon-Peretz, J., Israel, O., Goldsher, D. and Peretz, 1213–1219.
A. (1999). Posterior cortical atrophy variants of Breitve, M.H., Chwiszczuk, L.J., Hynninen, M.J. et al.
Alzheimer’s disease. Dementia and Geriatric Cognitive (2014). A systematic review of cognitive decline in
Disorders, 10 (6): 483. dementia with Lewy bodies versus Alzheimer’s dis-
Aisen, P.S., Schafer, K., Grundman, M. et al. (2003). ease. Alzheimer’s Research and Therapy, 6 (5-8): 53.
Results of a multicenter trial of rofexib and naproxen Brouwers, P., Cox, C., Martin, A. et al. (1984). Differential
in Alzheimer’s disease. JAMA: The Journal of the perceptual-spatial impairment in Huntington’s and
American Medical Association, 289: 2819–2826. Alzheimer’s dementias. Archives of Neurology, 41:
Aisen, P.S., Schneider, L.S., Sano, M. et al. (2008). High- 1073–1076.
dose B vitamin supplementation and cognitive decline Burns, A., Jacoby, R. and Levy, R. (1990). Psychiatric phe-
in Alzheimer disease: A randomized controlled nomena in Alzheimer’s disease. IV: Disorders of behav-
trial. JAMA: The Journal of the American Medical iour. The British Journal of Psychiatry, 157: 86–94.
Association, 300: 1774–1783. Burns, A., Jacoby, R. and Levy, R. (1991). Progression of
Backman, L., Jones, S., Small, B.J. et al. (2003). Rate of cognitive impairment in Alzheimer’s disease. Journal of
cognitive decline in preclinical Alzheimer’s disease: American Geriatric Society, 39: 39–45.
The role of comorbidity. The Journals of Gerontology Burns, A., Lewis, G., Jacoby, R. and Levy, R. (1991). Factors
Series B: Psychological Sciences and Social Sciences, affecting survival in Alzheimer’s disease. Psychological
58: 228–236. Medicine, 21: 363–370.
Bakchine, S., Lacomblez, L., Palisson, E. et al. (1989). Canevelli, M., Adali, N., Cantet, C. et al.; ICTUS/DSA
Relationship between primitive reflexes, extra-­ Group. (2013). Impact of behavioral subsyndromes
pyramidal signs, reflective apraxia and severity of on cognitive decline in Alzheimer’s disease: Data
­cognitive impairment in dementia of the Alzheimer from the ICTUS study. Journal of Neurology, 260 (7):
type. Acta Neurologica Scandinavica, 79: 38–46. 1859–1865.
Barclay, L.L., Zemcov, A., Blass, J.P. and McDowell, F.H. Canning, S.J., Leach, L., Stuss, D. et al. (2004).
(1985). Factors associated with duration of survival in Diagnostic utility of abbreviated fluency measures in
Alzheimer’s disease. Biological Psychiatry, 20: 86–93. Alzheimer disease and vascular dementia. Neurology,
Becker, J.T., Boller, F., Lopez, O.L. et al. (1994). The 62 (4): 556.
natural history of Alzheimer’s disease. Description of Carlesimo, G.A. and Oscar-Berman, M. (1992). Memory
study cohort and accuracy of diagnosis. Archives of deficits in Alzheimer’s patients: A comprehensive
Neurology, 51: 585–594. review. Neuropsychology Review, 3 (2): 119–169.
464 Dementia

Chen, J.Y., Stern, Y., Sano, M. and Mayeux, R. (1991). Dong, Y., Lee, W.Y., Basri, N.A. et al. (2012). The Montreal
Cumulative risks of developing extrapyramidal signs, Cognitive Assessment is superior to the Mini-Mental
psychosis, or myoclonus in the course of Alzheimer’s State Examination in detecting patients at higher risk
disease. Archives of Neurology, 48: 1141–1143. of dementia. International Psychogeriatrics, 24 (11):
Chui, H.C., Lyness, S.A., Sobel, E. and Schneider, L.S. 1749–1755.
(1994). Extrapyramidal signs and psychiatric symptoms Drachman, D.A., O’Donnell, B.F., Lew, R.A. and Swearer,
predict faster cognitive decline in Alzheimer’s disease. J.M. (1990). The prognosis in Alzheimer’s disease.
Archives of Neurology, 51: 676–681. ‘How far’ rather than ‘how fast’ best predicts the
Connor, D.J., Salmon, D.P., Sandy, T.J. et al. (1998). course. Archives of Neurology, 47: 851–856.
Cognitive profiles of autopsy-confirmed Lewy body Drevets, W.C. and Rubin, E.H. (1989). Psychotic
variant vs pure Alzheimer disease. Archives of ­symptoms and the longitudinal course of senile
Neurology, 55 (7): 994–1000. dementia of the Alzheimer type. Biological Psychiatry,
Cosentino, S., Scarmeas, N., Helzner, E. et al. (2008). 25: 39–48.
APOE epsilon 4 allele predicts faster cognitive decline Eaker, E.D., Vierkant, R.A. and Mickel, S.F. (2002).
in mild Alzheimer disease. Neurology, 70 (19 Pt 2): Predictors of nursing home admission and/or death in
1842–1849. incident Alzheimer’s disease and other dementia cases
Claus, J.J., van Gool, W.A., Teunisse, S. et al. (1998). compared to controls: A population-based study.
Predicting survival in patients with early Alzheimer’s Journal of Clinical Epidemiology, 55: 462–468.
disease. Dementia and Geriatric Cognitive Disorders, Evans, D.A., Smith, L.A., Scherr, P.A. et al. (1991). Risk
9: 284–293. of death from Alzheimer’s disease in a community
Cook, S.E., Miyahara, S., Bacanu, S.A. et al. (2003). population of older persons. American Journal of
Psychotic symptoms in Alzheimer disease: Evidence Epidemiology, 134: 403–412.
for subtypes. American Journal of Geriatric Psychiatry, Filley, C.M., Brownell, H.H. and Albert, M.L. (1985).
11: 406–413. Education provides no protection against Alzheimer’s
Corder, E.H., Saunders, A.M., Strittmatter, W.J. et al. disease. Neurology, 35: 1781–1784.
(1995). Apolipoprotein E, survival in Alzheimer’s dis- Fleisher, A.S. and Olichney, J.M. (2005).
ease patients, and the competing risks of death and Neurodegenerative disorders with diffuse cortical
Alzheimer’s disease. Neurology, 45: 1323–1328. Lewy bodies. Advances in Neurology, 96: 148–165.
Corey-Bloom, J., Galasko, D., Hofstetter, C.R. et al. Frisoni, G.B., Govoni, S., Geroldi, C. et al. (1995). Gene
(1993a). Clinical features distinguishing large cohorts dose of the epsilon 4 allele of apolipoprotein E and
with possible AD, probable AD, and mixed dementia. disease progression in sporadic late-onset Alzheimer’s
Journal of American Geriatric Society, 41: 31–37. disease. Annals of Neurology, 37: 596–604.
Corey-Bloom, J., Galasko, D., Hofstetter, C.R. et al. Galasko, D., Kwo-on-Yuen, P.F., Klauber, M.R. and Thal,
(1993b). Clinical features distinguishing large cohorts L.J. (1990). Neurological findings in Alzheimer’s
with possible AD, probable AD, and mixed dementia. disease and normal aging. Archives of Neurology, 47:
Journal of American Geriatric Society, 41: 31–37. 625–627.
Costa, A.S., Reich, A., Fimm, B. et al. (2014). Evidence of Gambassi, G., Landi, F., Lapane, K.L. et al. (1999).
the sensitivity of the MoCA alternate forms in moni- Predictors of mortality in patients with Alzheimer’s
toring cognitive change in early Alzheimer’s disease. disease living in nursing homes. Journal of Neurology,
Dementia and Geriatric Cognitive Disorders, 37 (1-2): Neurosurgery and Psychiatry, 67: 59–65.
95–103. Gaugler, J.E., Kane, R.L., Kane, R.A. et al. (2003).
Craft, S., Teri, L., Edland, S.D. et al. (1998). Accelerated Caregiving and institutionalization of cognitively
decline in apolipoprotein E-epsilon4 homozygotes impaired older people: Utilizing dynamic predictors of
with Alzheimer’s disease. Neurology, 51: 149–153. change. Gerontologist, 43: 219–229.
Cummings, J.L., Benson, F., Hill, M.A. and Read, S. (1985). Gaugler, J.E., Yu, F., Krichbaum, K. and Wyman, J.F.
Aphasia in dementia of the Alzheimer type. Neurology, (2009). Predictors of nursing home admission for
35: 394–397. ­persons with dementia. Medical Care, 47: 191–198.
Cummings, J.L. and Cole, G. (2002). Alzheimer dis- Gluhm, S., Goldstein, J., Loc, K. et al. (2013). Cognitive
ease. JAMA: The Journal of the American Medical performance on the mini-mental state examina-
Association, 287: 2335–2338. tion and the montreal cognitive assessment across
Cummings, J.L., Miller, B., Hill, M.A. and Neshkes, R. the healthy adult lifespan. Cognitive and Behavioral
(1987). Neuropsychiatric aspects of multi-infarct Neurology: Official Journal of the Society for
dementia and dementia of the Alzheimer type. Behavioral and Cognitive Neurology, 26 (1): 1–5.
Archives of Neurology, 44: 389–393. Geerlings, M.I., Deeg, D.J., Schmand, B. et al. (1997).
Dal Forno, G., Carson, K.A., Brookmeyer, R. et al. (2002). Increased risk of mortality in Alzheimer’s disease
APOE genotype and survival in men and women with patients with higher education? A replication study.
Alzheimer’s disease. Neurology, 58: 1045–1050. Neurology, 49: 798–802.
The natural history of Alzheimer’s disease 465

Gomez-Isla, T., West, H.L., Rebeck, G.W. et al. (1996). Heyman, A., Peterson, B., Fillenbaum, G. and Pieper, C.
Clinical and pathological correlates of apolipopro- (1997). Predictors of time to institutionalization of
tein E epsilon 4 in Alzheimer’s disease. Annals of patients with Alzheimer’s disease: The CERAD experi-
Neurology, 39: 62–70. ence, part XVII. Neurology, 48 (5): 1304–1309.
Growdon, J.H., Locascio, J.J., Corkin, S. et al. (1996). Heyman, A., Wilkinson, W.E., Hurwitz, B.J. et al. (1987).
Apolipoprotein E genotype does not influence rates Early-onset Alzheimer’s disease: Clinical predic-
of cognitive decline in Alzheimer’s disease. Neurology, tors of institutionalization and death. Neurology, 37:
47: 444–448. 980–984.
Haan, M.N., Jagust, W.J., Galasko, D. and Kaye, J. (2002). Hier, D.B., Warach, J.D., Gorelick, P.B. and Thomas, J.
Effect of extrapyramidal signs and Lewy bodies on (1989). Predictors of survival in clinically diagnosed
survival in patients with Alzheimer disease. Archives of Alzheimer’s disease and multi-infarct dementia.
Neurology, 59: 588–593. Archives of Neurology, 46: 1213–1216.
Hall, C.B., Derby, C., LeValley, A. et al. (2007). Education Hirono, N., Hashimoto, M., Yasuda, M. et al. (2003).
delays accelerated decline on a memory test in Accelerated memory decline in Alzheimer’s disease
persons who develop dementia. Neurology, 69 (17): with apolipoprotein epsilon4 allele. The Journal of
1657–1664. Neuropsychiatry and Clinical Neurosciences, 15:
Hansen, L.A. (1997). The Lewy body variant of Alzheimer 354–358.
disease. Journal of Neural Transmission Supplementa, Hooker, K., Bowman, S.R., Coehlo, D.P. et al. (2002).
51: 83–93. Behavioral change in persons with dementia:
Hanyu, H., Sato, T., Hirao, K. et al. (2009). Differences in Relationships with mental and physical health of
clinical course between dementia with Lewy bod- caregivers. The Journals of Gerontology Series B:
ies and Alzheimer’s disease. European Journal of Psychological Sciences and Social Sciences, 57:
Neurology, 16: 212–217. 453–460.
Haskell, S.G., Richardson, E.D. and Horwitz, R.I. (1997). Hughes, C.P., Berg, L., Danziger, W.L. et al. (1982). A new
The effect of estrogen replacement therapy on clinical scale for the staging of dementia. The British
­cognitive function in women: A critical review of Journal of Psychiatry, 140: 566–572.
the literature. Journal of Clinical Epidemiology, 50: Hutton, J.T., Dippel, R.L., Loewenson, R.B. et al. (1985).
1249–1264. Predictors of nursing home placement of patients with
Haupt, M. and Kurz, A. (1993). Predictors of nursing Alzheimer’s disease. Texas Medicine, 81: 40–43.
home placement in patients with Alzheimer’s dis- Irizarry, M.C., Jin, S., He, F. et al. (2012). Incidence of new-
ease. International Journal of Geriatric Psychiatry, 8: onset seizures in mild to moderate Alzheimer disease.
741–746. Archives of Neurology, 69 (3): 368–372.
Hauser, W.A., Morris, M.L., Heston, L.L. and Anderson, Ito, K., Ahadieh, S., Corrigan, B. et al. (2010). Disease pro-
V.E. (1986). Seizures and myoclonus in patients with gression meta-analysis model in Alzheimer’s disease.
Alzheimer’s disease. Neurology, 36: 1226–1230. Alzheimer’s & Dementia: Journal of the Alzheimer’s
Hebert, R., Dubois, M.F., Wolfson, C. et al. (2001). Factors Association, 6 (1): 39–53.
associated with long-term institutionalization of older Ito, K., Corrigan, B., Zhao, Q. et al. (2011). Disease
people with dementia: Data from the Canadian study progression model for cognitive deterioration from
of health and aging. The Journals of Gerontology Alzheimer’s Disease Neuroimaging Initiative database.
Series A: Biological Sciences and Medical Sciences, 56 Alzheimer’s & Dementia: Journal of the Alzheimer’s
(11): M693–M699. Association, 7 (2): 151–160.
Helmes, E. and Ostbye, T. (2002). Beyond memory impair- Jack, C.R., Jr., Knopman, D.S., Jagust, W.J. et al. (2010).
ment: Cognitive changes in Alzheimer’s disease. Hypothetical model of dynamic biomarkers of the
Archives of Clinical Neuropsychology, 17: 179–193. Alzheimer’s pathological cascade. Lancet Neurology, 9
Helzner, E.P., Luchsinger, J.A., Scarmeas, N. et al. (2009). (1): 119–128.
Contribution of vascular risk factors to the progres- Jacobs, D., Sano, M., Marder, K. et al. (1994). Age at onset
sion in Alzheimer disease. Archives of Neurology, 66: of Alzheimer’s disease: Relation to pattern of cogni-
343–348. tive dysfunction and rate of decline. Neurology, 44:
Heston, L.L., Mastri, A.R., Anderson, V.E. and White, J. 1215–1220.
(1981). Dementia of the Alzheimer type. Clinical genet- Jagger, C., Clarke, M. and Stone, A. (1995). Predictors of
ics, natural history, and associated conditions. Archives survival with Alzheimer’s disease: A community-based
of General Psychiatry, 38: 1085–1090. study. Psychological Medicine, 25: 171–177.
Heyman, A., Peterson, B., Fillenbaum, G. and Pieper, Jones, R.W., Schwam, E., Wilkinson, D. et al. (2009).
C. (1996). The consortium to establish a registry for Rates of cognitive change in Alzheimer disease:
Alzheimer’s disease (CERAD). Part XIV: Demographic Observations across a decade of placebo-controlled
and clinical predictors of survival in patients with clinical trials with donepezil. Alzheimer Disease and
Alzheimer’s disease. Neurology, 46: 656–660. Associated Disorders, 23 (4): 357–364.
466 Dementia

Jorm, A.F., Korten, A.E. and Henderson, A.S. (1987). The Lopez, O.L., Boller, F., Becker, J.T. et al. (1990). Alzheimer’s
prevalence of dementia: A quantitative integration disease and depression: Neuropsychological impair-
of the literature. Acta Psychiatrica Scandinavica, 76: ment and progression of the illness. American Journal
465–479. of Psychiatry, 147 (7): 855–860.
Jost, B.C. and Grossberg, G.T. (1995). The natural history Lopez, O.L., Wisniewski, S.R., Becker, J.T. et al. (1999).
of Alzheimer’s disease: A brain bank study. Journal of Psychiatric medication and abnormal behavior as pre-
American Geriatric Society, 43: 1248–1255. dictors of progression in probable Alzheimer disease.
Jost, B.C. and Grossberg, G.T. (1996). The evolution of Archives of Neurology, 56: 1266–1272.
psychiatric symptoms in Alzheimer’s disease: A natural Lopez, O.L., Wisniewski, S., Hamilton, R.L. et al. (2000).
history study. Journal of American Geriatric Society, Predictors of progression in patients with AD and
44: 1078–1081. Lewy bodies. Neurology, 54: 1774–1779.
Katzman, R. (1976). Editorial: The prevalence and malig- Lucca, U., Comelli, M., Tettamanti, M. et al. (1993). Rate
nancy of Alzheimer disease. A major killer. Archives of of progression and prognostic factors in Alzheimer’s
Neurology, 33: 217–218. disease: A prospective study. Journal of American
Katzman, R., Brown, T., Thal, L.J. et al. (1988). Comparison Geriatric Society, 41: 45–49.
of rate of annual change of mental status score in Lyketsos, C.G., Lopez, O., Jones, B. et al. (2002).
four independent studies of patients with Alzheimer’s Prevalence of neuropsychiatric symptoms in demen-
disease. Annals of Neurology, 24: 384–389. tia and mild cognitive impairment: Results from the
Katzman, R., Galasko, D., Saitoh, T. et al. (1995). Genetic cardiovascular health study. JAMA: The Journal of the
evidence that the Lewy body variant is indeed a American Medical Association, 288: 1475–1483.
phenotypic variant of Alzheimer’s disease. Brain and Martin, D.C., Miller, J.K., Kapoor, W. et al. (1987). A con-
Cognition, 28 (3): 259–265. trolled study of survival with dementia. Archives of
Kavcic, V. and Duffy, C.J. (2003). Attentional dynamics and Neurology, 44: 1122–1126.
visual perception: Mechanisms of spatial disorientation Mayeux, R., Stern, Y. and Spanton, S. (1985).
in Alzheimer’s disease. Brain, 126: 1173–1181. Heterogeneity in dementia of the Alzheimer type:
Knopman, D.S., Kitto, J., Deinard, S. and Heiring, J. (1988). Evidence of subgroups. Neurology, 35: 453–461.
Longitudinal study of death and institutionalization in McGonigal, G., McQuade, C.A., Thomas, B.M. and Whalley,
patients with primary degenerative dementia. Journal L.J. (1992). Survival in presenile Alzheimer’s and multi-
of American Geriatric Society, 36: 108–112. infarct dementias. Neuroepidemiology, 11: 121–126.
Kramer-Ginsberg, E., Mohs, R.C., Aryan, M. et al. Mega, M.S., Masterman, D.M., O’Connor, S.M. et al.
(1988). Clinical predictors of course for Alzheimer (1999). The spectrum of behavioral responses to
patients in a longitudinal study: A preliminary report. cholinesterase inhibitor therapy in Alzheimer disease.
Psychopharmacology Bulletin, 24: 458–462. Archives of Neurology, 56: 1388–1393.
Kurz, A., Egensperger, R., Haupt, M. et al. (1996). Mielke, M.M., Rosenberg, P.B., Tschanz, J. et al. (2007).
Apolipoprotein E epsilon 4 allele, cognitive decline, Vascular factors predict rate of progression in
and deterioration of everyday performance in Alzheimer disease. Neurology, 69: 1850–1858.
Alzheimer’s disease. Neurology, 47: 440–443. Miller, T.P., Tinklenberg, J.R., Brooks, J.O. and Yesavage,
Lapane, K.L., Gambassi, G., Landi, F. et al. (2001). Gender J.A. (1991). Cognitive decline in patients with
differences in predictors of mortality in nursing home Alzheimer disease: Differences in patients with and
residents with AD. Neurology, 56: 650–654. without extrapyramidal signs. Alzheimer Disease and
Lazarus, L.W., Newton, N., Cohler, B. et al. (1987). Associated Disorders, 5: 251–256.
Frequency and presentation of depressive symptoms Molsa, P.K., Marttila, R.J. and Rinne, U.K. (1986). Survival
in patients with primary degenerative dementia. and cause of death in Alzheimer’s disease and multi-
American Journal of Psychiatry, 144: 41–45. infarct dementia. Acta Neurologica Scandinavica, 74:
Lee, J.H., Olichney, J.M., Hansen, L.A. et al. (2000). Small 103–107.
concomitant vascular lesions do not influence rates of Molsa, P.K., Marttila, R.J. and Rinne, U.K. (1995). Long-
cognitive decline in patients with Alzheimer disease. term survival and predictors of mortality in Alzheimer’s
Archives of Neurology, 57: 1474–1479. disease and multi-infarct dementia. Acta Neurologica
Li, M.M., Yu, J.T., Wang, H.F. et al. (2014). Efficacy of Scandinavica, 91: 159–164.
vitamins B supplementation on mild cognitive impair- Moritz, D.J., Fox, P.J., Luscombe, F.A. and Kraemer, H.C.
ment and Alzheimer’s disease: A systematic review (1997). Neurological and psychiatric predictors of mor-
and meta-analysis. Current Alzheimer Research, 11 (9): tality in patients with Alzheimer disease in California.
844–852. Archives of Neurology, 54: 878–885.
Lo, R.Y., Jagust, W.J. and Alzheimer’s Disease Morris, J.C. (1997). Clinical dementia rating: A reliable and
Neuroimaging Initiative. (2012). Vascular burden and valid diagnostic and staging measure for dementia of
Alzheimer disease pathologic progression. Neurology, the Alzheimer type. International Psychogeriatrics, 9:
79 (13): 1349–1355. 173–176.
The natural history of Alzheimer’s disease 467

Morris, J.C., Edland, S., Clark, C. et al. (1993). The con- Reisberg, B., Ferris, S.H., de Leon, M.J. and Crook, T.
sortium to establish a registry for Alzheimer’s disease (1982). The Global Deterioration Scale for assessment
(CERAD). Part IV. Rates of cognitive change in the lon- of primary degenerative dementia. American Journal
gitudinal assessment of probable Alzheimer’s disease. of Psychiatry, 139: 1136–1139.
Neurology, 43: 2457–2465. Reisberg, B., Ferris, S.H., de Leon, M.J. et al. (1989). The
Morris, J.C., Ernesto, C., Schafer, K. et al. (1997). Clinical stage specific temporal course of Alzheimer’s disease:
dementia rating training and reliability in multicenter Functional and behavioral concomitants based upon
studies: The Alzheimer’s Disease Cooperative Study cross-sectional and longitudinal observation. Progress
experience. Neurology, 48: 1508–1510. in Clinical Biological Research, 317: 23–41.
Mortimer, J.A., Ebbitt, B., Jun, S.P. and Finch, M.D. Reisberg, B., Ferris, S.H., Franssen, E.H. et al. (1996).
(1992). Predictors of cognitive and functional progres- Mortality and temporal course of probable Alzheimer’s
sion in patients with probable Alzheimer’s disease. disease: A 5-year prospective study. International
Neurology, 42: 1689–1696. Psychogeriatrics, 8: 291–311.
Mungas, D., Reed, B.R., Ellis, W.G. and Jagust, W.J. Reisberg, B., Ferris, S.H., Shulman, E. et al. (1986).
(2001). The effects of age on rate of progression of Longitudinal course of normal aging and progres-
Alzheimer disease and dementia with associated sive dementia of the Alzheimer’s type: A prospective
cerebrovascular disease. Archives of Neurology, 58: study of 106 subjects over a 3.6 year mean interval.
1243–1247. Progress in Neuropsychopharmacology and Biological
Murdoch, B.E., Chenery, H.J., Wilks, V. and Boyle, Psychiatry, 10: 571–578.
R.S. (1987). Language disorders in dementia of the Rich, J.B., Rasmusson, D.X., Folstein, M.F. et al. (1995).
Alzheimer type. Brain and Language, 31: 122–137. Nonsteroidal anti-inflammatory drugs in Alzheimer’s
Musicco, M., Palmer, K., Salamone, G. et al. (2009). disease. Neurology, 45: 51–55.
Predictors of progression of cognitive decline in Rockwood, K., Kirkland, S., Hogan, D.B. et al. (2002). Use
Alzheimer’s disease: The role of vascular and sociode- of lipid-lowering agents, indication bias, and the risk
mographic factors. Journal of Neurology, 256: of dementia in community-dwelling elderly people.
1288–1295. Archives of Neurology, 59: 223–227.
Nasreddine, Z.S., Phillips, N.A., Bédirian, V. et al. (2005). Rosen, W.G., Mohs, R.C. and Davis, K.L. (1984). A new rat-
The Montreal Cognitive Assessment, MoCA: A brief ing scale for Alzheimer’s disease. American Journal of
screening tool for mild cognitive impairment. Journal Psychiatry, 141: 1356–1364.
of American Geriatric Society, 53 (4): 695–699. Rosen, J. and Zubenko, G.S. (1991). Emergence of psy-
Ortof, E. and Crystal, H.A. (1989). Rate of progression chosis and depression in the longitudinal evaluation of
of Alzheimer’s disease. Journal of American Geriatric Alzheimer’s disease. Biological Psychiatry, 29: 224–232.
Society, 37: 511–514. Rossetti, H.C., Munro Cullum, C., Hynan, L.S. and Lacritz,
Paulsen, J.S., Salmon, D.P., Thal, L.J. et al. (2000). L.H. (2010). The CERAD Neuropsychologic Battery
Incidence of and risk factors for hallucinations and Total Score and the progression of Alzheimer disease.
delusions in patients with probable AD. Neurology, 54 Alzheimer Disease and Associated Disorders, 24 (2):
(10): 1965–1971. 138–142.
Pearson, J.L., Teri, L., Reifler, B.V. and Raskind, M.A. Roth, M., Tym, E., Mountjoy, C.Q. et al. (1986). CAMDEX.
(1989). Functional status and cognitive impairment A standardised instrument for the diagnosis of
in Alzheimer’s patients with and without depression. mental disorder in the elderly with special reference
Journal of American Geriatric Society, 37: 1117–1121. to the early detection of dementia. British Journal of
Portet, F., Scarmeas, N., Cosentino, S. et al. (2009). Psychiatry, 149: 698–709.
Extrapyramidal signs before and after diagnosis of Salmon, D.P., Thal, L.J., Butters, N. and Heindel, W.C.
incident Alzheimer disease in a prospective population (1990). Longitudinal evaluation of dementia of the
study. Archives of Neurology, 66 (9): 1120–1126. Alzheimer type: A comparison of 3 standardized men-
Quinn, J.F., Raman, R., Thomas, R.G. et al. (2009). tal status examinations. Neurology, 40: 1225–1230.
A clinical trial of docosohexanoic acid (DHA) for the Samson, W.N., van Duijn, C.M., Hop, W.C. and Hofman,
treatment of Alzheimer’s disease. In Alzheimer’s A. (1996). Clinical features and mortality in patients
Association International Conference on Alzheimer’s with early-onset Alzheimer’s disease. European of
Disease, Vienna, Austria, pp. 01-04-02. Neurology, 36: 103–106.
Rapcsak, S.Z., Croswell, S.C. and Rubens, A.B. (1989). Sano, M., Bell, K.L., Galasko, D. et al. (2011). A random-
Apraxia in Alzheimer’s disease. Neurology, 39: ized, double-blind, placebo-controlled trial of simv-
664–668. astatin to treat Alzheimer disease. Neurology, 77 (6):
Reisberg, B., Borenstein, J., Salob, S.P. et al. (1987). 556–563.
Behavioral symptoms in Alzheimer’s disease: Sano, M., Ernesto, C., Thomas, R.G. et al. (1997). A con-
Phenomenology and treatment. Journal of Clinical trolled trial of selegiline, alpha-tocopherol, or both as
Psychiatry, 48 (Suppl.): 9–15. treatment for Alzheimer’s disease. The Alzheimer’s
468 Dementia

Disease Cooperative Study. New England Journal of Storandt, M., Grant, E.A., Miller, J.P. and Morris, J.C.
Medicine, 336: 1216–1222. (2002). Rates of progression in mild cognitive impair-
Scarmeas, N., Luchsinger, J.A., Brickman, A.M. et al. ment and early Alzheimer’s disease. Neurology, 59 (7):
(2011). Physical activity and Alzheimer disease course. 1034–1041.
American Journal of Geriatric Psychiatry, 19 (5): Sullivan, E.V., Corkin, S. and Growdon, J.H. (1986). Verbal
471–481. and nonverbal short-term memory in patients with
Scarmeas, N., Luchsinger, J.A., Mayeux, R. and Stern, Y. Alzheimer’s disease and in healthy elderly subjects.
(2007). Mediterranean diet and Alzheimer disease Developmental Neuropsychology, 2: 387–400.
mortality. Neurology, 69 (11): 1084–1093. Swanwick, G.R., Coen, R.F., Coakley, D. and Lawlor, B.A.
Schneider, L.S. (1992). Tracking dementia by the IMC and (1998). Assessment of progression and prognosis
the MMSE. Journal of American Geriatric Society, 40: in ‘possible’ and ‘probable’ Alzheimer’s disease.
537–538. International Journal of Geriatric Psychiatry, 13:
Schneider, L.S. and Dagerman, K.S. (2004). Psychosis of 331–335.
Alzheimer’s disease: Clinical characteristics and his- Teri, L., Hughes, J.P. and Larson, E.B. (1990). Cognitive
tory. Journal of Psychiatric Research, 38 (1): 105–111. deterioration in Alzheimer’s disease: Behavioral and
Schoenberg, B.S., Kokmen, E. and Okazaki, H. (1987). health factors. Journal of Gerontology, 45: 58–63.
Alzheimer’s disease and other dementing illnesses in a Teri, L., Larson, E.B. and Reifler, B.V. (1988). Behavioral dis-
defined United States population: Incidence rates and turbance in dementia of the Alzheimer’s type. Journal
clinical features. Annals of Neurology, 22: 724–729. of the American Geriatrics Society, 36: 1–6.
Shumaker, S.A., Legault, C., Rapp, S.R. et al. (2003). Teri, L., McCurry, S.M., Edland, S.D. et al. (1995). Cognitive
Estrogen plus progestin and the incidence of demen- decline in Alzheimer’s disease: A longitudinal investi-
tia and mild cognitive impairment in postmenopausal gation of risk factors for accelerated decline. Journals
women: The Women’s Health Initiative Memory Study: of Gerontology, Series A: Biological Sciences and
A randomized controlled trial. JAMA: The Journal of Medical Sciences, 50 (A): M49–M55.
the American Medical Association, 289: 2651–2662. Thal, L.J., Calvani, M., Amato, A. and Carta, A. (2000).
Snowden, M.B., Atkins, D.C., Steinman, L.E. et al. (2015). A 1-year controlled trial of acetyl-l-carnitine in early-
Longitudinal association of dementia and depres- onset AD. Neurology, 55: 805–810.
sion. American Journal of Geriatric Psychiatry, 23 (9): Thal, L.J., Grundman, M. and Klauber, M.R. (1988).
897–905. Dementia: Characteristics of a referral population and
Starkstein, S.E., Mizrahi, R. and Power, B.D. (2008). factors associated with progression. Neurology, 38:
Depression in Alzheimer’s disease: Phenomenology, 1083–1090.
clinical correlates and treatment. International Review Thal, L.J., Thomas, R.G., Mulnard, R. et al. (2003).
of Psychiatry, 20: 382–388. Estrogen levels do not correlate with improvement in
Steele, C., Rovner, B., Chase, G.A. and Folstein, M. (1990). cognition. Archives of Neurology, 60: 209–212.
Psychiatric symptoms and nursing home placement of Tilvis, R.S., Strandberg, T.E. and Juva, K. (1998).
patients with Alzheimer’s disease. American Journal of Apolipoprotein E phenotypes, dementia and mortal-
Psychiatry, 147 (8): 1049–1051. ity in a prospective population sample. Journal of
Stern, Y., Albert, M., Brandt, J. et al. (1994). Utility of American Geriatric Society, 46: 712–715.
extrapyramidal signs and psychosis as predictors of Tschampa, H.J., Neumann, M., Zerr, I. et al. (2001).
cognitive and functional decline, nursing home admis- Patients with Alzheimer’s disease and dementia with
sion, and death in Alzheimer’s disease: Prospective Lewy bodies mistaken for Creutzfeldt-Jakob disease.
analyses from the Predictors Study. Neurology, 44: Journal of Neurology, Neurosurgery and Psychiatry, 71:
2300–2307. 33–39.
Stern, Y., Brandt, J., Albert, M. et al. (1997). The absence Tschanz, J.T., Corcoran, C.D., Schwartz, S. et al. (2011).
of an apolipoprotein epsilon4 allele is associated with Progression of cognitive, functional, and neuropsychi-
a more aggressive form of Alzheimer’s disease. Annals atric symptom domains in a population cohort with
of Neurology, 41: 615–620. Alzheimer’s dementia: The Cache County Dementia
Stern, Y., Hesdorffer, D., Sano, M. and Mayeux, R. (1990). Progression study. American Journal of Geriatric
Measurement and prediction of functional capacity in Psychiatry, 19 (6): 532–542.
Alzheimer’s disease. Neurology, 40: 8–14. Ueki, A., Shinjo, H., Shimode, H. et al. (2001). Factors
Stern, Y., Mayeux, R., Sano, M. et al. (1987). Predictors of associated with mortality in patients with early-onset
disease course in patients with probable Alzheimer’s Alzheimer’s disease: A five-year longitudinal study.
disease. Neurology, 37: 1649–1653. International Journal of Geriatric Psychiatry, 16:
Stern, Y., Tang, M.X., Denaro, J. and Mayeux, R. (1995). 810–815.
Increased risk of mortality in Alzheimer’s disease van Dijk, P.T., Dippel, D.W. and Habbema, J.D. (1991).
patients with more advanced educational and occupa- Survival of patients with dementia. Journal of
tional attainment. Annals of Neurology, 37: 590–595. American Geriatric Society, 39: 603–610.
The natural history of Alzheimer’s disease 469

Walsh, J.S., Welch, H.G. and Larson, E.B. (1990). Survival Wolfson, C., Oremus, M., Shukla, V. et al. (2002).
of outpatients with Alzheimer-type dementia. Annals Donepezil and rivastigmine in the treatment of
of Internal Medicine, 113: 429–434. Alzheimer’s disease: A best-evidence synthesis of the
Wattmo, C., Wallin, A.K., Londos, E. and Minthon, L. published data on their efficacy and cost-effective-
(2011). Risk factors for nursing home placement in ness. Clinical Therapeutics, 24: 862–886.
Alzheimer’s disease: A longitudinal study of cognition, Wragg, R.E. and Jeste, D.V. (1989). Overview of depres-
ADL, service utilization, and cholinesterase inhibitor sion and psychosis in Alzheimer’s disease. American
treatment. Gerontologist, 51 (1): 17–27. Journal of Psychiatry, 146: 577–587.
Weisman, D., Cho, M., Taylor, C. et al. (2007). In dementia Yaffe, K., Grady, D., Pressman, A. and Cummings, S.
with Lewy bodies, Braak stage determines phenotype, (1998). Serum estrogen levels, cognitive performance,
not Lewy body distribution. Neurology, 69: 356–359. and risk of cognitive decline in older community
Williams, P.T. (2015). Lower risk of Alzheimer’s disease women. Journal of American Geriatric Society, 46:
mortality with exercise, statin, and fruit intake. Journal 816–821.
of Alzheimer’s Disease, 44 (4): 1121–1129. Yesavage, J.A., Poulsen, S.L., Sheikh, J. and Tanke, E.
Williams, M.M., Storandt, M., Roe, C.M. and Morris, J.C. (1988). Rates of change of common measures of
(2013). Progression of Alzheimer’s disease as measured impairment in senile dementia of the Alzheimer’s type.
by Clinical Dementia Rating sum of boxes scores. Psychopharmacology Bulletin, 24: 531–534.
Alzheimer’s & Dementia: Journal of the Alzheimer’s Yu, L., Boyle, P., Schneider, J.A. et al. (2013). APOE
Association, 9 (1 Suppl.): S39–S44. ε4, Alzheimer’s disease pathology, cerebrovascu-
Wilson, R.S., Bacon, L.D., Fox, J.H. and Kaszniak, A.W. lar disease, and cognitive change over the years
(1983). Primary memory and secondary memory in prior to death. Psychology and Aging, 28 (4):
dementia of the Alzheimer type. Journal of Clinical 1015–1023.
Neuropsychology, 5: 337–344. Zola-Morgan, S., Squire, L.R. and Amaral, D.G. (1986).
Winblad, B., Engedal, K., Soininen, H. et al. (2001). A Human amnesia and the medial temporal region:
1-year, randomized, placebo-controlled study of done- Enduring memory impairment following a bilateral
pezil in patients with mild to moderate AD. Neurology, lesion limited to field CA1 of the hippocampus. The
57: 489–495. Journal of Neuroscience, 6 (10): 2950.
46
The neuropathology of Alzheimer’s disease

COLIN L. MASTERS

cells (Blanks et al., 1989) show degenerative changes and


46.1 INTRODUCTION the anterior olfactory nucleus develops NFTs (Esiri and
Wilcock, 1984). Aβ deposits have been reported in the
In 2015, the centenary of the death of Aloysius Alzheimer retinal pigment epithelial layer, detectable with fluorescent
(1864–1915) was celebrated. Although Alzheimer highlighted β-sheet ligands. Abnormalities in the processes of olfactory
the two key histological lesions, amyloid plaques and neurofi- neurones in the nasal mucosa have been reported (Talamo
brillary tangles (NFTs) (Alzheimer, 1907), it is only in the last et al., 1989) and tau-reactive filaments, absent from con-
30 years that developments in molecular biology, molecular trols, have been described in the olfactory mucosa in biopsy
genetics, immunohistochemistry, image analysis and protein specimens of patients with probable AD (Tabaton et al.,
chemistry have transformed our understanding of the patho- 1991a). Deposition of Aβ in the periphery of the lens may
genesis of Alzheimer’s disease (AD). For laboratory-based also be linked to AD (Goldstein et al., 2003), although more
diagnostic pathologists, brain immunohistochemistry and studies are required to confirm this association.
cerebrospinal fluid (CSF) immunoassays in particular have
become an essential part of the diagnostic armoury.

46.3 MACROSCOPIC
NEUROPATHOLOGIC
46.2 PATHOLOGIC CHANGES OUTSIDE CHANGES
THE CENTRAL NERVOUS SYSTEM
(CNS) The naked-eye appearances of the brain range from
unremarkable to grossly abnormal. The leptomeninges
General post-mortem examination does not reveal any may be thickened, particularly over the convexity, and may
specific abnormalities outside the nervous system. Patients show orange-brown areas of various sizes, indicating old,
with AD usually die from terminal respiratory illness, circumscribed subarachnoid haemorrhage, which most
and bronchopneumonia is most often the cause of death. often result from amyloid congophilic angiopathy (ACA).
Ongoing investigations have, however, revealed that subtle The cranial nerves are normal and the large cerebral vessels
biochemical abnormalities may not be restricted to the brain. in uncomplicated cases of AD have not been damaged more
AD may, like a systemic illness, cause secondary changes in by atherosclerosis than expected for the patient’s age. Brain
the CSF and peripheral blood that reflect neuronal metabo- weight is quite often reduced, sometimes below 1000 g. Since
lism of the amyloid β (Aβ) and tau proteins. As the disease brain weight normally depends on the patient’s age, sex and
progresses, the total biochemical pools of Aβ and tau constitution, the degree of atrophy should be considered in
increase within the brain, CSF and blood, even though the the light of these factors. A discrepancy between the findings
Aβ levels in CSF and blood decrease as the disease develops of neuroimaging and neuropathology is not unusual: cere-
(Ritchie et al., 2003; Bates et al., 2009; Craig-Shapiro et al., bral atrophy reported on computed tomography (CT) scans
2009; Fagan et al., 2009). or magnetic resonance imaging (MRI) may not always be
That sensory outposts associated with the brain under- confirmed at post-mortem examination. Agonal changes
going degeneration has been suspected for some time. of haemodynamics, post-mortem delay and the effects
The optic nerve (Hinton et al., 1986) and retinal ganglion of fixation on the brain may be partly responsible for this
470
The neuropathology of Alzheimer’s disease 471

discrepancy. The normal ratio of 8:1 of total brain weight to scale is now able to be visualised by positron emission
that of the brainstem and cerebellum may decrease, indicat- tomography (PET)-Aβ tracers or measured by CSF Aβ levels
ing loss of tissue from the cerebral hemispheres. The atrophy as the disease begins and progresses (Rowe et al., 2007; Jack
is usually diffuse and symmetrical, although the frontopa- et al., 2009; Villemagne et al., 2013) (see Chapter 12). These
rietal region and the temporal lobes may be more severely new techniques and assays may be reporting on different
affected than the rest of the brain (Figure 46.1). Moderate- aggregation states of Aβ (Matveev et al., 2014).
to-severe atrophy is easy to discern: the sulci are widened
and the gyri are narrowed both on the outside and on coro-
nal slices. The latter also reveal enlarged lateral ventricles
with rounded angles and additional space between the hip- 46.4 HISTOLOGY, ULTRASTRUCTURE,
pocampi and the wall of the temporal horns. Decrease in IMMUNOCYTOCHEMISTRY AND
the thickness of neocortical ribbon, apart from extreme MORPHOMETRY
cases of atrophy, is difficult, and may prove deceptive, to
assess on naked-eye appearances. A quantitative study, The neurohistological features of AD are complex and vari-
however, revealed a decrease of neocortical areas of all lobes able. The two hallmark lesions, Aβ amyloid plaques and
in AD, whereas in patients over the age of 80, only the tem- NFTs (Figure 46.2) are complemented by granulovacuolar
poral cortex was atrophied (Hubbard and Anderson, 1981, degeneration, Hirano bodies, neuronal and synaptic loss,
1985). Reduction in cortical area may result from a decrease abnormalities of neuronal processes and synapses, astro-
in the length rather than in the width of cortex, indicating cytic and microglial response, and vascular changes. The
loss of columns of cells and fibres perpendicular to the pial white matter may also be affected (Esiri et al., 1997).
surface. A correlation appears to exist between the length,
but not the thickness, of the cortical ribbon and dementia
46.4.1 AMYLOID PLAQUES
score (Duyckaerts et al., 1985). Apart from occasional small,
orange-coloured old haemorrhages in the cortex, suggestive The amyloid or neuritic plaque (‘senile’ plaque) is one of
of ACA, uncomplicated AD usually is not associated with the major lesions found in the AD brain, first described by
any other focal lesions. There is a growing awareness that the Blocq and Marinesco (1892). These structures, ranging in
topographic dispersion of the lesions of AD are related to an size from 50 to 200 µm (Terry, 1985), can be readily demon-
underlying functional property of the higher nervous sys- strated in frozen and paraffin-embedded sections by silver
tem. The brain’s ‘default network’ may explain why certain impregnation methods (Figure 46.3) and immunocyto-
areas are more affected by atrophy and Aβ amyloid deposi- chemistry. The lesion consists of an Aβ amyloid core with
tion (Buckner et al., 2009; Dickerson et al., 2009). In turn, a corona of argyrophilic axonal and dendritic processes,
the evolution of Aβ amyloid deposition at the macroscopic Aβ amyloid fibrils, glial cell processes and microglial cells.
Neuritic processes in the periphery of the plaque are fre-
quently dystrophic and contain paired helical filaments,
which are composed largely of ubiquinated and phosphory-
lated tau protein (Gonatas et al., 1967; Hanger et al., 1991).
Divry (1927) demonstrated that the core of the AD plaque
contained a congophilic amyloid substance, which gave
an apple green colour under polarised light. The amyloid
plaque is composed of 5–10 nm filaments made up of a 38-
to 43-amino acid (4 kDa) peptide (Masters et al., 1985), now
referred to as Aβ peptide, derived from neurones by proteo-
lytic cleavage of the amyloid precursor protein (APP) (Kang
et al., 1987).
Three types of plaque have been identified in conven-
tional silver staining preparations: ‘primitive’ or ‘early’,
‘classical’ or ‘mature’, and ‘burnt out’ or ‘compact’. On the
basis of light and electron microscopic studies, a three-
stage evolution of the plaque has been proposed. The first
stage is the ‘primitive’ plaque, composed of a small num-
Figure 46.1 On the left, a coronal slice of the brain of a
ber of distorted neurites, largely presynaptic in origin
patient with severe Alzheimer’s disease showing atrophy:
The lateral ventricle is enlarged with rounding of its angle
with few amyloid fibres, astrocytic processes and the occa-
and there is additional space between the hippocampus sional microglial cell. The second stage is the ‘classical’ or
and the inferior horn of the lateral ventricle. Several gyri ‘mature’ plaque with a dense amyloid core with a halo of
are narrowed and the lateral fissure is widened. On the dystrophic neurites, astrocytic processes and cell bodies
right, a slice of the right hemibrain of a normal subject. and the occasional microglial cell. The final stage in this
472 Dementia

Figure 46.2 Neurofibrillary tangles and neurotic amyloid plaques in the neocortex. Modified Bielschowsky silver impregnation.

stages of plaque development have not been confirmed in


experimental transgenic mice and therefore the temporal
relationships suggested above remain speculative: it may yet
transpire that the three types of plaques are independent of
each other.
Studies using antibodies raised against Aβ peptide
reveal a much more widespread deposition of amyloid than
is visualized by traditional staining methods (Majocha
et al., 1988; Gentleman et al., 1989; Armstrong et al., 1996)
(Figures 46.4 through 46.8). Aβ immunoreactivity has been
detected throughout the CNS including the neocortex, hip-
pocampus, thalamus, amygdala, caudate nucleus, putamen,
nucleus basalis of Meynert (nbM), midbrain, pons, medulla
oblongata, the cerebellar cortex and spinal cord (Joachim
et al., 1989; Ogomori et al., 1989). These deposits take a vari-
ety of forms and include subpial, vascular, dyshoric, punc-
tate or granular, diffuse, stellar, ring-with-core and compact
deposits (Ogomori et al., 1989). A laminar pattern of Aβ
deposits in the neocortex has been described with concen-
trations in layers II, III and V (Majocha et al., 1988).
As noted, the Aβ peptide is a 38–43 residue cleavage
product of a larger precursor protein, the APP (Kang et al.,
1987). Although most cells have the potential to produce
APP, neurones and platelets express APP to high levels.
Moreover, neurones process APP with β-secretase (BACE)
Figure 46.3 An argyrophilic amyloid plaque in the to a greater extent than most other cell types, which in turn
neocortex showing a neurotic ring with an amyloid core
leads to the formation of full Aβ sequences. The theory that
formation. Modified Bielschowsky silver impregnation.
Aβ deposition is an early event in AD pathogenesis has been
given support by the presence of extracellular Aβ in diffuse
sequence is the ‘burnt out’ plaque consisting of a dense core plaques not associated with any neuritic change or astro-
of amyloid (Terry et al., 1981; Esiri and Morris, 1997). The cytic involvement (Yamaguchi et al., 1991). More condensed
relationship between dystrophic neurites, neurofibrillary Aβ is, however, associated with a neuritic change, reactive
degeneration and Aβ amyloid deposition is not fully under- astrocytosis and microglial infiltration and phagocytosis.
stood (Adalbert et al., 2009), but the neurites and tangles Most species of Aβ have been detected by immunohisto-
appear to be downstream of Aβ accumulation. These three chemistry using antibodies that recognize epitopes within
The neuropathology of Alzheimer’s disease 473

Figure 46.6 A high-power electron micrograph reveal-


ing (Aβ) fibrils (A) of a plaque. (Courtesy of Dr I. Janota,
Institute of Psychiatry, London.)

Figure 46.4 An amyloid plaque with a dense core and


corona. Amyloid β (Aβ) immunohistochemistry.

(a)

Figure 46.5 Aβ- protein deposits in the temporal lobe of


a patient with Alzheimer’s disease. Several deposit mor-
phologies can be seen. Aβ immunohistochemistry.
(b)
the full-length Aβ(1–42) or a truncated Aβ(1–40). The pre-
dominant species in sporadic AD is Aβ42(43) and this is Figure 46.7 Congophilic angiopathy. Blood vessels
stained with Congo red showing birefringence under
found in plaques of all morphological types. Diffuse plaques
polarised light (a) and β immunohistochemistry (b).
contain mainly Aβ42(43) (Mann et al., 1996a, 1996b).
A number of other proteins co-localize with Aβ includ-
ing apolipoprotein E (ApoE), a widely distributed protein AD. ApoE has been shown to bind to Aβ and a high propor-
involved in the transport of cholesterol. There are three tion of Aβ and ApoE deposits tend to be co-localized. Thus,
common isoforms of ApoE, which are encoded by three both diffuse and compact Aβ plaques may be immunola-
alleles, ε2, ε3 and ε4, and there is a strong association belled by anti-Aβ and anti-ApoE antibodies (Cairns et al.,
between the presence of the ε4 allele and the age of onset of 1997b). The presence of one or more ε4 alleles leads to both
474 Dementia

Figure 46.8 A high-power electron micrograph showing Aβ fibrils (A) surrounding a blood vessel: dysphoric angiopathy.
(Courtesy of Dr I. Janota, Institute of Psychiatry, London.)

an earlier-onset and a more severe Aβ amyloidosis (Roses, to Aβ may be required to cause aggregation of tau. Tangle-
1994; Agosta et al., 2009). The relationship between Aβ and bearing neurones generally project into areas with Aβ depo-
ApoE is still imperfectly understood, as ApoE knockout sition, suggesting some form of retrograde transport of the
mice have an increased amount of Aβ deposition on a trans- toxic elements that are associated with Aβ.
genic APP background.
The plaques are also immunohistochemically positive for 46.4.2 NEUROFIBRILLARY TANGLES
a number of neuroactive substances. Acetylcholinesterase,
a marker of the cholinergic system, can be demonstrated The second histological hallmark of AD is the NFT. These
within and around the neuritic elements of the corona. are not specific to AD, since they occur in other neurode-
Many of these elements derive from cholinergic neurones in generative disorders, including Down’s syndrome, posten-
the basal forebrain, particularly the nbM, the diagonal band cephalitic parkinsonism, dementia pugilistica, amyotrophic
of Broca and the medial septal nuclei. Several other neu- lateral sclerosis–parkinsonism–dementia complex of Guam,
rotransmitters have been demonstrated in the plaque. These subacute sclerosing panencephalitis, dementia with tangles
include substance P, neuropeptide Y, neurotensin, cholecys- with and without calcification, and in myotonic dystrophy
tokinin, 5-hydroxytryptamine (5HT) and catecholamine (Kiuchi et al., 1991). Globose NFTs also develop in progres-
(Walker et al., 1988; Hauw et al., 1991). Ubiquitin, a protein sive supranuclear palsy and sporadic motor neurone dis-
acting as a signal for degradation of abnormal proteins, is ease, but their morphology and ultrastructure differ from
also present in intracellular neurites and NFTs (Perry et al., those seen in AD. Current controversy surrounds the con-
1987). The protease inhibitor α1-antichymotrypsin has cept of primary age-related tauopathy (PART) and whether
been localized to the plaque using immunohistochemical this occurs in the absence of co-morbidity with other dis-
methods (Walker et al., 1988). Serum amyloid P compo- eases such as AD (Crary et al., 2014).
nent (SAP) is a glycoprotein complex produced in the liver, In AD, NFTs are common in the medial temporal struc-
distributed in serum and has been detected in diffuse and tures, in the hippocampus, amygdala and parahippocam-
consolidated Aβ deposits, NFTs and neuritic degeneration. pal gyrus, and also occur throughout the neocortex and the
The plaques are more numerous in associative regions of deep grey matter including the lentiform nucleus, the nbM,
neocortex than in sensory areas and are also largest in those the thalamus, the mamillary bodies, substantia nigra, locus
laminae characterised by large pyramidal neurones (Rogers caeruleus, periaqueductal grey matter, the raphé nuclei of
and Morrison, 1985). Accumulations of Aβ peptide are the brainstem and the pedunculopontine nucleus.
found in the neocortex of non-demented elderly individu- NFTs are intracellular inclusions composed of ubiqui-
als without any neurofibrillary change, suggesting that Aβ nated and phosphorylated tau. Their configuration is
deposition precedes neurofibrillary changes. However, NFT determined in part by the shape of neurones in which they
formation has been reported in areas devoid of Aβ (Braak develop. NFTs are flame shaped in pyramidal neurones,
et al., 1986; Duyckaerts et al., 1988; Delaère et al., 1990; while in the neurones of the brainstem they assume more
Armstrong et al., 1993), indicating that factors in addition complex, globose forms. They can be easily discerned in
The neuropathology of Alzheimer’s disease 475

histological sections stained with haematoxylin and eosin,


silver impregnation techniques or with Congo red, which
renders them birefringent under polarised light. They can
be best demonstrated by immunocytochemistry using anti-
bodies against tau and ubiquitin (Figure 46.9).
Electron microscopy reveals the fine structural details
(Figure 46.10). NFTs are mainly composed of paired heli-
cal filaments (Kidd, 1963), which in turn are formed by two
­fi laments wound around each other. Each filament has a
diameter of 10 nm with crossover points every 80 nm, result-
ing in the typical periodicity of a double helix. A negative
staining technique has demonstrated that each filament is
composed of four protofilaments with a diameter of 3–5 nm
(Wisniewski et al., 1984). NFTs also contain straight fila-
ments with an average diameter of 15 nm. These straight and
the paired helical filaments may form hybrid filaments, dis-
playing both morphologies, share surface epitopes and have
identically shaped structural units. These common features Figure 46.10 A high-power electron micrograph reveal-
indicate that paired helical and straight filaments are related ing paired helical filaments of a neurofibrillary tangle.
(Courtesy of the late Professor L.W. Duchen, Institute of
and they represent only somewhat different assemblies of
Psychiatry, London.)
the same basic molecular unit (Crowther, 1991).
A major component of the NFT is a microtubule-­
associated protein called tau, which is involved in microtu- Biochemically, NFTs are composed of phosphorylated
bule assembly and stabilization. In the adult human brain isoforms of tau (Goedert et al., 1988), and monoclonal anti-
there are six isoforms of tau and all of these are hyper- bodies can be used to quantify NFTs (Harrington et al.,
phosphorylated in AD (Hanger et al., 1991; Goedert et al., 1991). They also contain ubiquitin and anti-ubiquitin anti-
1992). Antibodies that recognize specific phosphorylation-­ bodies giving strongly positive reaction with NFTs (Mori
dependent epitopes may be used to identify the three sites et al., 1987; Perry et al., 1987).
of tau aggregation: the NFT, dystrophic neurites of plaques While NFTs are intracytoplasmic neuronal inclusions,
and neuropil threads. Phosphorylation-dependent antibod- they may become extracellular after the neurone that con-
ies may also be used to distinguish the tauopathy in AD tained them has vanished. These extraneuronal NFTs are
from other neurological disorders (Cairns et al., 1997a). most often seen in the hippocampus and entorhinal cortex
Other neuronal inclusions that have confused neuropa- in advanced disease, and both their antigenic properties
thologists until recently have been elucidated as contain- and ultrastructural features differ from their intraneuronal
ing the TAR DNA-binding (TDP)-43 protein (Arai et al., counterparts (Tabaton et al., 1991b). They are chiefly com-
2009). This type of neuronal inclusion is prominent in the posed of straight, not paired, helical filaments and react dif-
frontotemporal lobar atrophies of the non-Pick type, amyo- ferently with antibodies to tau and ubiquitin. A small subset
trophic lateral sclerosis (ALS), and also in selected areas of of extraneuronal tangles gives positive reactions with anti-
the AD-affected brain. Its significance in AD is the subject bodies to both tau and Aβ protein. The degradation of the
of intense scrutiny. NFT (the transition from intracellular to extracellular form)
is a series of distinct stages involving complex molecular
events that alter both the antigenicity and configuration of
the tangle (Bondareff et al., 1990). It is the insolubility of
NFTs that is most likely to be responsible for the occurrence
of extraneuronal forms (Terry, 1990). The extraneuronal
NFTs may be enveloped by glial processes and eventually be
internalized as part of their degradative cycle.

46.4.3 GRANULOVACUOLAR
DEGENERATION
Granulovacuolar degeneration was first described by
Simchowicz (1911) in the hippocampal neurones of senile
dementia patients (Figure 46.11). Granulovacuoles are
abnormal cytoplasmic structures: one or more vacuoles of
Figure 46.9 A neuritic plaque (P) and neurofibrillary tan- 3.5 mm in diameter each containing a single granule. The
gles (T) demonstrated by anti-tau immunohistochemistry. electron microscope shows a dense granular core, embedded
476 Dementia

to actin (Goldman, 1983) and immunostaining for tau


(Galloway et al., 1987).

46.4.5 NEURONAL LOSS
Neuronal loss is far more difficult to assess than the his-
tological abnormalities previously described, although the
decrease of particular neuronal populations is more readily
accepted than in normal ageing (Terry, 1990). While well-
defined, neuronal groups, like the locus ceruleus, are easier
to count, the cerebral cortex presents considerable technical
problems. Nevertheless, it has been established that there
is a significant decrease of 36%–46% in the concentration
of large neurones, particularly from layers III and V of the
neocortex (Terry et al., 1981). The loss of the large corti-
Figure 46.11 Granulovacuolar degeneration (arrows) and cal neurones is age dependent: the column of these cells is
Hirano bodies (arrowheads) in the pyramidal cell layer of reduced in patients under 80 years of age, but appears to be
the hippocampus. Haematoxylin and eosin.
normal for age in older subjects (Hubbard and Anderson,
1985). This neuronal loss, however, does not affect the
in a translucent matrix. This granular component gives a neocortex evenly; it appears to be restricted to the fron-
positive reaction with antibodies to tubulin (Price et al., tal and temporal lobes, whereas the parietal and occipital
1985), to phosphorylated epitopes of the heavy neurofila- regions are less involved (Mountjoy et al., 1983). The hip-
ment peptide (Kahn et al., 1985), to tau (Dickson et al., 1987) pocampus is even more severely affected: neuronal fall out
and, in some neurones, to ubiquitin (Leigh et al., 1989). reaches an average of 47% (Ball, 1977) and the number of
A detailed immunocytochemical study has demonstrated pyramidal neurones is reduced by 40% in area H1 (Mann
that most epitopes of the tau molecule are sequestered in et al., 1985). This loss appears to correlate with the num-
granulovacuoles and this tau protein is antigenically similar ber of tangle-bearing cells, while the end plate and area H2
to that found in paired helical filaments. Granulovacuoles are less affected. There is also a substantial decrease of neu-
are now considered to be special autophagosomes that are rones in the cholinergic nbM (Whitehouse et al., 1981), in
formed by the sequestration of electron-dense material by the cholinergic pedunculopontine nucleus (Jellinger, 1988),
a two-layered membrane. Granulovacuoles are virtually in the noradrenergic locus ceruleus (Tomlinson et al., 1981;
restricted to the pyramidal neurones of the hippocampus Marcyniuk et al., 1986) and in the serotonergic raphé nuclei
and in severe AD cases, the neuronal cytoplasm is replete in the brainstem (Yamamoto and Hirano, 1985).
with these abnormal structures. They also occur, much less
often, in a variety of diseases, including progressive supra- 46.4.6 ABNORMALITIES OF NEURONAL
nuclear palsy, amyotrophic lateral sclerosis–parkinsonism– PROCESSES AND SYNAPSES
dementia complex of Guam and tuberous sclerosis.
Neuropil threads or ‘curly’ fibres and dystrophic neurites have
46.4.4 HIRANO BODIES been well documented in silver impregnated preparations
developed by Gallyas (1971), but they attracted attention when
These abnormal structures were first described by Hirano it was realized that their occurrence was associated with the
(1965). They are most commonly seen in and among the severity of dementia (Braak et al., 1986). Neuropil threads or
pyramidal cells of the hippocampus. Hirano bodies occur curly fibres are slender, short structures found in the neuropil
in normal subjects and their frequency increases with age. of the cortex. Dystrophic neurites, some of which contribute to
However, AD patients have significantly more Hirano bod- the formation of plaques, also appear as somewhat contorted,
ies than age-matched controls. They also occur in various thread-like structures in these preparations. These abnormal
neurological diseases, including Pick’s disease, motor neu- neurites are likely to be a heterogeneous population of den-
rone disease and kuru, and in animals infected with kuru drites and axons (Tourtellotte and Van Hoesen, 1991). Their
and scrapie (Gibson and Tomlinson, 1977). Hirano bodies ultrastructural and immunocytochemical features are strik-
stand out in sections stained with haematoxylin and eosin ingly similar to those of NFTs, indicating that they also origi-
as bright pink, homogeneous structures that are circular in nate from the altered neuronal cytoskeleton. Neuropil threads
cross section with a diameter up to 25 µm, and rectangu- immunostain with antibodies to tau (Kowall and Kosik, 1987;
lar or spindle shaped up to 30 µm in length in longitudinal Probst et al., 1989) and to ubiquitin (Joachim et al., 1987).
sections. Ultrastructurally, they are composed of a complex, Electron microscopy has revealed that they are often in den-
crystalline array of interlacing filaments forming a lattice- drites and contain straight filaments of 14–16 nm in diameter
like or ‘herringbone’ configuration (Tomonaga, 1974). It in addition to paired helical filaments (Tabaton et al., 1989;
has been reported that they stain positively with antibodies Yamaguchi et al., 1990b). Moreover, they are often in the
The neuropathology of Alzheimer’s disease 477

dendrites of those nerve cells that contain NFTs in their cell 1992), where perivascular gliosis is prominent throughout
body (Braak and Braak, 1988). There is increasing evidence the cerebrum. In AD, extensive gliosis has a laminar pat-
that their distribution is more closely associated with NFTs tern in the neocortex, but there does not appear to be an
than with plaques. Considerable decrease of synapses also AD-specific pattern of subcortical gliosis. In addition to
occurs and this loss is likely to contribute to the development NFTs, plaques and dystrophic neurites, reactive astrocyto-
of the disease (Davis et al., 1987). Moreover, immunocyto- sis is associated with degenerating neurones in AD. In the
chemical quantification of synaptophysin, a protein localized pathogenesis of the plaque Aβ deposition appears to be an
in presynaptic terminals, revealed an average decrease of 50% early event preceding gliosis (Yamaguchi et al., 1990a).
in the density of the granular neuropil immunoreaction in The presence of major histocompatibility complex
the parietal, temporal and midfrontal cortex (Masliah et al., (MHC) class II antigens on reactive microglia in association
1989). In the electron microscope, some presynaptic terminals with plaques in AD is indicative of an immune response,
were distended and contained altered synaptic organelles, although the significance of such expression on the cell
including swollen vesicles and dense bodies, similar to those surfaces of reactive microglial cells is uncertain. Reactive
seen in dystrophic neurites. These latter structures gave posi- microglial cells have been found in association with dif-
tive staining with antibodies to synaptophysin and immu- fuse Aβ deposits and, more frequently, with compact cores
noreactivity was mainly localized to the outer membranes of of plaques (Itagakai et al., 1989). Leucocyte common anti-
synaptic vesicles and dense bodies. This finding lends further gen and complement receptors – including the cell adhe-
support to the view that progressive synaptic derangements sion molecules, the β-2 integrins – have been localised on
occur in the neocortex of AD (Masliah et al., 1991). microglial cells around plaques. These data are consistent
with a phagocytic role for microglia and provide evidence
46.4.7 GLIAL CHANGES for an innate immune response of brain tissue in AD.

Astrocytes can be readily observed immunohistochemi- 46.4.8 VASCULAR ABNORMALITIES


cally by using an antibody to glial fibrillary acidic protein
(GFAP; see Figure 46.12). Both NFTs and plaques are more When stained with the dye Congo red, cerebral blood ves-
prevalent in the pyramidal cell-rich layers II, III and V than sels containing amyloid appear apple green under polarized
in other laminae (Mann et al., 1985; Majocha et al., 1988; light. Pantelakis (1954) called this deposition congophilic
Braak et al., 1989), and both have been associated with pat- angiopathy (see Figure 46.8). These changes affect lepto-
terns of gliosis in AD (Itagakai et al., 1989; Cairns et al., meningeal and cortical arterioles, and occasionally intra-
cortical capillaries and venules. The parieto-occipital cortex
is usually more affected than that in the frontal and tem-
poral lobes and the change is most easily identified in the
striate cortex in the occipital lobe. The brainstem and cer-
ebellar cortex are less frequently affected. The occurrence
of ACA in conjunction with AD is quite common (Keage
et al., 2009).
Vascular amyloid is the same protein as the Aβ amyloid
found in plaques. Although vascular amyloid is frequently
present in AD, it may be absent even when there are abundant
plaques and parenchymal deposits. Occasionally, Aβ appears
to extend from the vessel wall into the surrounding cerebral
tissue; this is referred to as dyshoric angiopathy (or the drusige
Entartung of Scholz). This is commonly seen in lamina III of
the striate cortex and adjacent occipital cortex, occasionally
in Ammon’s horn and in the cerebellum (Hauw et al., 1991).
At least three forms of autosomal dominant familial cere-
bral amyloid angiopathy have been identified at the molecu-
lar level. In the Dutch (Levy et al., 1990) and Icelandic forms,
patients suffer from recurrent cerebral haemorrhages that lead
to an early death (before 40 years of age in Icelandic patients,
and between 50 and 60 years in Dutch patients). The amyloid
fibrils in patients with the Icelandic type are made up of cys-
tatin C, a degradation product of a larger protease inhibitor.
In the Dutch patients the amyloid fibrils are composed of the
Figure 46.12 Astrocyte cell bodies and processes same Aβ protein found in amyloid angiopathy in AD, Down’s
encircling a plaque. Glial fibrillary acidic protein syndrome and sporadic amyloid angiopathy. In British and
immunohistochemistry. Danish pedigrees, another form of diffuse cerebrovascular
478 Dementia

amyloidosis has elucidated the Worster-Drought syndrome


(Masters and Beyreuther, 2001) in which another novel amy-
loidogenic peptide aggregates in the brain.

46.4.9 WHITE MATTER CHANGES


Although the degenerative process in AD primarily affects
the grey matter, the white matter may not be spared. CT
scans may show areas of hypodensity. The precise patholog-
ical substrate remains controversial. Since amyloid angiop-
athy is often part of AD, it is possible that the white matter
changes result mainly from vascular causes. Symmetrical,
incomplete infarctions of the white matter histologically
correspond to partial loss of myelin sheaths, axons and oli-
godendrocytes. These changes are accompanied by mild Figure 46.13 Lewy bodies (L) and dystrophic neurites
astrocytosis and macrophage response, while there is hya- (N) in the midbrain of a patient who died with famil-
line fibrosis of small vessels. Cavitating infarctions do not ial Alzheimer’s disease with the amyloid precursor
protein 717 valine–isoleucine mutation. α-Synuclein
develop and this white matter damage, which occurs in the
immunohistochemistry.
absence of hypertensive vascular degeneration, is thought
to be caused by hypoperfusion (Brun and Englund, 1986).
However, severe loss of cortical neurones may also contrib- dementia with Lewy bodies contain the protein α-synuclein
ute to fibre loss and consequent pallor of white matter. (Spillantini et al., 1997). Familial and sporadic forms of AD
may also be found in combination with cerebrovascular dis-
ease. Rare sporadic cases of AD have been found in com-
46.5 FAMILIAL AUTOSOMAL bination with other neurodegenerative disorders including
DOMINANTLY INHERITED AD Pick’s disease, Creutzfeldt–Jakob disease, motor neurone
(FAD) disease and progressive supranuclear palsy.

AD is a disease primarily of old age and it is difficult to deter-


mine the proportion of genetic cases as many family members
46.6 PATHOGENESIS
will die before expressing the disease. Familial forms of AD
with multiple affected individuals are rare and account for While the cause of AD is now clearly defined as the cere-
probably fewer than 5% of AD cases. Mutations in the APP bral deposition of Aβ amyloid, the pathogenesis of AD is
gene and two related genes, presenilin-1 (PS1) and preseni- not fully understood. Although many risk factors have been
lin-2 (PS2), account for the majority of early-onset autosomal proposed, the evidence for most of these is weak except for
dominant cases of familial AD (Lamb, 1997) (see Chapter age, family history and ApoE genotype (Alafuzoff et al.,
50). Although clinical features may differ between fami- 2009; Chen et al., 2009). A genetic contribution to the aeti-
lies, the neuropathological features are not markedly differ- ology of AD has received strong support in recent years and
ent between FAD and early-onset sporadic cases. However, it is likely that more genes (such as BIN1, CLU, ABCA7,
prominent cerebellar plaques and Aβ deposition have been CRI, PICALM, MS4A6A, CD33, MS4A4E, CD2AP from the
reported in familial cases (Iseki et al., 1990; Struble et al., 1991; Alzgene top results, April 2015) will be associated with the
Fukutani et al., 1996, 1997). Computerized methods of image disease. In the more common non-familial, sporadic cases,
analysis have been used to more accurately define the patho- and in discordant monozygotic twins, environmental fac-
logical phenotype of AD. Using these techniques, differences tors may be important in the aetiology.
in Aβ load have been reported. Mann et al. (1996a, 1996b) The discovery of mutations in the APP and PS genes, all of
have shown that both APP and presenilin (PS) mutations lead which lead to overproduction of Aβ or preferential production
to measurable increases in Aβ load. More recent studies using of a long 42-amino acid form of Aβ, has confirmed the cen-
PET-amyloid ligands show a very distinctive accentuation of tral role of APP and Aβ deposition in the pathogenesis of AD
Aβ amyloid in the striatum (Villemagne et al., 2009). (Lamb, 1997). Deposition of Aβ appears to be an early event in
In those cases with an early onset, below 65 years of age, both AD and Down’s syndrome and precedes the development
the duration of illness tends to be shorter, and the density of plaques, NFTs, microgliosis and astrocytosis (Rozemuller
of NFTs and plaques is often greater than in sporadic, late- et al., 1989; Woltjer et al., 2009). The amyloid deposits char-
onset patients. Cortical and brainstem Lewy bodies may acteristic of AD, 42- to 43-amino acid Aβ fragments, are the
coexist with AD in APP (Lantos et al., 1992) and PS muta- abnormal proteolytic cleavage products of APP. Aβ has both
tion patients (Figure 46.13). It is now known that the Lewy toxic (Yanker et al., 1989) and trophic effects on neurones in
body and dystrophic neurites of Parkinson’s disease and culture, which may relate to plaque and NFT formation.
The neuropathology of Alzheimer’s disease 479

Metal ions such as aluminium, iron, copper and zinc (75%) of 51 unselected non-demented patients who died
have been implicated as an environmental toxic agent in the between the ages of 55 and 64 years (Ulrich, 1985). These
pathogenesis of AD. Accumulations of aluminosilicates and lesions were consistently found in the entorhinal cortex, the
iron in the central region of the core of plaques and within hippocampus, or both, suggesting that this region is a site
neurones containing NFTs have been reported (Candy of origin of the lesions. A morphometric analysis of cere-
et al., 1986). Another environmental factor implicated in Aβ bral slices by de la Monte (1989) showed that patients with
deposition is head trauma. Although Roberts et al. (1991) ‘preclinical’ AD had shrinkage of white matter comparable
showed that in a series of patients suffering sustained head to that found in AD, yet the cortical areas were normal,
injury, there were extensive deposits of Aβ in the neocortex, suggesting that white matter degeneration is an intrinsic
longer-term follow-up studies have failed to confirm any component of AD. In clinical AD, there was global cerebral
association (Chen et al., 2009). atrophy of both cortex and white matter. Neuronal death
Although varying amounts of Aβ with a range of morphol- in patients with AD is closely associated with extensive
ogies may be found in cognitively normal elderly individuals, synapse loss in the neocortex and this has been correlated
it is quantitatively much more severe and cytoarchitectoni- with cognitive impairment (Terry et al., 1991). Measures of
cally defined in AD (Majocha et al., 1988; Cairns et al., 1991). the morphological substrates of brain function frequently
The importance of overexpression of APP and its abnormal show an overlap between AD and control groups (Coleman
cleavage product, Aβ, in the pathogenesis of AD has been and Flood, 1987). There does not appear to be a consistent
demonstrated by transgenic mouse models (Borchelt et al., relationship between the pattern of vascular amyloid and
1998). Transgenic mice expressing the full-length human APP the distribution of plaques. Neither is there a correlation
develop Aβ deposits similar in morphology to those found in between vascular amyloid and dementia (Mountjoy et al.,
AD, but as noted above, differing somewhat in their human 1982). More recent studies have confirmed that the Aβ
‘subtype’ counterparts. These animal models will be useful for amyloid per se is probably not the best indicator of severity
testing anti-amyloid drugs (Morrissette et al., 2009). of disease – rather, the soluble Aβ, which are a prelude to
plaque formation show a closer correlation with the degree
of neurodegeneration (Mclean et al., 1999). With the advent
of molecular imaging probes for Aβ, many of the above
46.7 NEUROPATHOLOGY AND issues will be resolved (Jack et al., 2009).
CLINICAL FINDINGS

Alzheimer first suggested a correlation between the presence


46.8 DIAGNOSTIC PROBLEMS AND
of NFTs, plaques and dementia in 1906. Roth et al. (1966) and
Wilcock and Esiri (1982) demonstrated that elderly patients’
CRITERIA
mental test scores correlated with the neuropathological
lesions characteristic of AD. Many other neuropathological Despite the fact that the neurohistological abnormalities of
(Nelson et al., 2009) and neurochemical changes (Zubenko AD have been established, the neuropathological diagnosis
et al., 1991) have been associated with AD and the accuracy is not always straightforward (Cruz-Sánchez et al., 1995).
of clinical diagnosis ranges from 43% to 87% (Boller et al., There are several reasons to explain this problem. First, the
1989; Jellinger et al., 1990) and, in one sample of 26 cases, histopathology of AD is complex and the various abnormal-
100% (Morris et al., 1988). Conversely, inaccuracy in the ante-­ ities described previously, when severe and extensive, enable
mortem diagnosis of AD is common, and for this reason, post- a diagnosis to be made. However, there are considerable
mortem examination remains an essential component of AD differences from case to case and this histological hetero-
research. An accurate differential diagnosis is made difficult geneity of disease presents the most formidable difficulties
by the problems of distinguishing between mild dementia for the neuropathologist. The severity and distribution of
and the cognitive changes of ‘normal ageing’. Mild cognitive any of the histological lesions may considerably vary and
impairment is the earliest clinically recognizable form of AD, are influenced by the age and genetic background of the
which is now being evaluated with in vivo neuroimaging of patient, among other factors. Quantitative morphometry
Aβ load using PET radioliogands (Villemagne et al., 2013). and neurochemistry have supported the view that AD is not
Although not all studies have produced positive findings, a uniformly diffuse disorder: morphological abnormalities
there is general agreement that cases with earlier onset, and neurochemical deficits tend to be more severe and more
shorter duration of illness and greater cognitive impair- extensive in younger patients. There may be subtle differ-
ment have more cortical plaques, whereas late-onset cases ences in the pathology of familial and sporadic cases, and
that take a milder course tend to show less cortical change atypical cases exist both in the familial and sporadic forms.
(Bowen et al., 1979; Hansen et al., 1988). High correlations Second, although there are several histological abnormali-
were observed between cognitive performance and plaque ties in AD, only Aβ deposition approaches a level of speci-
counts in brain biopsies from dementing patients exam- ficity, since many of these lesions occur in normal ageing
ined earlier in the course of illness (Mann et al., 1986, 1988; and in other diseases of the nervous system. Third, AD may
Neary et al., 1986). Plaques and NFTs were detected in 38 concurrently occur with other diseases, most commonly
480 Dementia

with vascular disease and more rarely with other neurode- et al., 2002, 2004; Bates et al., 2009; Craig-Shapiro et al.,
generative disorders. 2009; Fagan et al., 2009; Woltjer et al., 2009).
For these reasons, it is important to establish a set of
neuropathological or neurochemical criteria. Although REFERENCES
several attempts have been made, no universally accepted
criteria exist for the neuropathological diagnosis of AD. Adalbert, R., Nogradi, A., Babetto, E. et al. (2009).
An attempt has been made to define AD in terms of hip- Severely dystrophic axons at amyloid plaques remain
pocampal pathology. Based on quantitative morphometry, continuous and connected to viable cell bodies. Brain,
it was suggested that more than 20 tangle-bearing neurones 132: 402–416.
per mm3 and fewer than 5600 neurones per mm3 should Agosta, F., Vossel, K.A., Miller, B.L. et al. (2009).
be diagnostic. This threshold, however, has proven too low Apolipoprotein E ε4 is associated with disease-specific
and would include 30% of mentally normal elderly people effects on brain atrophy in Alzheimer’s disease and
(Anderson and Hubbard, 1985). A North American panel frontotemporal dementia. Proceedings of the National
of neuropathologists has recommended age-adjusted plaque Academy of Sciences of the United States of America,
and tangle counts. In patients under 50, plaques and tangles 106: 2018–2022.
in excess of 2–5 per mm2 in the neocortex should secure the Alafuzoff, I., Aho, L., Helisalmi, S. et al. (2009). β-Amyloid
diagnosis of AD. Between 60 and 65 years, there should be deposition in brains of subjects with diabetes.
more than 8 plaques per mm2, between 66 and 75 years more Neuropathology and Applied Neurobiology, 35:
than 10 and over the age of 75, more than 15. In the first two 60–68.
age groups, some neurofibrillary tangles may be found, but Alzheimer, A. (1907). Über eine eigenartige Erkrankung
in the last group these may not be necessary for diagnosis der Hirnrinde. Allgemeine Zeitschrift für Psychiatrie,
(Khachaturian, 1985). The value of these criteria has, how- 64: 146–148.
ever, been diminished by the statement that these figures Anderson, J.M. and Hubbard, B.M. (1985). Definition of
can be revised downwards by as much as 50% in the pres- Alzheimer’s disease. Lancet, 1: 408.
ence of positive clinical history of AD. A survey of the prac- Arai, T., Mackenzie, I.R.A., Hasegawa, M. et al. (2009).
tical application of these criteria has revealed considerable Phosphorylated TDP-43 in Alzheimer’s disease and
variations in the use of criteria and the methods of exami- dementia with Lewy bodies. Acta Neuropathologica,
nation (Wisniewski et al., 1989). A European Multicentre 117: 125–136.
Study found that subjective assessment currently was more Armstrong, R.A., Cairns, N.J., Myers, D. et al. (1996).
reliable to compare cases from different laboratories, since A comparison of β-amyloid deposition in the
quantitative values are greatly influenced by the neurohisto- medial temporal lobe in sporadic Alzheimer’s dis-
logical technique employed (Duyckaerts et al., 1990). ease, Down’s syndrome and normal elderly brains.
The most widely used protocol for neuropathologi- Neurodegeneration, 5: 35–41.
cal assessment is that developed by the Task Force of the Armstrong, R.A., Myers, D. and Smith, C.U.M. (1993). The
Consortium to Establish a Registry for AD (CERAD). This spatial patterns of β/4 deposit subtypes in Alzheimer’s
takes clinical findings into consideration, and neuropatho- disease. Acta Neuropathologica, 86: 36–41.
logical criteria for ‘definite’, ‘probable’ and ‘possible’ AD Ball, M.J. (1977). Neuronal loss, neurofibrillary tangles and
are proposed (Mirra et al., 1991). This system is based on granulovacuolar degeneration in the hippocampus
a semi-quantitative assessment of plaque scores. The pres- with ageing brain and senile dementia: A quantitative
ence or absence of tangles does not enter into the formal study. Acta Neuropathologica, 37: 111–118.
assessment. The more recent National Institute on Aging Bates, K.A., Verdile, G., Li, Q.-X. et al. (2009). Clearance
and Reagan Institute (NIA-RI) criteria require both semi- mechanisms of Alzheimer’s amyloid-β peptide:
quantitative measures of plaque and tangle distribution Implications for therapeutic design and diagnostic
and additional sampling (Hyman and Trojanowski, 1997). tests. Molecular Psychiatry, 14: 469–486.
A change from previous criteria is the opinion that any Blanks, J.C., Hinton, D.R., Sadun, A.A. and Miller,
Alzheimer lesions should be considered as pathological C.A. (1989). Retinal ganglion cell degeneration in
even when they appear to be incidental. By acknowledging Alzheimer’s disease. Brain Research, 501: 364–372.
that a spectrum of changes exists within AD, these criteria Blocq, P. and Marinesco, G. (1892). Sur les lesions et la
may prove more successful than previous ones (Hyman and pathogénie de l’epilepsie dite essentielle. Semaine
Trojanowski, 1997). Medicale, 12: 445–446.
In the final analysis, it is likely that a combination of Boller, F., Lopez, O.L. and Moossy, J. (1989). Diagnosis of
morphologic and biochemical assays will yield a set of cri- dementia: Clinicopathologic correlations. Neurology,
teria through which a secure diagnosis of AD will be made. 39: 76–79.
Current indications are that blood, CSF and brain tissue Bondareff, E., Wischik, C.M., Novak, M. et al. (1990).
levels of Aβ (soluble, insoluble), tau, ApoE and BACE may Molecular analysis of neurofibrillary degeneration in
provide a more practical method for the diagnosis and ther- Alzheimer’s disease. American Journal of Pathology,
apeutic monitoring of AD (McLean et al., 1999; Holsinger 137: 711–723.
The neuropathology of Alzheimer’s disease 481

Borchelt, D.R., Woing, P.C., Sisodia, S.S. and Price, D.L. Crary, J.F., Trojanowski, J.Q., Schneider, J.A. et al. (2014).
(1998). Transgenic mouse models of Alzheimer’s Primary age-related tauopathy (PART): A com-
disease and amyotrophic lateral sclerosis. Brain mon pathology associated with human aging. Acta
Pathology, 8: 735–757. Neuropathologica, 128: 755–766.
Bowen, D.M., Spillane, J.A., Curzon, G. et al. (1979). Crowther, R.A. (1991). Straight and paired helical fila-
Accelerated ageing or selective neuronal loss as an ments in Alzheimer’s have a common structural unit.
important cause of dementia? Lancet, 1: 11–14. Proceedings of the National Academy of Sciences of
Braak, H. and Braak, E. (1988). Neuropil threads the United States of America, 88: 2288–2292.
occur in dendrites of tangle-bearing nerve cells. Cruz-Sánchez, F.F., Ravid, R. and Cuzner, M.L. (1995).
Neuropathology and Applied Neurobiology, 14: 39–44. Neuropathological Diagnostic Criteria for Brain
Braak, H., Braak, E., Grundke-Iqbal, I. and Iqbal, K. (1986). Banking. Oxford, United Kingdom: IOS Press.
Occurrence of neuropil thread, in the senile human Davis, C.A., Mann, D.M.A., Sumpter, P.Q. and Yates,
brain and in Alzheimer’s disease: A third location P.O. (1987). A quantitative morphometric analysis of
of paired helical filaments outside of neurofibrillary the neuronal and synaptic content of the frontal and
tangles and neuritic plaques. Neuroscience Letters, 65: temporal cortex in patients with Alzheimer’s disease.
351–355. Journal of the Neurological Sciences, 78: 151–164.
Braak, H., Braak, E. and Kalus, P. (1989). Alzheimer’s de la Monte, S.M. (1989). Quantitation of cerebral atro-
disease areal and laminar pathology in the occipital phy in preclinical and end-stage Alzheimer’s disease.
isocortex. Acta Neuropathologica, 77: 494–506. Annals of Neurology, 25: 450–459.
Brun, A. and Englund, E. (1986). A white matter disorder Delaère, P., Duyckaerts, C., Masters, C. et al. (1990). Large
in dementia of the Alzheimer type: A pathoanatomical amounts of neocortical βA4 deposits without neuritic
study. Annals of Neurology, 19: 253–262. plaques nor tangles in a psychometrically assessed, non-
Buckner, R.L., Sepulcre, J., Talukdar, T. et al. (2009). demented person. Neuroscience Letters, 116: 87–93.
Cortical hubs revealed by intrinsic functional connec- Dickerson, B.C., Bakkour, A., Salat, D.H. et al. (2009). The
tivity: Mapping, assessment of stability and relation cortical signature of Alzheimer’s disease: Regionally
to Alzheimer’s disease. Journal of Neuroscience, 29: specific cortical thinning relates to symptom severity
1860–1873. in very mild to mild AD dementia and is detectable in
Cairns, N.J., Atkinson, P.F., Hanger, D.P. et al. (1997a). Tau asymptomatic amyloid-positive individuals. Cerebral
protein in the glial cytoplasmic inclusions of multiple sys- Cortex, 19: 497–510.
tem atrophy can be distinguished from abnormal tau in Dickson, D.W., Ksiezak-Reding, H., Davies, P. and Yen,
Alzheimer’s disease. Neuroscience Letters, 230: 49–52. S.H. (1987). A monoclonal antibody that recognises
Cairns, N.J., Chadwick, A., Luthert, P.J. and Lantos, P.L. a phosphorylated epitope in Alzheimer neurofi-
(1991). β-Amyloid protein load is relatively uniform brillary tangles, neurofilaments and tau proteins
throughout neocortex and hippocampus in elderly immunostains granulovacuolar degeneration. Acta
Alzheimer’s disease patients. Neuroscience Letters, Neuropathologica, 73: 254–258.
129: 115–118. Divry, P. (1927). Etude histo-chimique des plaques séniles.
Cairns, N.J., Chadwick, A., Luthert, P.J. and Lantos, P.L. Journal Belge de Neurologie et de Psychiatrie, 27:
(1992). Astrocytosis, βA4-protein deposition and 643–657.
paired helical filament formation in Alzheimer’s dis- Duyckaerts, C., Delaère, P., Hauw, J.-J. et al. (1990). Rating
ease. Journal of the Neurological Sciences, 112: 68–75. of the lesions in senile dementia of the Alzheimer type:
Cairns, N.J., Fukutani, Y., Chadwick, A. et al. (1997b). A concordance between laboratories. A European
Apolipoprotein E, β-amyloid (Aβ), phosphorylated tau multicenter study under the auspices of EURAGE.
and apolipoprotein E genotype in Alzheimer’s disease. Journal of the Neurological Sciences, 97: 295–323.
Alzheimer Research, 3: 109–114. Duyckaerts, C., Delaère, P., Poulain, V. et al. (1988). Does
Candy, J.M., Oakley, A.E., Klinowski, J. et al. (1986). amyloid precede paired helical filaments in the senile
Aluminoscilicates and senile plaque formation in plaques? A study of 15 cases with graded intellectual
Alzheimer’s disease. Lancet, 1: 354–357. status in aging and Alzheimer’s disease. Neuroscience
Chen, X.H., Johnson, V.E., Uryu, K. et al. (2009). A lack of Letters, 91: 354–359.
amyloid β plaques despite persistent accumulation of Duyckaerts, C., Hauw, J.-J., Piette, F. et al. (1985). Cortical
amyloid β in axons of long-term survivors of traumatic atrophy in senile dementia of the Alzheimer type
brain injury. Brain Pathology, 19: 214–223. is mainly due to a decrease in cortical length. Acta
Coleman, P.D. and Flood, D.G. (1987). Neuron numbers Neuropathologica, 66: 72–74.
and dendritic extent in normal aging and Alzheimer’s Esiri, M.M., Hyman, B.T., Beyreuther, K. and Masters, C.L.
disease. Neurobiology of Aging, 8: 521–545. (1997). Ageing and dementia. In Graham, D.I. and
Craig-Shapiro, R., Fagan, A.M. and Holtzman, D.M. (2009). Lantos, P.L. (eds.), Greenfield’s Neuropathology, Sixth
Biomarkers of Alzheimer’s disease. Neurobiology of Edition, Vol. 2. London, United Kingdom: Arnold, pp.
Disease, 35: 128–140. 153–233.
482 Dementia

Esiri, M.M. and Morris, J.H. (1997). The Neuropathology Hanger, D.P., Brion, J.P., Gallo, J.M. et al. (1991). Tau in
of Dementia. Cambridge, NY: Cambridge University Alzheimer’s disease and Down’s syndrome is insoluble
Press. and abnormally phosphorylated. Biochemical Journal,
Esiri, M.M. and Wilcock, G.K. (1984). The olfactory 275: 99–104.
bulbs in Alzheimer’s disease. Journal of Neurology, Hansen, L.A., De Teresa, R., Davies, P. and Terry, R.D.
Neurosurgery and Psychiatry, 47: 56–60. (1988). Neocortical morphometry, lesion counts, and
Fagan, A.M., Head, D., Shah, A.R. et al. (2009). Decreased choline acetyltransferase levels in the age spectrum of
cerebrospinal fluid Aβ42 correlates with brain atrophy Alzheimer’s disease. Neurology, 38: 48–55.
in cognitively normal elderly. Annals of Neurology, 65: Harrington, C.R., Mukaetova-Ladinska, E.B., Hills, R. et al.
176–183. (1991). Measurement of distinct immunochemical
Fukutani, Y., Cairns, N.J., Rossor, M.N. and Lantos, P.L. presentations of tau protein in Alzheimer’s disease.
(1996). Purkinje cell loss and astrocytosis in the cer- Proceedings of the National Academy of Sciences of
ebellum in familial and sporadic Alzheimer’s disease. the United States of America, 88: 5842–5846.
Neuroscience Letters, 214: 33–36. Hauw, J.J., Duyckaerts, C. and Delaere, P. (1991).
Fukutani, Y., Cairns, N.J., Rossor, M.N. and Lantos, P.L. Alzheimer’s disease. In Duckett, S. (ed.), The Pathology
(1997). Cerebellar pathology in sporadic and familial of the Aging Nervous System. London, United
Alzheimer’s disease including APP 717(val-ile) mutation Kingdom: Lea and Febiger, pp. 113–147.
cases: A morphometric investigation. Journal of the Hinton, D.R., Sadun, A.A., Blanks, J.C. and Miller, C.A.
Neurological Sciences, 149: 177–184. (1986). Optic nerve degeneration in Alzheimer’s
Galloway, P.G., Perry, G., Kosik, K.S. and Gambetti, disease. New England Journal of Medicine, 315:
P. (1987). Hirano bodies contain tau protein. Brain 485–487.
Research, 403: 337–340. Hirano, A. (1965). Pathology of amyotrophic lateral scle-
Gallyas, F. (1971). Silver staining of Alzheimer’s neurofibril- rosis. In Gajdusek, D.C., Gibbs Jr., C.J. and Alpers, M.
lary changes by means of physical development. Acta (eds.), Slow, Latent and Temperate Virus Infections.
Morphologica Academiae Scientifica Hungarica, 19: Washington, DC: U.S. Government Printing Office, pp.
1–8. 23–26.
Gentleman, S.M., Bruton, C., Allsop, D. et al. (1989). Holsinger, R.M.D., McLean, C.A., Beyreuther, K. et al.
A demonstration of the advantages of immunostain- (2002). Increased expression of the amyloid precur-
ing in the quantification of amyloid plaque deposits. sor β-secretase in Alzheimer’s disease. Annals of
Histochemistry, 92: 355–358. Neurology, 51: 783–786.
Gibson, P.H. and Tomlinson, B.E. (1977). Numbers of Holsinger, R.M.D., Mclean, C.A., Collins, S.J. et al. (2004).
Hirano bodies in the hippocampus of normal and Increased β-secretase activity in cerebrospinal fluid of
demented people with Alzheimer’s disease. Journal of Alzheimer’s disease subjects. Annals of Neurology, 55:
the Neurological Sciences, 33: 199–206. 898–899.
Goedert, M., Spillantini, M.G., Cairns, N.J. and Crowther, Hubbard, B.M. and Anderson, J.M. (1981). A quantitative
R.A. (1992). Tau proteins of Alzheimer paired helical study of cerebral atrophy in old age and senile demen-
filaments; abnormal phosphorylation of all six iso- tia. Journal of the Neurological Sciences, 50: 135–145.
forms. Neuron, 8: 159–168. Hubbard, B.M. and Anderson, J.M. (1985). Age-related
Goedert, M., Wischik, C.M., Crowther, R.A. et al. (1988). variations in the neuron content of the cerebral cortex
Cloning and sequencing of the cDNA encoding a core in senile dementia of Alzheimer type. Neuropathology
protein of the paired helical filament of Alzheimer’s and Applied Neurobiology, 11: 369–382.
disease: Identification as the microtubule-associated Hyman, B.T. and Trojanowski, J.Q. (1997). Editorial on
protein tau. Proceedings of the National Academy consensus recommendations for the postmortem
of Sciences of the United States of America, 85: diagnosis of Alzheimer disease from the National
4051–4055. Institute on Aging and the Reagan Institute Working
Goldman, J.E. (1983). The association of actin with Hirano Group on diagnostic criteria for the neuropatho-
bodies. Journal of Neuropathology and Experimental logical assessment of Alzheimer’s disease. Journal
Neurology, 42: 146–152. of Neuropathology and Experimental Neurology, 56:
Goldstein, L.E., Muffat, J.A., Cherny, R.A. et al. (2003). 1095–1097.
Cystolic beta-amyloid deposition and supranuclear Iseki, E., Matsushita, M., Kosaka, K. et al. (1990).
cataracts in lenses from people with Alzheimer’s dis- Morphological characteristics of senile plaques in
ease. Lancet, 361: 1258–1265. familial Alzheimer’s disease. Acta Neuropathologica,
Gonatas, N.K., Anderson, A. and Vongelista, I. (1967). The 80: 227–232.
contribution of altered synapses in the senile plaque: Itagakai, S., McGeer, P.L., Akiyama, H. et al. (1989).
An electronmicroscopic study in Alzheimer’s demen- Relationship of microglia and astrocytes to amy-
tia. Journal of Neuropathology and Experimental loid deposits of Alzheimer’s disease. Journal of
Neurology, 26: 25–39. Neuroimmunology, 24: 173–182.
The neuropathology of Alzheimer’s disease 483

Jack, C.R., Lowe, V.J., Weigand, S.D. et al; and the Leigh, P.N., Probst, A., Dale, G.E. et al. (1989). New
Alzheimer’s Disease Neuroimaging Initiative. (2009). aspects of the pathology of neurodegenerative
Serial PIB and MRI in normal, mild cognitive impair- disorders as revealed by ubiquitin antibodies. Acta
ment and Alzheimer’s disease: Implications for Neuropathologica, 79: 61–72.
sequence of pathological events in Alzheimer’s dis- Levy, E., Carman, M.D., Fernandez-Madrid, I.J. et al.
ease. Brain, 132: 1355–1365. (1990). Mutation of the Alzheimer’s disease amyloid
Jellinger, K. (1988). The pedunculopontine nucleus in gene in hereditary cerebral hemorrhage, Dutch type.
Parkinson’s disease, progressive supranuclear palsy Science, 248: 1124–1126.
and Alzheimer’s disease. Journal of Neurology, Majocha, R.E., Benes, F.M., Reifel, J.L. et al. (1988).
Neurosurgery and Psychiatry, 51: 540–543. Laminar-specific distribution and infrastructural detail
Jellinger, K., Danielczyk, W., Fischer, P. and Gabriel, of amyloid in the Alzheimer’s disease cortex visualized
E. (1990). Clinicopathological analysis of dementia by computer-enhanced imaging of epitopes recog-
disorders in the elderly. Journal of the Neurological nized by monoclonal antibodies. Proceedings of the
Sciences, 95: 239–258. National Academy of Sciences of the United States of
Joachim, C.L., Morris, J.H. and Selkoe, D.J. (1989). Diffuse America, 85: 6182–6186.
senile plaques occur commonly in the cerebellum in Mann, D.M.A., Iwatsubo, T., Cairns, N.J. et al. (1996a).
Alzheimer’s disease. American Journal of Pathology, Amyloid β protein (Aβ) deposition in chromosome
135: 309–319. 14-linked Alzheimer’s disease: Predominance of
Joachim, C.L., Morris, J.H., Selkoe, D.J. and Kosik, K.S. Aβ42(43). Annals of Neurology, 40: 149–156.
(1987). Tau epitopes are incorporated into a wide Mann, D.M.A., Iwatsubo, T., Ihara, Y. et al. (1996b).
range of lesions in Alzheimer’s disease. Journal of Predominant deposition of amyloid-β42(43) in plaques
Neuropathology and Experimental Neurology, 46: in cases of Alzheimer’s disease and hereditary cere-
611–622. bral hemorrhage associated with mutations in the
Kahn, J., Anderton, B.H., Probert, A. et al. (1985). amyloid precursor protein gene. American Journal of
Immunohistological study of granulovacuolar degener- Pathology, 148: 1257–1266.
ation using monoclonal antibodies to neurofilaments. Mann, D.M.A., Yates, P.O. and Marcynink, B. (1985). Some
Journal of Neurology, Neurosurgery and Psychiatry, 48: morphometric observations in the cerebral cortex and
924–926. hippocampus in presenile Alzheimer’s disease, senile
Kang, J., Lemaire, H.G., Unterbeck, A. et al. (1987). The dementia of Alzheimer type and Down’s syndrome in
precursor of Alzheimer’s disease amyloid A4 pro- middle age. Journal of the Neurological Sciences, 69:
tein resembles a cell-surface receptor. Nature, 325: 139–159.
733–736. Mann, D.M.A., Yates, P.O. and Marcyniuk, B. (1986).
Keage, H.A.D., Carare, R.O., Friedland, R.P. et al. (2009). A comparison of nerve cell loss in cortical and sub-
Population studies of sporadic cerebral amyloid cortical structures in Alzheimer’s disease. Journal of
angiopathy and dementia: A systematic review. BMC Neurology, Neurosurgery and Psychiatry, 49: 310–312.
Neurology, 9: 3. Mann, D.M.A., Yates, P.O., Marcyniuk, B. et al. (1988). The
Khachaturian, Z.S. (1985). Diagnosis of Alzheimer’s dis- progression of the pathological changes of Alzheimer’s
ease. Archives of Neurology, 42: 1097–1104. disease in frontal and temporal neocortex examined
Kidd, M. (1963). Paired helical filaments in electron both at biopsy and at autopsy. Neuropathology and
­microscopy of Alzheimer’s disease. Nature, 197: Applied Neurobiology, 14: 177–195.
192–193. Marcyniuk, B., Mann, D.M.A. and Yates, P.O. (1986).
Kiuchi, A., Otsuka, N., Namba, Y. et al. (1991). Presenile The topography of cell loss from locus ceruleus in
appearance of abundant neurofibrillary tangles with- Alzheimer’s disease. Journal of the Neurological
out senile plaques in the brain in myotonic dystrophy. Sciences, 76: 335–345.
Acta Neuropathologica, 82: 1–5. Masliah, E., Hansen, L., Albright, T. et al. (1991).
Kowall, N.W. and Kosik, K.S. (1987). Axonal disruption and Immunoelectron microscopic study of synaptic pathol-
aberrant localization of tau protein characterize the ogy in Alzheimer’s disease. Acta Neuropathologica, 81:
neuropil pathology of Alzheimer’s disease. Annals of 428–433.
Neurology, 22: 639–643. Masliah, E., Terry, R.D., DeTeresa, R.M. and Hansen, L.A.
Lamb, B.T. (1997). Presenilins, amyloid-β and Alzheimer’s (1989). Immunohistochemical quantification of the
disease. Nature Medicine, 3: 28–29. synapse-related protein synaptophysin in Alzheimer
Lantos, P.L., Luthert, P.J., Hanger, D. et al. (1992). disease. Neuroscience Letters, 103: 234–239.
Familial Alzheimer’s disease with the amyloid Masters, C.L. and Beyreuther, K. (2001). The Worster-
­precursor protein position 717 mutation and Drought syndrome and other syndromes of dementia
sporadic Alzheimer’s disease have the same with spastic paraparesis: The paradox of molecu-
cytoskeletal pathology. Neuroscience Letters, lar pathology. Journal of Neuropathology and
137: 221–224. Experimental Neurology, 60: 317–319.
484 Dementia

Masters, C.L., Simms, G., Weinman, N.A. et al. (1985). Perry, G., Friedman, R., Shaw, G. and Chau, V. (1987).
Amyloid plaque core protein in Alzheimer disease Ubiquitin is detected in neurofibrillary tangles and
and Down syndrome. Proceedings of the National senile plaque neurites of Alzheimer’s disease brains.
Academy of Sciences of the United States of America, Proceedings of the National Academy of Sciences of
82: 4245–4249. the United States of America, 84: 3033–3036.
Matveev, S.V., Spielmann, H.P., Metts, B.M. et al. (2014). Price, D.L., Stuble, R.G., Altschuler, R.J. et al. (1985).
A distinct subfraction of Abeta is responsible for the Aggregation of tubulin in neurons in Alzheimer’s
high-affinity Pittsburgh compound B-binding site in disease. Journal of Neuropathology and Experimental
Alzheimer’s disease brain. Journal of Neurochemistry, Neurology, 44: 366.
131: 356–368. Probst, A., Anderton, B.H., Brion, J.P. and Ulrich, J. (1989).
McLean, C.A., Cherny, R.A., Fraser, F.W. et al. (1999). Senile plaque neurites fail to demonstrate anti-paired
Soluble pool of Aβ as a determinant of severity of helical filament and anti-microtubule associated
neurodegeration in Alzheimer’s disease. Annals of protein-tau immunoreactive proteins in the absence
Neurology, 46: 860–866. of neurofibrillary tangles in the neocortex. Acta
Mirra, S.S., Heyman, A., McKeel, D. et al. (1991). The con- Neuropathologica, 77: 430–436.
sortium to establish a registry for Alzheimer’s disease Ritchie, C.W., Bush, A.I., Mackinnon, A. et al. (2003).
(CERAD). Part II. Standardization of the neuropatho- Metal-protein attenuation with iodochlorhydroxyquin
logic assessment of Alzheimer’s disease. Neurology, (clioquinol) targeting Aβ amyloid deposition and toxic-
41: 479–486. ity in Alzheimer disease: A pilot phase 2 clinical tiral.
Mori, H., Kondo, J. and Ihara, Y. (1987). Ubiquitin is a Archives of Neurology, 60: 1678–1691.
component of paired helical filaments in Alzheimer’s Roberts, G.W., Gentleman, S.M., Lynch, A. and Graham,
disease. Science, 235: 1641–1644. D.I. (1991). βA4 amyloid protein deposition in brain
Morris, J.C., McKeel, D.W., Fulling, K. et al. (1988). after head trauma. Lancet, 338: 1422–1423.
Validation of clinical diagnostic criteria for Alzheimer’s Rogers, J. and Morrison, J.H. (1985). Quantitative mor-
disease. Annals of Neurology, 24: 17–22. phology and laminar distribution of senile plaques
Morrissette, D.A., Parachikova, A., Green, K.N., LaFerla, in Alzheimer’s disease. Journal of Neuroscience, 5:
F.M. (2009). Relevance of transgenic mouse models 2801–2808.
to human Alzheimer disease. Journal of Biological Roses, A.D. (1994). Apolipoprotein E affects the rate of
Chemistry, 284: 6033–6037. Alzheimer’s disease expression: β amyloid burden is a
Mountjoy, C.Q., Roth, M., Evans, N.J.R. and Evans, H.M. secondary consequence dependent on APOE genotype
(1983). Cortical neuronal counts in normal elderly con- and duration of disease. Journal of Neuropathology and
trols and demented patients. Neurobiology of Aging, Experimental Neurology, 53: 429–437.
4: 1–11. Roth, M., Tomlinson, B.E. and Blessed, G. (1966).
Mountjoy, C.Q., Thomlinson, B.E. and Gibson, P.H. (1982). Correlations between scores for dementia and counts
Amyloid senile plaques and cerebral blood ves- of ‘senile plaques’ in cerebral grey matter of elderly
sels. A semi-quantitative investigation of a possible subjects. Nature, 209: 109–110.
relationship. Journal of the Neurological Sciences, Rowe, C.C., Ng, S., Ackermann, U. et al. (2007). Imaging
57: 89–103. β-amyloid burden in aging and dementia. Neurology,
Neary, D., Snowden, J.S., Bowen, D.M. et al. (1986). 68: 1718–1725.
Neuropsychological syndromes in presenile Rozemuller, J.M., Eikelenboom, P., Stam, F.C. et al. (1989).
­dementia due to cerebral atrophy. Journal A4 protein in Alzheimer’s disease: Primary and second-
of Neurology, Neurosurgery and Psychiatry, ary cellular events in extracellular amyloid deposi-
49: 163–174. tion. Journal of Neuropathology and Experimental
Nelson, P.T., Braak, H. and Markesbery, W.R. (2009). Neurology, 48: 674–691.
Neuropathology and cognitive impairment in Simchowicz, T. (1911). Histologische Studien über die
Alzheimer disease: A complex but coherent relation- senile Demenz. Histologische und Histopathologische
ship. Journal of Neuropathology and Experimental Arbeiten, 4: 267–444.
Neurology, 68: 1–14. Spillantini, M.G., Schmidt, M.L., Lee, V.M. et al. (1997).
Ogomori, K., Kitamoto, T., Tateishi, J. et al. (1989). α-Synuclein in Lewy bodies. Nature, 388: 839–840.
β-Protein amyloid is widely distributed in Struble, R.G., Polinsky, R.J., Hedreen, J.C. et al. (1991).
the ­central nervous system of patients with Hippocampal lesions in dominantly inherited
Alzheimer’s disease. American Journal of Pathology, Alzheimer’s disease. Journal of Neuropathology and
134: 243–251. Experimental Neurology, 50: 82–94.
Pantelakis, S. (1954). Un type particulier d’angiopathie Tabaton, M., Cammarata, S., Mancardi, G.L. et al. (1991a).
sénile du système nervaux central: L’angiopathie con- Abnormal tau-reactive filaments in olfactory mucosa
gophile. Topographie et fréquence. Monatsschrift für in biopsy specimens of patients with probable
Psychiatrie und Neurologie, 128: 219–256. Alzheimer’s disease. Neurology, 41: 391–394.
The neuropathology of Alzheimer’s disease 485

Tabaton, M., Cammarata, S., Mancardi, G. et al. (1991b). Walker, L.C., Kitt, C.A., Cork, L.C. et al. (1988). Multiple
Ultrastructural localization of β-amyloid, tau, and ubiq- transmitter systems contribute neurites to individual
uitin epitopes in extracellular neurofibrillary tangles. senile plaques. Journal of Neuropathology and
Proceedings of the National Academy of Sciences of Experimental Neurology, 47: 138–144.
the United States of America, 88: 2098–2102. Whitehouse, P.J., Price, D.L., Clark, A.W. et al. (1981).
Tabaton, M., Mandybur, T.I., Perry, G. et al. (1989). The Alzheimer’s disease: Evidence for selective loss of
widespread alteration of neurites in Alzheimer’s dis- cholinergic neurons in the nucleus basalis. Annals of
ease may be unrelated to amyloid deposition. Annals Neurology, 10: 122–126.
of Neurology, 26: 771–778. Wilcock, G.K. and Esiri, M.M. (1982). Plaques, tangles and
Talamo, B.R., Ruder, R.A., Kosik, K.S. et al. (1989). dementia. Journal of the Neurological Sciences, 56:
Pathological changes in olfactory neurons in patients 343–356.
with Alzheimer’s disease. Nature, 337: 736–739. Wisniewski, H.M., Merz, P.A. and Iqbal, K. (1984).
Terry, R.D. (1985). Alzheimer’s disease. In Davis, R.L. and Ultrastructure of paired helical filaments of Alzheimer’s
Robertson, D.M. (eds.), Textbook of Neuropathology. neurofibrillary tangle. Journal of Neuropathology and
Baltimore, MD: Williams & Wilkins, pp. 824–841. Experimental Neurology, 43: 643–656.
Terry, R.D. (1990). Normal aging and Alzheimer’s disease: Wisniewski, H.M., Rabe, A., Zigman, W. and Silverman,
Growing problems. In Cancilla, P.A., Vogel, F.S. and W. (1989). Neuropathological diagnosis of Alzheimer’s
Kaufman, N. (eds.), Neuropathology. International disease. Journal of Neuropathology and Experimental
Academy of Pathology Monograph. Baltimore, MD: Neurology, 48: 606–609.
Williams & Wilkins, pp. 41–54. Woltjer, R.L., Sonnen, J.A., Sokal, I. et al. (2009).
Terry, R.D., Masliah, E., Salmon, D.P. et al. (1991). Physical Quantitation and mapping of cerebral detergent-
basis of cognitive alterations in Alzheimer’s disease: insoluble proteins in the elderly. Brain Pathology, 19:
Synapse loss is the major correlate of cognitive impair- 365–374.
ment. Annals of Neurology, 30: 572–580. Yamaguchi, H., Nakazato, Y., Hirai, S. and Shoji, M.
Terry, R.D., Peck, A., DeTeresa, R. et al. (1981). Some mor- (1990a). Immunoelectron microscopic localiza-
phometric aspects of the brain in senile dementia of tion of amyloid β protein in the diffuse plaques of
the Alzheimer type. Annals of Neurology, 10: 184–192. Alzheimer-type dementia. Brain Research, 508:
Tomlinson, B.E., Irving, D. and Blessed, G. (1981). Cell loss 320–324.
in the locus ceruleus in senile dementia of Alzheimer Yamaguchi, H., Nakazato, Y., Shoji, M. et al. (1990b).
type. Journal of the Neurological Sciences, 49: 419–428. Ultrastructure of the neuropil threads in the
Tomonaga, M. (1974). Ultrastructure of Hirano bodies. Alzheimer brain: Their dendritic origin and accumula-
Acta Neuropathologica, 28: 365–366. tion in the senile plaques. Acta Neuropathologica,
Tourtellotte, W.G. and Van Hoesen, G.W. (1991). The axo- 80: 368–374.
nal origin of a subpopulation of dystrophic neurites in Yamaguchi, H., Nakazato, Y., Shoji, M. et al. (1991).
Alzheimer’s disease. Neuroscience Letters, 129: 11–16. Ultrastructure of diffuse plaques in senile dementia
Ulrich, J. (1985). Alzheimer changes in nondemented of the Alzheimer type: Comparison with primitive
patients younger than sixty-five: Possible early stages plaques. Acta Neuropathologica, 82: 13–20.
of Alzheimer’s disease and senile dementia of the Yamamoto, T. and Hirano, A. (1985). Nucleus raphe
Alzheimer type. Annals of Neurology, 17: 273–277. dorsalis in Alzheimer’s disease: Neurofibrillary tangles
Villemagne, V.L., Ataka, S., Mizuno, T. et al. (2009). High and loss of large neurons. Annals of Neurology, 17:
striatal amyloid beta-peptide deposition across dif- 573–577.
ferent autosomal Alzheimer disease mutation types. Yanker, B.A., Dawes, L.R., Fisher. S. et al. (1989).
Archives of Neurology, 66: 1537–1544. Neurotoxicity of a fragment of the amyloid precur-
Villemagne, V.L., Burnham, S., Bourgeat, P. et al; for the sor associated with Alzheimer’s disease. Science, 245:
Australian Imaging Biomarkers and Lifestyle (AIBL) 417–420.
Research Group. (2013). Amyloid β deposition, neu- Zubenko, G.S., Moossy, J., Martinez, J. et al. (1991).
rodegeneration and cognitive decline in sporadic Neuropathologic and neurochemical correlates of
Alzheimer’s disease: A prospective cohort study. The psychosis in primary dementia. Archives of Neurology,
Lancet Neurology, 12: 357–367. 48: 619–624.
47
Neurochemistry of Alzheimer’s disease

PAUL T. FRANCIS

(Francis et al., 1993, 2010b). This chapter examines both


47.1 INTRODUCTION cognitive and non-cognitive (behavioural) correlates of
neurotransmitter dysfunction, the latter also being consid-
The demonstration of deficits in choline acetyltransfer- ered to relate to structural and functional alterations in the
ase (ChAT) activity in patients dying with Alzheimer’s central nervous system (CNS). Such changes are important,
disease (AD) (Bowen et al., 1976; Davies and Maloney, partially because carers find behavioural disturbances dif-
1976), together with human anticholinergic drug studies ficult to cope with and the presence of such behaviours in
(Drachman and Leavitt, 1974) led to the eventual develop- AD patients often leads to institutionalization. This chap-
ment of cholinesterase inhibitors (ChEIs) for the symptom- ter focuses on cholinergic and glutamatergic systems; for a
atic treatment of AD. Present neurochemical investigations comprehensive review of neurotransmission in AD see Lai
of this system attempt to understand how early the choliner- et al. (2007).
gic deficit develops and the extent to which it occurs in mild
cognitive impairment (MCI). In addition, recognition of the
role of the glutamatergic system in learning and memory 47.2.1 ACETYLCHOLINE
has prompted study of biochemical markers of the major
Neuropathologically, loss of neurones from the nucleus of
excitatory neurotransmitter system. Anatomical studies
Meynert (Ch4 cholinergic nucleus) is well documented in
suggest interconnectivity between glutamatergic and cho-
AD, although, the extent of the loss reported varies from
linergic systems but their exact role in AD still requires
moderate to severe and it has been suggested that in AD
further elucidation. Just as cholinergic and glutamatergic
cholinergic dysfunction exceeds neuronal degeneration
disruption may be additive in contributing to cognitive and
(Perry et al., 1992). This is consistent with studies of cholin-
behavioural symptoms of dementia, so can pharmacologi-
ergic markers in biopsy samples from patients who had AD
cal replacement therapy be synergistic (Francis et al., 1993,
for approximately 3 years, where reduction in functional
2010b; Francis, 2005).
cholinergic measures, acetylcholine synthesis and choline
The amyloid cascade hypothesis has undergone many
uptake exceeded the reduction in choline acetyltransferase
revisions but still provides a framework in which to under-
(ChAT) activity (Francis et al., 1985).
stand disease progression in AD (Hardy, 2006). It is widely
On the basis of the aforementioned evidence, neocortical
accepted that neurotransmitter deficits, neuronal and syn-
cholinergic innervation appears to be lost at an early stage
apse loss and subsequent decline are final consequences of
of the disease. Beach et al. (2000) reported neurofibrillary
this cascade.
tangles (NFTs) in entorhinal cortex and a 20%–30% loss of
ChAT activity in brains from patients in the earliest stages of
AD, namely Braak stages I and II and tangle formation in the
main cholinergic nucleus is present in MCI (Mesulam et al.,
47.2 NEUROCHEMISTRY 2004). However, another study using the Clinical Dementia
Rating scale (CDR) suggests that the greatest reduction in
From a neurochemical standpoint, acetylcholine (ACh), markers of the cholinergic system occurs between moderate
glutamate, serotonin and noradrenaline are the major (CDR 2.0) and severe (CDR 5.0) disease with little change
transmitter systems affected in AD with relative sparing of between the non-demented patients and those in the mild
dopamine, γ-aminobutyric acid (GABA) and most peptides stages of dementia (CDR 0-2) (Davis et al., 1999). In contrast
486
Neurochemistry of Alzheimer’s disease 487

to a study on MCI, the suggested AD prodromal reported was increased in the frontal cortex of AD patients with
an increase in ChAT activity in some cortical regions delusions and in the temporal cortex of those with halluci-
(DeKosky et al., 2002). The acknowledged problems of these nations and then compared with patients without psychotic
latter studies is that ChAT activity is not the rate-limiting symptoms. This suggests a role of M2 receptors in the psy-
step of ACh synthesis and hence, there could be cholinergic chosis of AD and may provide the rationale for treatment of
dysfunction before structural loss, possibly linked to aber- behaviourally disturbed AD patients with M2 antagonists
rant sprouting. Changes in cholinergic neurones appear to (Lai et al., 2001).
relate to aspects of cognitive function (Francis et al., 1985) Related to these findings in AD, a study of rivastigmine
as well as non-cognitive behaviour (Minger et al., 2000; in patients with Parkinson’s disease dementia showed a
Gauthier et al., 2002). It has also been suggested that ace- positive outcome (Emre et al., 2004); such patients have a
tylcholine (Ach) is centrally involved in the process of con- greater cholinergic deficit than AD patients (Perry et al.,
scious awareness (Perry et al., 1999) and that, the variety 1994).
of clinical symptoms associated with cholinergic dysfunc-
tion in AD and related disorders reflects disturbances in the 47.2.3 GLUTAMATE
conscious processing of information.
Nicotinic and muscarinic (M2) ACh receptors, most Glutamate is the principal excitatory neurotransmitter of
of which are considered to be located on cholinergic ter- the brain, being used at approximately two-thirds of syn-
minals, are reduced in AD (Court et al., 2001; Lai et al., apses and as a consequence, the majority of neurones and
2001). Reductions in nicotinic receptors are confined to the glia have receptors for glutamate. An integral part of pro-
α4β2 subtype. The postsynaptic cholinergic system (usu- tein, energy and ammonia metabolism of all cells with a
ally present upon glutamatergic neurones) appears to be high intracellular concentration, it has been difficult to dis-
less affected, which is a hopeful sign for therapeutic inter- tinguish the presynaptic transmitter pool of glutamate from
vention. Postsynaptic muscarinic M1 receptors (Perry et the metabolic pool (Francis et al., 1993). Considered to be
al., 1990) are relatively preserved, although, there may be the main neurotransmitter of neocortical and hippocampal
a degree of functional uncoupling (Tsang et al., 2006). The pyramidal neurones, glutamate is thus involved in higher
enzyme responsible for the breakdown of ACh acetylcho- mental functions such as cognition and memory (Francis
linesterase, is reduced, perhaps in part, due to a loss of cho- et al., 1993). An important mechanism by which glutamate
linergic terminals, while butyrylcholinesterase activity (the may contribute to learning and memory functions is via
function of which is not yet fully understood and which is long-term potentiation (LTP) at pyramidal neurone syn-
present in extrasynaptic areas and plaques), increases (Perry apses (Baudry and Lynch, 2001). LTP is a form of synaptic
et al., 1978). strengthening following brief high-frequency stimulation.
Histological AD studies indicate loss of pyramidal neu-
47.2.2 ACETYLCHOLINE AND COGNITION rones and their synapses together with surrounding neu-
ropil (Esiri, 1991). Corticortical and corticofugal-projecting
A prediction of the cholinergic hypothesis is that, drugs pyramidal neurones are lost together with those of the ento-
likely to potentiate this function should improve cognition rhinal and hippocampal CA1 region. Remaining neurones
in AD patients. There are a number of treatment approaches are subject to NFT formation. Uptake of d-aspartate, a
to the amelioration of the cholinergic deficit; however, the putative marker of glutamatergic nerve endings, is reduced
use of acetylcholinesterase inhibitors (AChEIs) is the most in many cortical areas in AD brains (Procter et al., 1988).
well-developed approach till date (Francis et al., 1999). Biochemical evidence suggests a presynaptic ‘double
During the late 1980s and early 1990s, the first cholino- blow’ as the activity of glutamatergic neurones is heavily
mimetic compound, tacrine, underwent large-scale clinical influenced by the cholinergic system, also dysfunctional
studies and the ChEI treatment benefits were established in in AD patients. The clinical relevance of these changes is
patients with probable AD. A so-called second-generation emphasized because glutamatergic and cholinergic dys-
of ChEIs has been developed including donepezil, rivastig- function are both strong correlates of cognitive decline in
mine and galantamine (Francis et al., 1999; Wilkinson et AD. Glutamatergic (and cholinergic) cells die over a period
al., 2004). Such compounds demonstrate a clinical effect of years and in a very specific regional and neurochemi-
and magnitude of benefit of at least that reported for tacrine, cally selective pattern (Francis et al., 2012). The selectiv-
but with a more favourable clinical profile. Evidence has ity is intriguing because cholinergic neurones of the basal
emerged from clinical trials of ChEIs that such drugs forebrain dies while those of the pons are spared; glutama-
may improve the behavioural symptoms of AD patients. tergic neurones of the frontal, temporal and parietal cor-
Physostigmine, tacrine, rivastigmine and donepezil have tex are lost while apparently similar neurones in the motor
variously been reported in placebo-controlled trials to and sensory cortex are unaffected. Factors that may lead to
decrease psychotic symptoms, agitation, apathy, anxiety, necrosis or apoptosis in selected groups of neurones in AD
disinhibition, pacing, aberrant motor behaviour (inability may include tangles, Aβ toxicity, microglia, free radical gen-
to sit still) and lack of cooperation (Cummings et al., 2008). eration, excitotoxicity (too much and too little glutamater-
In one study, muscarinic acetylcholine M2 receptor density gic neurotransmission) and withdrawal of trophic factors.
488 Dementia

The strongest evidence suggests that most of glutamatergic and memory (Lynch, 1998). Circumstantial evidence of the
pyramidal neurones die as a consequence of the presence involvement of glutamatergic pathways includes the well-
of NFTs within the cytoplasm (Kowall and Beal, 1991). established role of structures such as, the hippocampus in
However, this may only account for up to 50% of such cells. learning and memory. More specifically, lesions of certain
Glutamate is synthesized in nerve terminals by one of glutamatergic pathways impair learning and memory. In
several possible enzymes. First, glutamine can be converted addition, glutamate and glutamate receptors are involved
to glutamate by the action of the mitochondrial enzyme in mechanisms of synaptic plasticity (LTP and long-term
glutaminase (Procter et al., 1988); alternatively glutamate depression or LTD, of the synapse), which are considered to
can be produced by transamination from aspartate in underlie learning and memory (Baudry and Lynch, 2001).
the cytosol. Direct measurement of glutaminase activity Loss of synapses and pyramidal cell perikarya from the
was unaffected in AD (Procter et al., 1988). By contrast, neocortex of AD patients correlates with measures of cog-
glutaminase-positive neurones were reduced in number nitive decline and is considered to be the best evidence of
and subject to tangle formation (Kowall and Beal, 1991). a functional role of glutamatergic involvement in cognitive
Several groups have shown reductions in the concentra- dysfunction of AD patients (Francis et al., 1993).
tion of glutamate in AD tissue and lumbar CSF (Lowe et al.,
1990). Although, glutamate neurotransmission failure was 47.2.5 THE GLUTAMATERGIC SYSTEM
not extensive in these studies, glutamate concentration was AND AD PATHOLOGY
reduced by 14% in temporal lobe biopsy samples and by 86%
in the terminal zone of the perforant pathway at autopsy of Excitotoxic cell death involves excess activation of receptors,
AD patients (Hyman et al., 1987). leading to raised intracellular Ca2+ and consequent acti-
More recent investigations have examined the status of vation of a cascade of enzymes, resulting in cell death by
the vesicular glutamate transporters (VGLUT), VGLUT1 necrosis or apoptosis (Lipton, 1999). During the 1980s
and VGLUT2 (Bellocchio et al., 2000). These proteins are it was also suggested that endogenous glutamate could
only present in the glutamatergic neurone terminals and accumulate and become excitotoxic, perhaps, as a result
therefore, represent a useful marker of their number. of impaired clearance (as a consequence of disrupted
Studies indicate that there is a reduction in VGLUT1 (but transporter function or indirectly in conditions of reduced
not VGLUT2) in parietal and occipital cortex in AD but energy availability). There is some evidence that energy lev-
not in temporal cortex (Kirvell et al., 2006). Other studies els may be reduced in AD due to perturbed mitochondrial
link this reduction to cognitive impairment (Kashani et al., function (Francis et al., 1993) and considerable evidence
2008). for oxidative damage of proteins including the glutamate
Upon release into the synapse, approximately 95% of transporter (Keller et al., 1997). Others have cautioned that
glutamate is removed by glutamate transporter proteins there is no simple relationship between raised extracellular
present upon glial cells named GLT, GLAST and EAAC glutamate concentrations and cell death in vivo. It remains
(Danbolt, 2001). Reductions in glutamate uptake in AD possible that changes in numbers of glutamate receptors or
cases have been reported in fresh (unfrozen) post-mortem changes in ion selectivity may lead to cell death over time.
brains (Procter et al., 1994). Antibodies directed against For instance, the large numbers of calcium-permeable
the individual glutamate transporters reveal conflicting AMPA receptors present on basal forebrain cholinergic
data. Reduced levels of GLT protein (but not its messen- neurones may be linked to their loss in AD (Ikonomovic
ger ribonucleic acid [mRNA]), with normal levels of both and Armstrong, 1996).
GLAST and EAAC have been reported (Danbolt, 2001). The glutamatergic system is a significant target for
Even on assuming there was no reduction of transporter AD-related pathology in which tangles occur principally in
protein, there is considerable evidence for oxidative dam- glutamatergic pyramidal neurones, these cells are lost and
age of proteins such as these glutamate transporters (Begni much of the observed synaptic pathology will involve these
et al., 2004). This might explain the functional deficit in cells. Tangles, cell and synapse loss are the strongest cor-
glutamate uptake identified above and is likely to lead to relates of cognitive impairment in AD patients (Neary et
elevations in synaptic concentrations of glutamate (Francis, al., 1986; Terry et al., 1991). There is also an emerging role
2009; Francis et al., 2012). of tangle formation within pyramidal neurones of specific
brain regions as a causative mechanism of agitation and
47.2.4 GLUTAMATE AND COGNITION aggression in AD (Tekin et al., 2001; Guadagna et al., 2012).

A role for glutamate and glutamate receptors in learning and 47.2.6 INTERACTIONS BETWEEN
memory is widely recognized. For example, N-methyl-d- CHOLINERGIC AND
aspartate (NMDA) antagonists impair learning and mem- GLUTAMATERGIC SYSTEMS
ory, while NMDA agonists and facilitators improve memory
(Francis et al., 1993). Likewise, AMPAkines (positive mod- It is important to remember that glutamatergic neurones of
ulators of alpha-amino-3-hydroxy-5-methyl-4-isoxazole- the neocortex and hippocampus are influenced by acetyl-
propionic acid [AMPA] receptor function) facilitate learning choline through nicotinic and muscarinic receptors (Dijk
Neurochemistry of Alzheimer’s disease 489

et al., 1995; Chessell and Humphrey, 1995). Terminals of complex. Reduction of 5-HT as well as its metabolites (nota-
cholinergic neurones are found in all layers of the neo- bly 5-hydroxyindolacetic acid, 5-HIAA) has been reported
cortex, synapsing with pyramidal neurones in layers II/III in many studies of post-mortem AD brains and studies of
and V (Turrini et al., 2001). Both muscarinic and nicotinic neurosurgical biopsy material (Francis et al., 2010a). There
receptors activate pyramidal neurones and hence, facili- is also evidence of plasticity in the serotonergic system as
tate glutamate release in rat model receptors (Chessell and the ratio 5-HIAA/5-HT is often found to increase in both
Humphrey, 1995; Dijk et al., 1995). Thus, under activity frontal and temporal cortex. With respect to serotonergic
of the cholinergic system, it is likely to have effects on the receptors, the picture is equally complex with region specific
activity of the glutamatergic system and conversely, it fol- changes that relate to the presence of behavioural symptoms
lows that treatment of patients with cholinomimetics which with particular links to agitation.
is likely to increase glutamatergic function. Because of the widespread influence of the serotonergic
system on pathways important for cognition and behaviour,
47.2.7 TREATMENT STRATEGIES BASED together with the receptor diversity, this system has con-
ON FACILITATING GLUTAMATERGIC siderable potential for pharmacological manipulation. The
NEUROTRANSMISSION rationale for targeting 5-HT2A receptors for some behav-
iours and 5-HT6 for cognition and behaviour is at the fore-
Cholinergic stimulation is one pathway to enhance gluta- front of clinical studies (Ramirez et al., 2014).
matergic function. Other treatment strategies that more
directly increase the activity of remaining glutamater-
gic neurones, without causing excitotoxicity, represent an
47.3 CONCLUSION
important target for the symptomatic treatment of AD and
may have a disease modifying effect. Several approaches
have been tried, including positive modulation of both The changes in cholinergic neurotransmission seen in the
AMPA and NMDA receptors. AMPAkines, which are con- brains of patients dying with AD provided the rationale
sidered to work by increasing the sensitivity of these recep- for the development of compounds aimed at symptomatic
tors, have been in clinical trial for MCI but with modest relief. Treatment of AD patients with ChEIs has confirmed
success (Johnson and Simmon, 2002). the relevance of this finding. However, it has long been rec-
Perhaps, the most surprising development is the suc- ognized that the major changes in AD are likely to involve
cess of the un-competitive NMDA antagonist meman- the abundant cortical glutamatergic pyramidal neurones.
tine in clinical trials in moderate and severe AD patients Hence, there is a need for this change to be addressed. Since
(Reisberg et al., 2003). One would normally consider that increasing cholinergic function will also increase glutama-
such an approach – blockade of a receptor that would tergic activity, there may be synergistic benefit from co-
normally be activated in learning and memory – would administration of drugs that target both the cholinergic
be counter-intuitive. However, there is evidence that this system (ChEIs) and glutamatergic systems (memantine).
molecule acts like the endogenous NMDA antagonist, There is renewed effort to identify improved symptomatic
magnesium ions, able to prevent background activation of treatments to combine with ChEIs and memantine and
the NMDA receptor (‘noise’), while allowing activation of serotonergic agents are at the forefront of these efforts. As
this receptor for LTP formation (Francis et al., 2012). The disease modifying treatments become available, it is likely
observed effects of memantine on agitation and aggression that symptomatic therapy will continue to be required and
may be explained by this mechanism; however, there is indeed there is evidence that such treatment may also slow-
preclinical evidence to suggest that this drug is capable of down the disease progression.
altering the phosphorylation state of tau, the precursor to
tangle formation (Francis, 2009). There is also evidence of REFERENCES
a benefit of the addition of memantine to established treat-
ment with a ChEI in AD patients (Tariot et al., 2004). Since Baudry, M. and Lynch, G. (2001). Remembrance of
ChEIs are likely to act in part by increasing glutamate arguments past: How well is the glutamate recep-
release (‘signal’) (Dijk et al., 1995; Francis et al., 2012), the tor hypothesis of LTP holding up after 20 years?
benefit may be hypothesized to come from the combina- Neurobiology of Learning and Memory, 76: 284–297.
tion of a reduction in glutamate ‘noise’ (by memantine) Beach, T.G., Kuo, Y.M., Spiegel, K. et al. (2000). The
and an increase in discrete glutamate signals (donepezil) cholinergic deficit coincides with A beta deposition
(Francis et al., 2012). at the earliest histopathologic stages of Alzheimer
disease. Journal of Neuropathology and Experimental
47.2.8 SEROTONERGIC SYSTEMS IN AD Neurology, 59: 308–313.
Begni, B., Brighina, L., Sirtori, E. et al. (2004). Oxidative
Extensive serotonergic denervation of the neocortex and stress impairs glutamate uptake in fibroblasts from
hippocampus has been reported in AD; however, the con- patients with Alzheimer’s disease. Free Radical Biology
tribution of such changes towards clinical symptoms is and Medicine, 37: 892–901.
490 Dementia

Bellocchio, E.E., Reimer, R.J., Fremeau, R.T. Jr. and Francis, P.T., Palmer, A.M., Snape, M. and Wilcock, G.K.
Edwards, R.H. (2000). Uptake of glutamate into syn- (1999). The cholinergic hypothesis of Alzheimer’s
aptic vesicles by an inorganic phosphate transporter. disease: A review of progress. Journal of Neurology,
Science, 289: 957–960. Neurosurgery and Psychiatry, 66: 137–147.
Bowen, D.M., Smith, C.B., White, P. and Davison, A.N. Francis, P.T., Parsons, C.G. and Jones, R.W. (2012).
(1976). Neurotransmitter-related enzymes and indices Rationale for combining glutamatergic and cho-
of hypoxia in senile dementia and other abiotrophies. linergic approaches in the symptomatic treat-
Brain, 99: 459–496. ment of Alzheimer’s disease. Expert Review of
Chessell, I.P. and Humphrey, P.P.A. (1995). Nicotinic Neurotherapeutics, 12: 1351–1365.
and muscarinic receptor-evoked depolarisations Francis, P.T., Ramirez, M.J. and Lai, M.K. (2010a).
recorded from a novel cortical brain slice preparation. Neurochemical basis for symptomatic treatment of
Neuropharmacology, 34: 1289–1296. Alzheimer’s disease. Neuropharmacology, 59: 221–229.
Court, J., Martin-Ruiz, C., Piggott, M. et al. (2001). Francis, P.T., Ramirez, M.J., Lai, M.K. et al. (2010b).
Nicotinic receptor abnormalities in Alzheimer’s dis- Neurochemical basis for symptomatic treatment of
ease. Biological Psychiatry, 49: 175–184. Alzheimer’s disease. Neuropharmacology, 59: 221–229.
Cummings, J.L., Mackell, J. and Kaufer, D. (2008). Francis, P.T., Sims, N.R., Procter, A.W. and Bowen, D.M.
Behavioral effects of current Alzheimer’s disease (1993). Cortical pyramidal neurone loss may cause
treatments: A descriptive review. Alzheimer’s and glutamatergic hypoactivity and cognitive impairment in
Dementia, 4 (1): 49–60. Alzheimer’s disease: Investigative and therapeutic per-
Danbolt, N.C. (2001). Glutamate Uptake. Progress in spectives. Journal of Neurochemistry, 60 (5): 1589–1604.
Neurobiology, 65: 1–105. Gauthier, S., Feldman, H., Hecker, J. et al. (2002). Efficacy
Davies, P. and Maloney, A.J.F. (1976). Selective loss of cen- of donepezil on behavioral symptoms in patients with
tral cholinergic neurons in Alzheimer’s disease. Lancet, moderate to severe Alzheimer’s disease. International
2 (8000): 1403. Psychogeriatrics, 14: 389–404.
Davis, K.L., Mohs, R.C., Marin, D. et al. (1999). Cholinergic Guadagna, S., Esiri, M.M., Williams, R.J. and Francis,
markers in elderly patients with early signs of P.T. (2012). Tau phosphorylation in human brain:
Alzheimer disease. JAMA, 281: 1401–1406. Relationship to behavioral disturbance in dementia.
DeKosky, S.T., Ikonomovic, M.D., Styren, S.D. et al. (2002). Neurobiology of Aging, 33: 2798–2806.
Upregulation of choline acetyltransferase activity in Hardy, J. (2006). A hundred years of Alzheimer’s disease
hippocampus and frontal cortex of elderly subjects research. Neuron, 52 (1): 3–13.
with mild cognitive impairment. Annals of Neurology, Hyman, B.T., Van Hoesen, G.W. and Damasio, A.R.
51: 145–155. (1987). Alzheimer’s disease: Glutamate depletion in
Dijk, S.N., Francis, P.T., Stratmann, G.C. and Bowen, D.M. the hippocampal perforant pathway zone. Annals of
(1995). Cholinomimetics increase glutamate outflow by Neurology, 22: 37–40.
an action on the corticostriatal pathway: Implications Ikonomovic, M.D. and Armstrong, D.M. (1996).
for Alzheimer’s disease. Journal of Neurochemistry, 65: Distribution of AMPA receptor subunits in the nucleus
2165–2169. basalis of Meynert in aged humans: Implications for
Drachman, D.A. and Leavitt, J. (1974). Human memory selective neuronal degeneration. Brain Research, 716:
and the cholinergic system. Archives of Neurology, 30: 229–232.
113–121. Johnson, S.A. and Simmon, V.F. (2002). Randomized,
Emre, M., Aarsland, D., Albanese, A. et al. (2004). double-blind, placebo-controlled international clinical
Rivastigmine for dementia associated with Parkinson’s trial of the Ampakine CX516 in elderly participants
disease. New England Journal of Medicine, 351: with mild cognitive impairment: A progress report.
2509–2518. Journal of Molecular Neuroscience, 19: 197–200.
Esiri, M. (1991). Neuropathology. In R. Jacoby and C. Kashani, A., Lepicard, E., Poirel, O. et al. (2008). Loss of
Oppenheimer (eds.), Psychiatry in the Elderly. Oxford: VGLUT1 and VGLUT2 in the prefrontal cortex is cor-
Oxford University Press, pp. 113–147. related with cognitive decline in Alzheimer disease.
Francis, P.T. (2005). The glutamatergic system and Neurobiology of Aging, 29: 1619–1630.
its role in higher mental function. Journal of Keller, J.N., Mark, R.J., Bruce, A.J. et al. (1997).
Psychopharmacology, 19: A7. 4-hydroxynonenal, an aldehydic product of mem-
Francis, P.T. (2009). Altered glutamate neurotransmission brane lipid peroxidation, impairs glutamate trans-
and behaviour in dementia: Evidence from studies of port and mitochondrial function in synaptosomes.
memantine. Current Molecular Pharmacology, 2: 77–82. Neuroscience, 80: 685–696.
Francis, P.T., Palmer, A.M., Sims, N.R. et al. (1985). Kirvell, S.L., Esiri, M.M. and Francis, P.T. (2006). Down
Neurochemical studies of early-onset Alzheimer’s regulation of vesicular glutamate transporters precede
disease. Possible influence on treatment. New England cell loss and pathology in Alzheimer’s disease. Journal
Journal of Medicine, 313: 7–11. of Neurochemistry, 98: 939–950.
Neurochemistry of Alzheimer’s disease 491

Kowall, N.W. and Beal, M.F. (1991). Glutamate-, types. Journal of Neural Transmission – Parkinsons
glutaminase-, and taurine-immunoreactive neurons Disease and Dementia Section, 2: 149–158.
develop neurofibrillary tangles in Alzheimer’s disease. Perry, E., Walker, M., Grace, J. and Perry, R. (1999).
Annals of Neurology, 29 (2): 162–167. Acetylcholine in mind: A neurotransmitter correlate
Lai, M.K., Lai, O.F., Keene, J. et al. (2001). Psychosis of of consciousness? Trends in Neuroscience, 22 (6):
Alzheimer’s disease is associated with elevated musca- 273–280.
rinic M2 binding in the cortex. Neurology, 57: 805–811. Procter, A.W., Francis, P.T., Holmes, C. et al. (1994). Beta-
Lai, M.K.P., Ramirez, M.J., Tsang, S.W. and Francis, P.T. amyloid precursor protein isoforms show correlations
(2007). Alzheimer’s disease as a neurotransmit- with neurones but not with glia of demented subjects.
ter disease. In D. Dawbarn and S.J. Allen (eds.), Acta Neuropathologica, 88 (6): 545–552.
Neurobiology of Alzheimer’s Disease, Vol. 3. Oxford: Procter, A.W., Palmer, A.M., Francis, P.T. et al. (1988).
Oxford University Press, pp. 245–282. Evidence of glutamatergic denervation and possible
Lipton, P. (1999). Ischemic cell death in brain neurons. abnormal metabolism in Alzheimer’s disease. Journal
Physiological Reviews, 79: 1431–1568. of Neurochemistry, 50: 790–802.
Lowe, S.L., Bowen, D.M., Francis, P.T. and Neary, D. (1990). Ramirez, M.J., Lai, M.K., Tordera, R.M. and Francis, P.T.
Ante mortem cerebral amino acid concentrations (2014). Serotonergic therapies for cognitive symptoms
indicate selective degeneration of glutamate-enriched in Alzheimer’s disease: Rationale and current status.
neurons in Alzheimer’s disease. Neuroscience, 38: Drugs, 74: 729–736.
571–577. Reisberg, B., Doody, R., Stoffler, A. et al. (2003).
Lynch, G. (1998). Memory and the brain: Unexpected Memantine in moderate-to-severe Alzheimer’s
chemistries and a new pharmacology. Neurobiology of disease. The New England Journal of Medicine, 348:
Learning and Memory, 70: 82–100. 1333–1341.
Mesulam, M., Shaw, P., Mash, D. and Weintraub, S. (2004). Tariot, P.N., Farlow, M.R., Grossberg, G.T. et al. (2004).
Cholinergic nucleus basalis tauopathy emerges early Memantine treatment in patients with moderate to
in the aging-MCI-AD continuum. Annals of Neurology, severe Alzheimer disease already receiving donepezil:
55: 815–828. A randomized controlled trial. JAMA, 291: 317–324.
Minger, S.L., Esiri, M.M., McDonald, B. et al. (2000). Tekin, S., Mega, M.S., Masterman, D.M. et al. (2001).
Cholinergic deficits contribute to behavioural dis- Orbitofrontal and anterior cingulate cortex neurofi-
turbance in patients with dementia. Neurology, 55: brillary tangle burden is associated with agitation in
1460–1467. Alzheimer disease. Annals of Neurology, 49: 355–361.
Neary, D., Snowden, J.S., Mann, D.M. et al. (1986). Terry, R.D., Masliah, E., Salmon, D.P. et al. (1991). Physical
Alzheimer’s disease: A correlative study. Journal of basis of cognitive alterations in Alzheimer’s disease:
Neurology, Neurosurgery and Psychiatry, 49: 229–237. Synapse loss is the major correlate of cognitive impair-
Perry, E.K., Haroutunian, V., Davis, K.L. et al. (1994). ment. Annals of Neurology, 30: 572–580.
Neocortical cholinergic activities differentiate Lewy Tsang, S.W.Y., Lai, M.K.P., Kirvell, S. et al. (2006). Impaired
body dementia from classical Alzheimers-disease. coupling of muscarinic M-1 receptors to G-proteins in
NeuroReport, 5: 747–749. the neocortex is associated with severity of dementia
Perry, E.K., Johnson, M., Kerwin, J.M. et al. (1992). in Alzheimer’s disease. Neurobiology of Aging, 27:
Convergent cholinergic activities in aging and 1216–1223.
Alzheimer’s disease. Neurobiology of Aging, 13: Turrini, P., Casu, M.A., Wong, T.P. et al. (2001). Cholinergic
393–400. nerve terminals establish classical synapses in the rat
Perry, E.K., Perry, R.H., Blessed, G. and Tomlinson, B.E. cerebral cortex: Synaptic pattern and age-related
(1978). Changes in brain cholinesterases in senile atrophy. Neuroscience, 105: 277–285.
dementia of Alzheimer type. Neuropathology and Wilkinson, D.G., Francis, P.T., Schwam, E. and Payne-
Applied Neurobiology, 4: 273–277. Parrish, J. (2004). Cholinesterase inhibitors used in
Perry, E.K., Smith, C.J., Court, J.A. and Perry, R.H. (1990). the treatment of Alzheimer’s disease: The relationship
Cholinergic nictotinic and muscarinic receptors in between pharmacological effects and clinical efficacy.
dementia of Alzheimer, Parkinson and Lewy body Drugs Aging, 21: 453–478.
48
The central role of Aβ amyloid and tau in the
pathogenesis of Alzheimer’s disease

CRAIG W. RITCHIE AND COLIN L. MASTERS

which case it may be a secondary ­phenomenon. Oxidative


48.1 INTRODUCTION damage may also be the result of Aβ or tau aggregation.
There is increasing evidence that sporadic AD is primarily
The details of the molecular neuropathology of Alzheimer’s the result of failure of clearance of Aβ over many decades. This
disease (AD), from the amyloid precursor protein (APP) is in contrast to the autosomal dominant inherited forms of
through to the production of amyloid Aβ, plaque formation, AD, in which pathogenic mutations in critical genes in the
neurodegeneration and to final neuronal death are discussed Aβ-biogenesis pathway cause an overproduction of Aβ from
in this chapter. This process, like Alexander’s Gordian the time of conception. These processes will be discussed in
knot, has been scrutinized over the last four decades with more detail later in the chapter.
the ongoing hope that a decisive therapeutic incision is not The biogenesis of Aβ from APP and putative mechanisms
too distant. Points in the biochemical pathway that may be for its neurotoxicity, together with genetic risk factors such
amenable to therapeutic intervention are highlighted. as apolipoprotein E (ApoE) that may interact with Aβ and
The theory that Aβ amyloid underlies the neurodegen- affect its clearance from the brain, is the focus of this chap-
erative changes in AD remains pre-eminent (Figure 48.1) ter. More detailed reviews on these subjects can be found
despite the emergence in recent years of alternative disease elsewhere (Masters et al., 2015).
pathways. It is clear that there are both proven and hypo-
thetical links between the alternate pathways (including
oxidative stress, altered innate immune reponses, elevation
48.2 Aβ AMYLOID ACCUMULATION
of cortisol and impaired axonal transport) and Aβ protein.
Integral to the pivotal role of Aβ and AD is the presumed
AND PLAQUE DEVELOPMENT
neurotoxicity of Aβ, especially in its soluble/diffusible oligo-
meric forms, while the polymerized fibrillar forms in the One of the two characteristic microscopic features of AD is
amyloid plaques may represent a toxic reservoir. the accumulation of Aβ amyloid in plaques of varying mor-
This Aβ peptide is the main constituent of the amyloid phology. These are extracellular or perivascular congophilic
plaque, one of the characteristic pathological features of deposits of aggregated Aβ with a high content of β-pleated
AD. It is also found in vessel walls as a congophilic amyloid sheet secondary structure. The amyloid plaque itself is the
angiopathy (Glenner and Wong, 1984; Masters et al., 1985). end result of a process of Aβ oligomerisation, fibril formation,
Neurofibrillary changes are also major features in the brains aggregation and precipitation occurring in several stages, with
of patients with AD. Tau protein, a microtubule-associated each stage potentially having a different impact on surround-
protein, is ubiquinated, phosphorylated and accumulates as ing neurones. Initially it was postulated that a soluble species
neurofibrillary tangles (NFT) and dystrophic neurites (Kosik of Aβ forms oligomers. Over time there developed a consen-
et al., 1986; Wood et al., 1986). The actual link between Aβ sus that it is the oligomeric form of Aβ that is the most syn-
accumulation and tau-associated NFT remains unknown, aptotoxic (Naylor et al., 2008; Shankar and Walsh, 2009) and
although NFT appear to be downstream of the events that neurotoxic (Mclean et al., 1999). Oligomers are formed from
surround Aβ accumulation in the AD brain. The hyperphos- monomeric Aβ through the formation of covalent dityrosine
phorylation of tau protein may occur after aggregation, in bonds or other post-translational changes that are relativeley
492
The central role of Aβ amyloid and tau in the pathogenesis of Alzheimer’s disease 493

Alzheimer’s disease

Neurodegeneration

NFT APC / ACA

Genetic
Aβ ApoE risk
factors
PS1,2

Environmental Pathogenetic
APP
risk mutations
factors

Figure 48.1 The amyloidocentric pathway that leads to Alzheimer’s disease proceeds from the proteolytic processing of
the amyloid β precursor protein (APP) into the Aβ amyloid. The amyloid forms visible plaques (amyloid plaque cores [APC];
amyloid congophilic angiopathy [ACA]). The relationship between Aβ and neurofibrillary tangle (NFT) formation remains
unclear. The mechanisms that underly the basic neurodegenerative changes in Alzheimer’s disease are also uncertain. This
pathway is subject to modulation by environmental and genetic factors at various points.

resistant to catabolism (Naylor et al., 2008). The formation the amyloid plaque therefore varies depending upon both
of oligomers is dependent upon the micro-environment in location and stage of development.
which Aβ is released from the cell membrane (Smith et al., Beyond a certain level, the number of amyloid plaques
2007). These oligomers may aggregate into protofibrillar at post-mortem does not correlate well with the severity
structures, which may first be seen as precipitates in diffuse of clinical disease (McKee et al., 1991; Morris et al., 1991;
amyloid plaques; this progresses with dystrophic neurite for- Terry et al., 1991; Berg et al., 1998). More recent studies
mation both within the neuropil and around dense crystalloid using positron emission tomography (PET)-Aβ molecular
precipitates of amyloid cores. Following this, an intermediate imaging show how the Aβ burden correlates with cognitive
stage is reached, where the plaque increases in complexity impairment and rates of decline (Villemagne et al., 2013,
before a final stage of a non-neuritic ‘burned out’ or ‘end-stage Lim et al., 2015). At high levels, the rates of Aβ increase may
plaque’ is reached. reach a plateau, explained in part by a process of growth
In autosomal dominantly inherited AD, processing of and resolution of plaques (Hyman and Tanzi, 1992). In
APP creates a high ratio of Aβ42 (‘long’ Aβ of 42 amino concert with traditional explanations of the disconnect
acids) to Aβ40 (‘short’ Aβ of 40 amino acids). These pro- between plaque number and disease severity, there is now
cesses are discussed later in this chapter. The more insoluble a gathering consensus that the soluble/diffusible Aβ42 load
long Aβ is the primary constituent of the amyloid plaque. may be more closely related to clinical severity (McLean
However, immunocytochemical techniques have demon- et al., 1999), whether or not the Aβ42 exists as an oligomer
strated that diffuse plaques contain not only Aβ42 but also or complexed to other proteins (Lorenzo and Yanker, 1994;
Aβ40 and, in a small proportion of plaques (most of which Howlett et al., 1995). A major argument raised by opponents
have a dense amyloid core), Aβ(16–40/42) (the p3 fragment) of the amyloid theory is that individuals can have numerous
(Dickson, 1997). Perivascular amyloid identified in the plaques but no clinical cognitive impairment. As we learn
congophilic angiopathies predominantly consists of Aβ40 more from PET-Aβ and cerebrospinal fluid-Aβ (CSF-Aβ)
(Suzuki et al., 1994; Barelli et al., 1997). measurements, this argument becomes less convincing.
Plaque morphology also varies as a function of topo- Aβ may mediate clinical symptomatology predominantly
graphic location. Smaller granular deposits are seen in through synaptotoxicity and neurotoxicity, which precedes
the deep grey matter nuclei, and linear streaks occur in frank neurodegeneration. Finally, Aβ accumulation and the
the molecular layer of the cerebellum. The morphology of plaques are components within a very complex biological
494 Dementia

system that relies on numerous other pathological processes APP gene is located on chromosome 21 (Kang et al., 1987).
before AD develops clinically. These processes are mediated The pathogenic APP mutations account for less than 5% of
by other biological, genetic and environmental risk factors. all cases of AD inherited in an autosomal dominant manner.
It may be that to trigger AD from the physiological produc- The precise physiological function of APP is unknown,
tion of monomeric Aβ requires the ‘perfect storm’ combin- though no doubt is complex and possibly related to synaptic
ing several biological (e.g. neuroinflammation, oxidated plasticity, repair and regeneration. The structural domains
stress and genetic vulnerability) and environmental (e.g. suggest that it may have a role in cell–cell (synapse) or cell–
cerebrovascular) risk factors – all of which are more likely matrix interactions (neurite stabilization). There is evidence
to accumulate and co-occur with advancing age. to suggest that APP is affected by metal ion binding, calcium
levels and heparin binding. APP has also been associated
with cell proliferation and neurite outgrowth in response to
48.3 GENETIC EVIDENCE FOR THE nerve growth factor (Milward et al., 1992; Small et al., 1994;
ROLE OF Aβ IN AD Yankner, 1996). Other in vitro experiments have suggested
that a deficiency of APP renders cells more susceptible to a
variety of neurotoxic insults.
The most convincing support for the central role of Aβ and
APP in the pathogenesis of AD is that most of the known
fully penetrant autosomal dominant gene mutations that 48.5 APP PROCESSING BY α-, β- AND
cause early-onset AD lead to an increase of Aβ42 produc- γ-SECRETASES
tion (Bateman et al., 2006; Mawuenyega et al., 2010) (see
Table 48.1). Mutations in the APP gene itself lead to the
development of aberrant APP that is preferentially pro- APP exists in three major isoforms of 695, 751 and 770 amino
cessed to produce an increase in the Aβ 42:40 ratio. More acids. The 695-amino acid isoform (APP695) is predomi-
recently, mutations near the β-secretase (BACE1) site have nantly expressed in neurones (Kang et al., 1987; Tanzi et al.,
been identified, which are protective (Jonsson et al., 2012). 1987). The APP751 and APP770 isoforms are predominantly
All causative APP mutations identified to date occur in expressed in peripheral tissues and astrocytes (Golde et al.,
proximity to the Aβ domain of the protein. Similarly, mul- 1990; Kang and Müller-Hill, 1990). APP is synthesised in the
tiple mutations in the two presenilin (PS) genes have a dra- endoplasmic reticulum and is then transported through the
matic effect on the Aβ 42:40 ratio (Szaruga et al., 2015). Golgi apparatus to the cell surface. In its transmembrane ori-
entation (as shown in Figure 48.2) it undergoes proteolytic
cleavage to produce Aβ. The half-life of APP is short and is
processed by at least two pathways (α- and β-/γ-secretase
48.4 AMYLOID PRECURSOR PROTEIN activities). In peripheral cells, α-secretase activity predomi-
nates, cleaving the Aβ domain of APP at position 16/17. This
The APP is a transmembrane protein that is found in most leads to the production of soluble ectodomain protein sAPPα
cell types including neuronal and glial cells (Figure 48.2). It (Evin et al., 1994). The soluble ectodomains of APP (both
is especially enriched in the alpha-granule of platelets. The sAPPα and sAPPβ) do not aggregate and do not participate

Table 48.1 Pathogenic mutations that cause Alzheimer’s disease

Mutations Mechanism of action Effect


APP gene dosage or aberrant regulation
Down’s Syndrome (trisomy 21) Upregulation of APP gene promoter Increased total Aβ
up to fivefold to sixfold
Pathogenic APP gene mutations
APP codons:
670/671 (‘Swedish’) Increase β-secretase cleavage Excess total Aβ production
692 and 693 (‘Dutch and Flemish’) Decrease α-secretase activity? Resultant increase in β- and γ-activity
with consequent increased total Aβ
715 (‘French’) Affects γ-secretase activity Increased amounts of Aβ p3 fragment
717 (‘London’) Altered γ-secretase activity Increased ratio of Aβ42:40
723 (‘Australian’) Affects γ-secretase activity Increased ratio of Aβ42:40

Pathogenic PS1, 2 gene mutations


More than 70 point or misense mutations Direct or indirect alteration of Increased ratio of Aβ42:40
and exon deletion γ-secretase
Abbreviation: APP, amyloid precursor protein.
The central role of Aβ amyloid and tau in the pathogenesis of Alzheimer’s disease 495

Poly-T GPD HBD-2 CS-GAG


NPXY
APP

P CHO
Clathrin/Fe65
binding
KPI OX-2 Exon 15
Aβ G0 binding

HBD-1 ZnBD Membrane


(a) β α γ anchor
Secretase sites

β-Secretase α-Secretase γ-Secretase

NH2 T E E I S E V K M D A E F R H D S G Y E V H H Q K L V F F A E D V G S N K G A I I G L M V G G V V I A T V I V I T L V ML K K K C O OH
1 10 20 30 40
I
G
Mutation NL GC MVF P

APP770 Codon 670/671 692 693 715 717 723


Swedish Flemish Dutch French London Australian
(b) 716
Florida

Figure 48.2 (a) The structural domains of APP are schematically shown. Within the large extracellular ectodomain
there are heparin-binding sites (HBD-1,2), metal-binding sites (ZnBD), growth-promoting domains (GPD), carbohydrate-­
attachment sites (CHO; CS-GAG), and alternatively spliced exons (KPI, OX2, Exon 15). The shorter cytoplasmic domain
has interacting motifs for G0 protein, clathrin (NPXY) and the Fe65 family of proteins. (b) The Aβ (juxtatransmembrane
and transmembrane) domains are shown in more detail. The β-, α- and γ- secretase sites are shown, with the major
­cleavage sites indicated by unbroken arrows. The critical pathogenic mutations are shown to be clustered near the
­secretase sites (see also Table 48.1).

in plaque formation. α-Secretase activity leaves in the mem- are associated with preferential activity of the β-secretase
brane a small carboxyl-terminal (C-terminal) fragment that pathway (Green et al., 2006) as well as hippocampal atro-
undergoes further processing. The α-secretase activity may phy and increased amyloid plaque development (Butters
be sequence non-specific and act on several other trans- et al., 2008) in patients with depression. The pharma-
membrane proteins. Furthermore, recent work has identi- cological inhibition of the enzyme 11β-hydroxysteroid
fied a disintegrin and metalloprotease (ADAM), as being a dehydrogenase (HSD) that converts inactive cortisone
potent effector of α-secretase activity (Lammich et al., 1999). to cortisol is attracting interest as a potential therapeutic
Increase of ADAM-10 expression and activity may theoreti- avenue (MacLullich et al., 2012). These biological observa-
cally be effective as a treatment for AD. tions provide a basis for the epidemiological and clinical
Aβ, therefore, exists in several forms depending on links between depression/stress and Alzheimer’s demen-
the site of cleavage by α-, β- or γ-secretases. The length tia (Jorm, 2001; Huang et al., 2009).
of the Aβ fragment varies depending on the site of cleav- Determination of the structure of γ-secretase could have
age leading to a product between 38 and 43 amino acids important therapeutic implications (De Strooper et al.,
in length. The amino-terminal (N-terminal) 28 residues 1999; Struhl and Greenwald, 1999; Wolfe et al., 1999; Ye
are from the extracellular portion of APP, and the 11–15 et al., 1999; Iwatsubo 2004). The macromolecular complex
C-terminal residues are derived from the transmembrane that constitutes γ-secretase activity is >400 kDa in mass
domain. It is the Aβ42 which is considered to be the more and contains at least three other molecules: Aph1, Pen2 and
insoluble and toxic moiety and is a product of γ-secretase nicastrin (Iwatsubo, 2004). Active development of inhibi-
cleavage of the APP molecule. β- and γ-secretase activity tors of γ-secretase has progressed at a radical pace, but the
involves cleavage of APP at sites corresponding to posi- results of clinical trials have been disappointing. Further
tions 1 (β-secretase) and 40 or 42 (γ-secretase) of the Aβ work on dosage and pharmacodynamics are required
fragment, releasing the insoluble amyloidogenic peptide to achieve more suitable inhibition and modulation of
Aβ(1–40/42) commonly referred to as Aβ42. It is intrigu- γ-secretase activity (Bai et al., 2015; De Strooper and Chávez
ing to note that in animal models, elevated levels of cortisol Gutiérrez, 2015).
496 Dementia

Another factor that influences the cleavage pathway may


48.6 APP MUTATIONS AND Aβ be the release of APP mediated by protein kinase C (PKC),
PRODUCTION which leads to a predominance of the α-secretase path-
way (Caporaso et al., 1992; Gillespie et al., 1992; Sinha and
The various APP mutations inevitably produce increased Lieberburg, 1992). Of interest, PKC-mediated α-secretase
ratios of Aβ42 through a variety of mechanisms (Table 48.1). activity may be induced by neurotransmitters and other
The mutations at codons 670/671 (the ‘Swedish’ mutation), first-messenger ligands (Buxbaum et al., 1992; Lahiri
increase β-secretase cleavage and lead to an overall excess et al., 1992; Nitsch et al., 1992). If these first- and second-­
amyloid production (Cai et al., 1993). Mutations at codon messenger pathways prove to directly influence the pro-
717 (the ‘London’ mutation), 715 (the ‘French’ mutation), 716 duction of Aβ42, then these systems could provide another
(the ‘Florida’ mutation) and 723 (the ‘Australian’ mutation) target for pharmacological intervention (Roh et al., 2012; Ju
act at the γ-secretase site, increasing the ratio of Aβ42:40 et al., 2013)
(Suzuki et al., 1994; Brooks et al., 1995; Eckman et al., 1997;
Ancolio et al., 1999; Kwok et al., 2000). Mutations at 692
and 693 (the ‘Dutch’ and ‘Flemish’ mutations) are either
associated with α-secretase activity, presumably leading to a
48.7 PRESENILINS 1 AND 2
decrease in this pathway with a resultant increase in β- and
γ-activity (Haass et al., 1994) or yield a mutant Aβ peptide The presenilin 1 (PS1) gene is located on chromosome 14
with enhanced toxicity or propensity to aggregation. In and PS2 on chromosome 1. In the brain, PS1 is found in
Down’s syndrome (trisomy 21), where there is an extra copy both neurones and glia. It has a multipass transmembrane
of the APP gene, there is a similar increase in the overall orientation (Figure 48.3). PS2 expression in the pancreas
levels of Aβ42 (Teller et al., 1996). and muscle exceeds other sites including the brain (Rogaev,
It is necessary to elucidate the mechanisms that regu- 1998). The PS molecules undergo cleavage by unidentified
late the activity of the secretase enzymes involved in APP mechanisms to yield N- and C-terminal fragments.
metabolism, as a greater understanding of these processes A link between PS mutations and Aβ production has
will aid the development of therapeutic interventions. One been confirmed as an increased ratio of Aβ42:40 is noted
factor that may predict which form of cleavage is utilized is in transfected cell lines and transgenic mice expressing
cell type, with glial cells and neurones (both differentiated mutant forms of PS1 (Borchelt et al., 1996; Citron et al.,
and undifferentiated) producing different isoforms of APP 1996; Duff et al., 1996; Lemere et al., 1996). Patients and
(Haass et al., 1991; Baskin et al., 1992; Hung et al., 1992). at-risk individuals with PS mutations have increased ratios

Exon 10

Exon 3 Alternative
(caspase)
cleavage site

Exon 9
Exon 12
Exon 4
Exon 8

Exon 6 Cytosol
`Presenilinase´
cleavage site
Lumen
Exon 7
Exon 11

PS-I residue change due to misense mutation


PS-I exon deletion mutations: ΔE4
ΔE9
Exon 5 ΔE10

Figure 48.3 A model of the presenilin 1 molecule. The multiple mutations that cause early-onset Alzheimer’s disease are
seen to cluster in the transmembrane domains and in proximity to the normal ‘presenilinase’ cleavage site. The cytosolic
loop between transmembrane domains six and seven may play a critical role in γ-secretase activity.
The central role of Aβ amyloid and tau in the pathogenesis of Alzheimer’s disease 497

of Aβ42:40 (Scheuner et al., 1996). PS1 may have an anti- observation that a free-radical-based process, mediated by
apoptotic activity (Roperch et al., 1998) and mutations in Aβ, leads to neuronal damage and that this process, in vitro,
PS1 may sensitise neuronal cells to apoptosis by disruption can be inhibited by antioxidants. The use of antioxidants
of intracellular calcium levels (Keller et al., 1998). in the treatment of AD has been investigated clinically in
In summary, most of the identified genotypes in auto- recent years and some clinical efficacy has been shown with
somal dominant AD have, as their common denominator, vitamin E (Sano et al., 1997) though a summation of the
an increase in the production of Aβ42. This observation evidence to date has concluded that this intervention may
suggests a direct causal relationship between Aβ42 and the well be too late in advanced disease or early dementia (Ames
development of AD. and Ritchie, 2007). Moreover, preliminary trials with com-
pounds that target the redox-active metal-binding sites on
Aβ have yielded encouraging results (Ritchie et al., 2003;
Lannfelt et al., 2008).
48.8 Aβ AND NEUROTOXICITY

Despite the above evidence that an increase in the propor-


tion of Aβ42 is a common end point for all the autosomal 48.9 Aβ INTERACTION WITH
dominant mutations that lead to AD, the exact mechanism APOLIPOPROTEIN E AND OTHER
of underlying Aβ42 and neuronal degeneration remains RISK FACTOR GENES
unclear (Table 48.2).
Aβ has been shown to be directly neurotoxic to hippo- 48.9.1 APOLIPOPROTEIN E
campal neurones that are particularly sensitive to oxidative
stress and are affected first in the spread of AD throughout ApoE isoforms represent a major genetic susceptibility fac-
the cerebral cortex. While some authors have been unable tor for sporadic AD, affecting all people over the age of 60
to demonstrate neurotoxicity of Aβ when it exists in an years. It is the strongest of a multiple set of genetic risk fac-
immature, non-fibrillar, amorphous aggregate (Lorenzo tors that have the potential to modify any putative environ-
and Yanker, 1994), there is now an emerging consensus that mental risk factor (Saunders et al., 1993; Huang et al., 2004).
the smaller oligomeric aggregates of Aβ, in association with The ApoE gene, located on chromosome 19, was the first
factors such as metal ions, constitute the principal forms polymorphic gene to be associated with a complex disease
of the toxic Aβ species. Aβ can potentiate the neuronal using positional cloning strategies. The ApoE gene products
insult of excitatory amino acids (Koh et al., 1990), oxida- vary depending upon the inherited pair of polymorphic
tive stress (Lockhart et al., 1994) and glucose deprivation alleles. There are three allelic varieties ε2, ε3 and ε4. ApoE is
(Copani et al., 1991). This may explain why the cumulative a glycoprotein of 299 amino acids that is a normal constitu-
exposure of the ageing brain to other risk factors renders it ent of plasma and CSF lipoprotein particles. The physiologi-
more vulnerable to the toxic effects of Aβ. Aβ has also been cal function of the protein is to mediate cholesterol uptake,
shown in vitro to cause plasma membrane lipid peroxida- storage, transport and metabolism. There is a binding site
tion, impairment of ion motive ATPases, glutamate uptake for Aβ present near the C-terminus of the ApoE molecule
and uncoupling of γ-protein-linked receptors (Behl et al., (Wisniewski et al., 1993). ApoE is a component of the very-
1994; Butterfield et al., 1994; Hensley et al., 1994; Mark et al., low-density lipoprotein (VLDL) and high-density lipo-
1996). These pathological processes may contribute to a loss protein (HDL) complexes involved in cellular uptake and
of intracellular calcium homeostasis that has been reported metabolism of cholesterol (Mahley, 1988). ApoE is normally
in cultured neurones (Mattson et al., 1993). Aβ, when intro- upregulated and released from astrocytes following neuro-
duced into neuronal cultures, leads to a gradual increase in nal injury and not only affects lipid metabolism following
intracellular calcium, though this activity is not affected by this trauma but may also complex with VLDL to increase
the addition of either calcium channel blockers or chelat- neurite outgrowth and synaptogenesis. However, the dif-
ing agents (Lorenzo and Yanker, 1994; Whitson and Appel, ferent ApoE genotypes differentially affect this process.
1995). Aβ may also impair redox activity in mitochon- Individuals carrying the ε4 allele have a decreased capac-
dria, leading to the production of free radicals (Shearman ity for compensatory neurite outgrowth and synaptogenesis
et al., 1994). Cellular damage mediated by Aβ is inhibited following neuronal injury (Poirier, 1994).
by vitamin E (Behl et al., 1992). This supports the above It has also been demonstrated that individuals with
at least one ε4 allele develop greater numbers of neuritic
Table 48.2 Aβ toxicity-proposed mechanisms plaques (Olichney et al., 1996) irrespective of whether or not
they have clinical AD (Rebeck et al., 1993; Schmechel et al.,
●● Generation of reactive oxygen species, metal
1993; Polvikoski et al., 1995). ApoE ε4 also complexes with
mediated
Aβ, whereas ApoE ε3 does not have the same affinity, thus
●● Physical disruption of cellular membrane
promoting protofibril formation, which, as has been alluded
●● Receptor-mediated uptake and apoptotic signalling
to already, may be the immediate neurotoxic product in
●● Disturbance of intracellular calcium homeostasis
AD. A model therefore exists that outlines the interaction
498 Dementia

between Aβ and ApoE, which explains why the latter exists However, there may only be a marginal increase in diag-
as one of many contributing genetic risk factors to the devel- nostic accuracy (American College of Medical Genetics/
opment of AD. At the very least, it links the metabolism of American Society of Human Genetics Working Group on
cholesterol into the Aβ-mediated pathway of AD causation. Apolipoprotein E and Alzheimer’s Disease, 1995; National
Not only the genotype that an individual carries is rele- Institute on Aging/Alzheimer’s Association Working
vant as a risk factor for AD but also variations in ApoE levels Group, 1996). In isolation, knowledge of the ApoE genotype
may also be an important factor in predicting disease onset. of any given individual lacks the sensitivity, specificity or
The promoter of ApoE that regulates the synthesis of the lipo- positive predictive value to be useful as a screening tool.
protein is polymorphic. The allelic variants at −491 (Bullido
et al., 1998), −427 (Artiga et al., 1998) and −219 (Lambert 48.9.2 OTHER GENETIC RISK FACTORS
et al., 1998) of the ApoE promoter region, when homozygous, FOR AD
may be associated with increased risk of AD, and a combina-
tion of the adverse alleles of the promoter and coding regions The AlzGene initiative has provided major insights into
confer even greater risk of developing AD (Artiga et al., 1998). genetic risks for AD. The project allows the knowledge
Clinically, possession of the ε4 allele increases the risk of gained from numerous studies and meta-analysis to pro-
developing AD at any particular age. In individuals who are vide dynamic conclusions on risk across a range of poten-
ε4 heterozygotes, there is an approximate doubling of risk tially relevant genes. From this, gene polymorphisms can be
and for those who are homozygous for ε4 the risk is increased ranked by their strength of association with AD (be that a
by a factor of between six and eight (National Institute on risk or protective) or by the p-value. It is the latter method
Aging/Alzheimer’s Association Working Group, 1996). of ranking that is currently used. The current ‘top 10’
Possession of an ε4 allele may lead to an earlier age of onset genes are summarized below (Table 48.3), which has been
(Blacker et al., 1997; Burlinson et al., 1998) and there may be extracted from the AlzGene website: http://www.alzgene
an even greater risk of developing AD in females who carry .org/TopResults.asp.
the ε4 allele (Poirier et al., 1993; Payami et al., 1994; Duara The relationship of the ‘top 5’ of these genes to Aβ and tau
et al., 1996). There are, however, conflicting reports regard- pathology is summarized. ApoE and AD has been dealt with
ing the rate of decline associated with different genotypes. in detail above. The bridging integrator 1 (BIN1) gene, also
More recent data obtained from longitudinal cohorts and known as amphiphysin 2, is primarily associated with AD
PET-Aβ molecular imaging clearly demonstrate the effect pathology through its role in tau modulation. It also has a role
of ApoE in the preclinical, prodromal and clinical phases in calcium homeostasis, apoptosis, endocytosis/­trafficking
of Aβ (Lim et al., 2015). The ApoE ε2 allele may confer pro- and inflammation. Epigenetic modifications are important
tection against the development of AD (Corder et al., 1994; for AD pathogenesis, and there are data that suggest the pos-
Myers et al., 1996). sible DNA methylation of the BIN1 promoter. Finally, given
The ε4 allele also confers additional risk of developing the potential contributions of BIN1 to AD pathogenesis, tar-
AD in those patients who have suffered strokes (Slooter geting BIN1 might present novel opportunities for AD ther-
et al., 1997), though other authors, in smaller studies, have apy (for a further review see Tan et al., 2013).
failed to replicate this finding (Burlinson et al., 1998). Other Clusterin or apolipoprotein J has also been associated
cerebral insults such as head injuries have been associated with AD risk for many years (see Li et al., 2014). Clusterin
with increased Aβ deposition in patients who carry an ε4 is a chaperone protein and can inhibit the aggregation of
allele (Nicoll et al., 1995). Determining the ApoE status of a Aβ. However, it has also been shown to promote intra-
patient may be of value in aiding the diagnosis of AD where cellular pathways of Aβ metabolism that are neurotoxic
there is already a high clinical suspicion of the condition. as well as promote the formation of neurotoxic, soluble

Table 48.3 Top 10 genes associated with increased risk of Alzheimer’s dementia from AlzGene Project

Rank Gene Odds ratio (CI) p-value


1 Apolipoprotein E 3.68 (3.3–4.12) <1−50
2 Bridging integrator (BIN) 1 1.16 (1.13–1.2) <1.6−26
3 Clusterin (Apolipoprotein J) 0.88 (0.86–0.9) <3.4−23
4 ATP-binding cassette sub-family A member (ABCA) 7 1.23 (1.18–1.28) <8.2−22
5 Complement component (3b/4b) receptor (CR1) 1.17 (1.14–1.21) <4.7−21
6 Phosphatidylinositol-binding clathrin assembly protein (PICALM) 0.88 (0.86–0.9) <2.8−20
7 Membrane-spanning 4-domains, subfamily A, member 6A (MS4A6A) 0.9 (0.88–0.93) <1.8−11
8 CD33 0.89 (0.86–0.93) <2.1−10
9 Membrane-spanning 4-domains, subfamily A, member 4E (MS4A4E) 1.08 (1.05–1.11) <9.5−10
10 CD2-associated protein (CD2AP) 1.12 (1.08–1.16) <2.8−9
Abbreviation: CI, confidence interval.
The central role of Aβ amyloid and tau in the pathogenesis of Alzheimer’s disease 499

Aβ species. To this end the impact that clusterin, and Several investigators have now shown that, like ApoE,
genetic and epigentic variants therein has on AD risk different LRP1 genotypes are associated with the develop-
remains uncertain with the biology of the protein to still ment of AD (Kang et al., 1997; Hollenbach et al., 1998),
be full characterized. ATP-binding cassette sub-family mediated possibly through an increase in Aβ burden. α2M
A member 7(ABCA7) is a member of the superfamily genotypes, unlike ApoE genotypes, have seemingly no
of ATP-binding cassette (ABC) transporters. These pro- effect on the age of onset in AD (Liao et al., 1998). The odds
teins transport various molecules across extracellular and ratio for AD associated with the γ/γ α2M genotype was
intracellular membranes. The function of this protein is shown to be 1.77 (1.16–2.70, p < 0.01) and in combination
not yet known; however, the expression pattern suggests with ApoE ε4 was 9.68 (3.91–24.0, p < 0.001). In the same
a role in lipid homeostasis in cells of the immune system study, there was no increased risk of AD demonstrated in
and elsewhere (Reitz et al., 2013). With relevance to AD, heterozygotic individuals with γ\γ genotype, and stratifi-
it has been demonstrated that ABCA7 is involved in APP cation of the sample based on the presence or absence of the
transport through the neuronal cell membrane (Chan et ApoE ε4 allele demonstrated a similar over-­representation
al., 2008), as well as being involved in the phagocytosis of the γ/γ genotype in both strata (Liao et al., 1998). This
by macrophages of apoptotic cells (Tanaka et al., 2011). finding was demonstrated in a population-based study.
Complement receptor 1 (CR1) is a gene involved in the This initial work suggests that the α2M γ/γ genotype may
complement cascade that has been linked to AD pathol- increase the risk of developing AD, that this risk is inde-
ogy. Its role is as a receptor protein for complement frag- pendent of the effects of ApoE ε4, and that the risks are
ments C3b and C4b especially in the circulatory system. additive and substantial. The studies to date require fur-
Activated complement is involved centrally in activation ther replication and the effect requires demonstration in
and recruitment of microglia around fibrillar amyloid different populations in a similar manner to studies defin-
plaques (Eikelenboom and Veerhuis, 1996), a process that ing the risks associated with ApoE ε4. If future studies do
is further enhanced by Aβ (Haga et al., 1993). The cyto- conclude that α2M is an independent risk factor for AD,
lytic effects of complement cascade in association with then predictive testing considering both genes (and also
Aβ have been recognized for over two decades (Rogers et including genotyping of the ApoE promoter regions) could
al., 1992), but the complex genetic association shown with be developed with much higher positive predictive values
ApoE status (Thambisetty et al., 2013), evidence of meta- than those demonstrated with ApoE testing alone. Whole
bolic alterations associated with AD (Darborg et al., 2012) genome search strategies (Blacker et al., 2003; Ertekin-
and evidence that CR1 inhibition can reduce the classical Taner et al., 2004) have yielded other ‘hot spots’ of inter-
complement cascade (Davis et al., 2008) has stimulated est, the most intriguing of which is that on chromosome
further interest in this area from both a mechanistic and 10, close to the insulin-degrading enzyme (IDE) locus.
therapeutic perspective. Attempts to identify IDE as a risk factor have so far failed,
but it remains an ­attractive candidate for its ­demonstrated
role in ­promoting the clearance of Aβ from the brain
α2-MACROGLOBULIN (α2M),
48.9.3  (Farris et al., 2004).
LOW-DENSITY LIPOPROTEIN
RECEPTOR-RELATED PROTEIN
(LRP1) AND INSULIN-DEGRADING
ENZYME 48.10 LINKING Aβ PRODUCTION AND
Over and above the genes listed above, there is an ongo-
AGGREGATION TO THE OTHER
ing interest in other genes and gene products related to Aβ OBSERVED PATHOLOGIES IN AD
metabolism. While the ApoE gene was the first polymor-
phic marker to be identified as a genetic modifier of risk 48.10.1 Aβ AND TAU
for AD, other candidate genetic loci are being investigated, HYPERPHOSPHORYLATION
including α2M, a protease inhibitor found in association
with amyloid plaques in AD. The gene for α2M is located on The two most well-defined pathological processes in AD
chromosome 12, binds Aβ with high affinity and may influ- are the aggregation of tau protein and subsequent destabi-
ence fibril formation and the toxicity of Aβ in vitro (Hughes lization of the cytoskeletal protein tubulin, and the aggre-
et al., 1998). The activity of α2M has also been associated gation of Aβ to form amyloid plaques. While traditionally
with LRP1. Both α2M and LRP1 are upregulated following there has been debate about which process is upstream from
neuronal injury (Lopes et al., 1994). Furthermore, α2M is the other and hence more ‘important’, less prosaic think-
thought to mediate Aβ degradation via endocytosis through ing would lead one to develop hypotheses about how these
LRP1 (Narita et al., 1997). As ApoE and APP are also ligands two important lesions are linked. One such hypothesis rec-
of LRP1, it is postulated that different isoforms of ApoE and ognizes the importance of the protein glycogen synthase
α2M may interact differentially and competitively for this kinase 3 (GSK3) having a pivotal role in the pathological
system, affecting the clearance of Aβ. events observed in AD (Hooper et al., 2008).
500 Dementia

GSK3 has a multiplicity of functions including roles in 2009). When activated, glial cells overproduce the excito-
maintaining microtubule stability, apoptosis and gene tran- toxic neurotransmitter glutamate, and pro-inflammatory
scription. Upregulation of the protein is associated with cytokines also influence concentrations of zinc (Vasto et al.,
induction of tau hyperphosphorylation (Lovestone et al., 2007). While the glutamate is directly neurotoxic through
1994) an effect inhibited by insulin (Lesort et al., 1999). The the N-methyl-d-aspartate (NMDA) receptor, the height-
role of insulin to not only inhibit GSK3 but also promote the ened levels of zinc may drive the oligomerization of Aβ lead-
expression of IDE, which is an Aβ protease as well as a pro- ing to both acute (synaptotoxic) delirium-generating events
moter of APP processing through the α-secretase pathway as well as – over time – neurotoxicity and neurodegenera-
has led to the proposition that AD could be labelled ‘Type 3 tion leading to more entrenched and irreversible symptoms.
Diabetes’ (de la Monte and Wands, 2008). This observation may explain why PBT2 was shown to have
Moreover, the GSK3α isoform has been shown to regu- an acute effect in Phase 2 trials despite being developed pre-
late APP processing leading to increased Aβ production dominantly as a disease-modifying agent (Lannfelt et al.,
(Phiel et al., 2003), which in turn has been shown in neu- 2008). While the above highlights the negative consequencs
rones to further increase GSK3β expression. Both the GSK3 of glial activation, it is recognized that phagocytosis by glial
isoforms are associated with tau hyperphosphorylation. cells of Aβ at earlier stages of the disease process is benefi-
Althouth there are other theories associating tau and Aβ cial. Therefore there is a both positive (early) and negative
protein in the genesis of the disease, GSK3 overexpression (later) effect of glial activation vis-a-vis the progression of
and its interaction with insulin would appear likely linking AD. This topic has been reviewed elsewhere (Gambuzza et
processes with much consistent evidence supporting this al., 2014) wherein the role of Toll-like receptors (proteins
theory. intimately involved in the innate immune system) can be
activated to either induce Aβ uptake or (in later disease)
neuroinflammation.
48.10.2 Aβ AND MITOCHONDRIAL
DYSFUNCTION
Mitochondria are the energy source of the cell and any dis- 48.11 A PHYSIOLOGICAL ROLE FOR Aβ
ruption of mitochondrial function will necessarily lead to
impaired neuronal function, defences, repair and integrity. It is debated whether an invariably toxic protein or
Again, the question is raised as to whether Aβ overproduc- single process explains the pathology of AD – rather it
tion leads to mitochondrial damage or whether the latter may be the interaction of many (normally) physiologi-
precedes the former. cal processes whose interactions associated with genetic
An energy-deficient neurone will tend to favour less- and environmnetal risks associated with ageing become
energy-dependent processes, where an alternate exists. In pathological. In this light, Aβ may well perform an
this regard it has been demonstrated that BACE increases important physiological role to act as an antioxidant and
its function in energy deprived neurones (Vassar et al., a chaperone elevating metals towards biological inactiv-
2009). Therefore – upstream mitochondrial damage may ity embedded within the β-pleated sheet of the plaque.
lead to increased Aβ42 production. Downstream from
When the system is in homeostatic equilibrium between
this though – mitochondrial membranes and multiple
trace metal elevation, Aβ genesis and aggregation – there
mitochondrial processes are damaged by intracellular Aβ is little if any synaptotoxicty, but when multiple systems
(Reddy and Beal, 2008) and the C-truncated fragment of start to fail under the influence of genetic vulnerability
APP can lead to further mitochondrial dysfunction through and in the context of ageing mediating accumulation of
disruption of translocase of outer m
­ itochondrial membrane risks, the normally benign systems tip into a pathological,
40 (TOMM40). It is also worth noting that the hyperphos- malignant cascade that evolution has not found a way (or
phorylation of tau protein is more likley to occur in energy- a need) to switch off.
deficient cells (Rhein and Eckert, 2007). Accordingly, to intervene beyond what is possible from a
preventative perspective, one must not throw the baby out
48.10.3 Aβ AND NEUROINFLAMMATION with the bathwater; interventions will need to be subtle and
no doubt used in combinations with other interventions
Finally, there has been a long-held hypothesis that glial acti- affecting other parts of the process.
vation is central to the pathology seen in AD (Meda et al.,
2001). Glial cells activate in response to inflammatory sig-
nalling from both peripheral and central sources (including
the amyloid plaque via process outlined above regarding the 48.12 INTERVENTIONS
role of complement). The response of glia to systemic inflam-
mation may explain the genesis of delirium in patients with Being able to determine the risk that a given individual
systemic infections and the apparent acceleration of decline has of developing Alzheimer’s dementia late in life has
in Alzheimer’s dementia as a result of delirium (Fong et al., important clinical implications, as risk modification
The central role of Aβ amyloid and tau in the pathogenesis of Alzheimer’s disease 501

strategies are most likely to be of greatest benefit when REFERENCES


disease processes are nascent or indolent. As proven
disease-modifying treatments for AD are still in early American College of Medical Genetics/American
­development, another benefit of defining a population Society of Human Genetics Working Group on ApoE
with AD but without dementia is to be able to define a and Alzheimer disease (1995). Statement on use of
cohort for ­clinical trials of drugs or biological agents with Apolipoprotein E testing for Alzheimer’s disease.
a ­postulated preventative or significant disease-modify- Journal of the American Medical Association,
ing action. 274: 1627–1629.
Current therapeutic interventions for AD operate far Ames, D. and Ritchie, C. (2007). Antioxidants and
downstream from the postulated initial toxicity of Aβ42. Alzheimer’s disease: Time to stop feeding vitamin E
Their efficacy is dependent upon augmenting the activ- to dementia patients? International Psychogeriatrics,
ity of surviving cholinergic neurones; the cholinesterase 19 (1): 1–8.
inhibitors (donepezil, galantamine and rivastigmine) Ancolio, K., Dumanchin, C., Barelli, H. et al. (1999).
have shown only modest effect on clinical progression of Unusual phenotypic alteration of β-amyloid precursor
AD and have no demonstrable disease-modifying effect protein maturation (βAPP) by a new Val-715 to met
(Ritchie et al., 2004; see Chapter 52). To prevent neuronal βAPP-770 mutation responsible for probable early-
damage and clinical impairment, effective therapies must onset Alzheimer’s disease. Proceedings of the National
either affect the pathological levels of production of Aβ42 Academy of Sciences of the United States of America,
from APP or mediate its toxicity or clearance. Therapeutic 96: 4119–4124.
paradigms may therefore (and this list is not exhaustive) Artiga, M.J., Bullido, M.J., Frank, A. et al. (1998). Risk for
target inhibition of γ-secretase, or BACE1, or upregulate Alzheimer’s disease correlates with transcriptional
the α-secretase pathway; influence the phosphorylation activity of the ApoE gene. Human Molecular Genetics,
state of the APP molecule; interdict metal–ions interac- 7: 1887–1892.
tion with Aβ and hence prevent oligomerization; scav- Bai, X.C., Yan, C., Yang, G. et al. (2015). An atomic
enge free radicals; affect the postulated overactivation ­structure of human [γ]-secretase. Nature,
of microglial cells in response to Aβ deposition and sys- 525: 212–217.
temic inflammatory signals; inhibit GSK3; stabilize or Barelli, H., Lebeau, A., Vizzavona, J. et al. (1997).
protect TOMM40; improve insulin sensitivity; protect α-7 Characterization of new polyclonal antibodies specific
nicotinic receptors and do so as a combination of these for 40 and 42 amino acid-long amyloid β peptides:
approaches. Their use to examine the cell biology of presenilins
These actions may, if the Aβ theory is correct, decrease and the immunohistochemistry of sporadic
Aβ42 oligomerization and downstream toxic effects thereby Alzheimer’s disease and cerebral a ­ myloid ­angiopathy
protecting both the synapse at the early stage of disease and cases. Molecular Medicine, 3: 695–707.
latterly the neurone itself. Baskin, F., Rosenberg, R. and Davis, R.M. (1992).
Morphological differentiation and proteoglycan
synthesis regulate Alzheimer amyloid precursor
protein processing in PC-12 and human astrocyte
cultures. Journal of Neuroscience Research, 32:
48.13 CONCLUSIONS 274–279.
Bateman, R.J., Munsell, L.Y., Morris, J.C. et al. (2006).
This chapter highlights the complexity and intricacies of Human amyloid-β synthesis and clearance rates
the various systems that tie Aβ to the neuronal degenera- as measured in cerebrospinal fluid in vivo. Nature
tion of AD. One sword blow upon the Gordian knot will Medicine, 12: 856–861.
not work, rather the interventions and untying should Behl, C., Davis, J., Cole, G.M. and Schubert, D. (1992).
represent a thoughtful, careful unpicking. Intensive Vitamin E protects nerve cells from amyloid β protein
investigation of these biochemical systems has elucidated toxicity. Biochemical and Biophysical Research
several potential targets for rational therapeutic interven- Communications, 186: 944–950.
tion. The test of the Aβ theory will be in the examina- Behl, C., Davis, J.B., Lesley, R. and Schubert, D. (1994).
tion of interventions that modulate the production and Hydrogen peroxide mediates amyloid β protein
accumulation of Aβ42, initially in mice before human tri- toxicity. Cell, 77: 817–827.
als take place. In preparation for these trials, defining a Berg, L., McKeel, D.W., Miller, P. et al. (1998).
high-risk population based on genetic risk factors and the Clinicopathologic studies in cognitively healthy
observation of biological changes in patient’s precognitive aging and Alzheimer’s disease. Relation of h ­ istologic
decline will continue with the hope that more effective ­markers to dementia severity, age, sex, and
disease-modifying drugs will be available in the not too apolipoprotein E genotype. Archives of Neurology,
distant future. 55: 326–335.
502 Dementia

Bertram, L. and Tanzi, R.E. (2004). Alzheimer’s disease: of amyloid precursor protein in vitro. Journal of
One disorder, too many genes? Human Molecular Neurochemistry, 106 (2): 793–804.
Genetics, 13: R135–R141. Citron, M., Diehl, T.S., Gordon, G. et al. (1996).
Blacker, D., Bertram, L., Saunders, A.J. et al. (2003). Evidence that the 42- and 40-amino acid forms
Results of a high-resolution genome screen of 437 of amyloid beta protein are generated from the
Alzheimer’s disease families. Human Molecular β-amyloid precursor protein by different protease
Genetics, 12: 23–32. activities. Proceedings of the National Academy
Blacker, D., Haines, J.L., Rodes, L. et al. (1997). ApoE4 of Sciences of the United States of America, 93:
and age at onset of Alzheimer’s disease: The NIMH 13170–13175.
genetics initiative. Neurology, 48: 139–147. Copani, A., Koh, J. and Cotman, C.W. (1991). β-Amyloid
Borchelt, D.R., Thinakaran, G., Eckman, C.B. et al. (1996). increases neuronal susceptibility to injury by
Familial Alzheimer’s disease-linked presenilin 1 glucose deprivation. Neuropharmacology and
variants elevate Aβ-1–42/1–40 ratio in vitro and in vivo. Neurotoxicology, 2: 763–765.
Neuron, 17: 1005–1013. Corder, E.H., Saunders, A.M., Risch, N.J. et al. (1994).
Brooks, W.S., Martins, R.N., Devoecht, J. et al. Protective effect of apolipoprotein E type 2 allele
(1995). A mutation in codon 717 of the amyloid for late onset Alzheimer’s disease. Nature Genetics,
­precursor ­protein gene in an Australian family 7: 180–184.
with Alzheimer’s disease. Neuroscience Letters, Darborg, J., Andreasson, V., Pekna, M. et al. (2012).
199: 183–186. Cerebrospinal fluid level of complement proteins C3,
Bullido, M.J., Artiga, M.J., Recuero, M. et al. (1998). C4 and CR1 in Alzheimer’s disease. Journal of Neural
A polymorphism in the regulatory region of ApoE Transmission, 119: 789–797.
associated with risk of Alzheimer’s dementia. Nature Davis, A.E. 3rd, Mejia, P. and Lu, F. (2008). Biological
Genetics, 18: 69–71. activities of C1 inhibitor. Molecular Immunology,
Burlinson, S., Burns, A., Mann, D. et al. (1998). Effect of 45 (16): 4057–4063.
apolipoprotein E status on clinical features of dementia. de la Monte, S.M. and Wands, J.R. (2008). Alzheimer’s
International Journal of Geriatric Psychology, 13: disease is type 3 diabetes-evidence reviewed.
177–185.Butterfield, D.A. (2002). Amyloid β-peptide Journal of Diabetes Science and Technology,
(1-42)-induced oxidative stress and neurotoxicity: 2 (6): 1101–1113.
Implications for neurodegeneration in Alzheimer’s De Strooper, B. and Chávez Gutiérrez, L. (2015).
disease brain. A review. Free Radical Research, 36: Learning by failing: Ideas and concepts to tackle
1307–1313. γ-secretases in Alzheimer’s disease and beyond.
Butterfield, D.A., Hensley, K., Harris, M. et al. (1994). Annual Review of Pharmacology and Toxicology,
β-Amyloid peptide free radical fragments initiate 55: 419–437.
synaptosomal lipoperoxidation in a sequence- De Strooper, B., Saftig, P., Craessaerts, K. et al. (1998).
specific fashion: Implications to Alzheimer’s Deficiency of presenilin-1 inhibits the normal
disease. Biochemical and Biophysical Research ­cleavage of amyloid precursor protein. Nature,
Communications, 200: 710–715. 391: 387–390.
Butters, M. A., Young, J.B, Lopez, O. et al. (2008). Dickson, D.W. (1997). The pathogenesis of senile plaques.
Pathways linking late-life depression to persistent Journal of Neuropathology and Experimental
cognitive impairment and dementia. Dialogues in Neurology, 56: 321–339.
Clinical Neuroscience, 10 (3): 345–357. Duara, R., Barker, W.W., Lopez-Alberola, R. et al. (1996).
Buxbaum, J.D., Oishi, M., Chen, H.I. et al. (1992). Alzheimer’s disease: Interaction of apolipoprotein
Cholinergic agonists and interleukin 1 regulate E genotype, family history of dementia, gender,
­processing and secretion of the Alzheimer β/A4 education, ethnicity and age of onset. Neurology,
­amyloid protein precursor. Proceedings of the 46: 1575–1579.
National Academy of Sciences of the United States of Duff, K., Eckman, C., Zehr, C. et al. (1996). Increased
America, 89: 10075–10078. amyloid-β42(43) in brains of mice expressing mutant
Cai, X.D., Golde, T.E. and Younkin, S.G. (1993). Release of presenilin 1. Nature, 383: 710–713.
excess amyloid beta protein from a mutant amyloid β Eckman, C.B., Mehta, N.D., Crook, R. et al. (1997).
protein precursor. Science, 259: 514–516. A new pathogenic mutation in the APP gene
Caporaso, G.L., Gandy, S.E. and Buxbaum, J.D. (1992). (I716V) increases the relative proportion of
Protein phosphorylation regulates secretion A beta 42(43). Human Molecular Genetics,
of Alzheimer β/A4 amyloid precursor protein. 6: 2087–2089.
Proceedings of the National Academy of Sciences of Eikelenboom, P. and Veerhuis, R. (1996). The role
the United States of America, 89: 3055–3059. of complement and activated microglia in the
Chan, S.L., Kim, W.S., Kwok, J.B. et al. (2008). ATP- pathogenesis of Alzheimer’s disease. Neurobiology of
binding cassette transporter A7 regulates processing Aging, 17 (5): 673–680.
The central role of Aβ amyloid and tau in the pathogenesis of Alzheimer’s disease 503

Ertekin-Taner, N., Allen, M., Fadale, D. et al. (2004). cultured microglial cells. Brain Research, 601
Genetic variants in haplotype block spanning IDE (1–2): 88–94.
are signficantly associated with plasma Aβ42 levels Hensley, K., Carney, J.M., Mattson, M.P. et al. (1994).
and risk for Alzheimer disease. Human Mutation, A model for β-amyloid aggregation and neurotoxicity
23: 334–342. based on free radical generation by the peptide:
Evin, G., Beyreuther, K. and Masters, C.L. (1994). Relevance to Alzheimer disease. Proceedings of the
Alzheimer’s disease amyloid precursor protein National Academy of Sciences of the United States of
(AβPP): Proteolytic processing, secretases and America, 91: 3270–3274.
βA4 amyloid production. Amyloid: International Hollenbach, E., Ackerman, S., Hyman, B.T. and Rebeck,
Journal of Experimental and Clinical Investigation, G.W. (1998). Confirmation of an association between a
1: 263–280. polymorphism in exon 3 of the low-density lipoprotein
Farris, W., Mansourian, S., Leissring, M.A. et al. (2004). receptor-related protein gene and Alzheimer’s
Partial loss-of-function mutations in insulin-degrading disease. Neurology, 50: 1905–1907.
enzyme that induce diabetes also impair degradation Hooper, C., Killick, R. and Lovestone, S. (2008). The
of amyloid β–protein. Amercian Journal of Pathology, GSK3 hypothesis of Alzheimer’s disease. Journal of
164: 1425–1434. Neurochemistry, 104 (6): 1433–1439.
Fong, T.G., Jones, R.N., Shi, P. et al. (2009). Delirium Howlett, D.R., Jennings, K.H., Lee, D.C. et al. (1995).
accelerates cognitive decline in Alzheimer disease. Aggregation state and neurotoxic properties of
Neurology, 72 (18): 1570–1575. Alzheimer beta-amyloid peptide. Neurodegeneration,
Gambuzza, M.E., Sofo, V., Salmeri, F.M. et al. (2014). Toll- 4: 23–32.
like receptors in Alzheimer’s disease: A therapeutic Huang, C.W., Lui, C.C., Chang, W.N. et al. (2009). Elevated
perspective. CNS & Neurological Disorders—Drug basal cortisol level predicts lower hippocampal volume
Targets, 13 (9): 1542–1558. and cognitive decline in Alzheimer’s disease. Journal
Gillespie, S.L., Golde, T.E. and Younkin, S.G. of Clinical Neuroscience, 16 (10): 1283–1286.
(1992). Secretory processing of the Alzheimer Huang, Y.D., Weisgraber, K.H., Mucke, L. and Mahley,
­amyloid β/A4 protein precursor is increased R.W. (2004). Apolipoprotein E: Diversity of cellular
by protein ­phosphorylation. Biochemical and origins, structural and biophysical properties, and
Biophysical Research Communications, 187: effects in Alzheimer’s disease. Journal of Molecular
1285–1290. Neuroscience, 23: 189–204.
Glenner, G.G. and Wong, C.W. (1984). Alzheimer’s disease: Hughes, S.R., Khorkova, O., Goyal, S. et al. (1998).
Initial report of the purification and characterization of α2-Macroglobulin associates with β-amyloid peptide
a novel cerebrovascular amyloid protein. Biochemical and prevents fibril formation. Proceedings of the
and Biophysical Research Communications, National Academy of Sciences of the United States of
120: 885–890. America, 95: 3275–3280.
Golde, T.E., Estus, S., Usiak, M. et al. (1990). Expression Hung, A.Y., Koo, E.H., Haass, C. and Selkoe, D.J. (1992).
of β amyloid protein precursor mRNAs: Recognition Increased expression of beta-amyloid precursor
of a novel alternatively spliced form and quantitation protein during neuronal differentiation is not
in Alzheimer’s disease using PCR. Neuron, accompanied by secretory cleavage. Journal of
4: 253–267. Biological Chemistry, 268: 22959–22962.
Green, K.N., Billings, L.M., Roozendaal, B. et al. (2006). Hyman, B.T. and Tanzi, R.E. (1992). Amyloid, dementia and
Glucocorticoids increase amyloid-beta and tau Alzheimer’s disease. Current Opinion in Neurology and
pathology in a mouse model of Alzheimer’s disease. Neurosurgery, 5: 88–93.
Journal of Neuroscience, 26 (35): 9047–9056. Iwatsubo, T. (2004). The γ-secretase complex: Machinery
Haass, C., Hung, A.Y. and Selkoe, D.J. (1991). Processing for intramembrane proteolysis. Current Opinion in
of β-amyloid precursor protein in microglia and Neurobiology, 14: 379–383.
astrocytes favors an internal localization over Jonsson, T., Atwal, J.K., Steinberg, S. et al. (2012).
constitutive secretion. Journal of Neuroscience, A mutation in APP protects against Alzheimer’s
11: 3783–3793. disease and age-related cognitive decline. Nature,
Haass, C., Hung, A.Y., Selkoe, D.J. and Teplow, D.B. 488: 96–99.
(1994). Mutations associated with a locus for familial Jorm, A.F. (2001). History of depression as a risk factor for
Alzheimer’s disease result in alternative processing dementia: An updated review. TheAustralian and New
of amyloid β protein precursor. Journal of Biological Zealand Journal of Psychiatry, 35 (6): 776–781.
Chemistry, 269: 17741–17748. Ju, Y.E., McLeland, J.S., Toedebusch, C.D. et al. (2013).
Haga, S., Ikeda, K., Sato, M. et al. (1993). Synthetic Sleep quality and preclinical Alzheimer disease.
Alzheimer amyloid beta/A4 peptides enhance Journal of the American Medical Association
­production of complement C3 component by Neurology, 70: 587–593.
504 Dementia

Kang, J., Lemaire, H., Unterbeck, A. et al. (1987). The Lemere, C.A., Lopera, F., Kosik, K.S. et al. (1996). The
precursor of Alzheimer’s disease amyloid A4 protein E280A presenilin 1 Alzheimer mutation produces
resembles a cell-surface receptor. Nature, 325: increased Aβ42 deposition and severe cerebellar
733–736. pathology. Nature Medicine, 2: 1146–1150.
Kang, J. and Müller-Hill, B. (1990). Differential splicing Lesort, M., Jope, R. and Johnson, G.V. (1999). Insulin
of Alzheimer’s disease amyloid A4 precursor RNA transiently increases tau phosphorylation: Involvement
in rat tissues: PreA4695 mRNA is predominantly in glycogen synthase kinase-3 and Fyn tyrosine kinase.
produced in rat and human brain. Biochemical Journal of Neurochemistry, 72 (9): 576–584.
and Biophysical Research Communications, 166: Li, X., Ma, Y., Wei, X. et al. (2014). Clusterin in Alzheimer’s
1192–1200. disease: A player in the biological behaviour of
Kang, D.E., Saitoh, T., Chen, X. et al. (1997). Genetic amyloid-beta. Neuroscience Bulletin, 30 (1): 162–168.
association of the low-density lipoprotein receptor- Liao, A., Nitsch, R.M., Greenberg, S.M. et al. (1998).
related protein gene (LRP), an apolipoprotein Genetic association of an α2-macroglobulin (Val 1000
E receptor, with late onset Alzheimer’s disease. Ile) polymorphism and Alzheimer’s disease. Human
Neurology, 49: 56–61. Molecular Genetics, 7: 1953–1956.
Keller, J.N., Guo, Q., Holtsberg, F.W. et al. Lim, Y.Y., Villemagne, V.L., Laws, S.M. et al. (2015). APOE
(1998). Increased sensitivity to mitochondrial and BDNF polymorphisms moderate amyloid β-related
toxin-induced apoptosis in neural cells cognitive decline in preclinical Alzheimer’s disease.
expressing mutant presenilin-1 is linked to Molecular Psychiatry, 20: 1322–1328.
perturbed calcium homeostasis and enhanced Lockhart, B.P., Benicourt, C., Junien, J.L. and Privat, A.
oxyradical production. Journal of Neuroscience, 18: (1994). Inhibitors of free radical formation fail to
4439–4450. attenuate direct β-amyloid(25–35) peptide-mediated
Koh, J., Yang, L.L. and Cotman, C.W. (1990). β-Amyloid neurotoxicity in rat hippocampal cultures. Journal of
protein increases the vulnerability of cultured cortical Neuroscience Research, 39: 494–505.
neurons to excitotoxic damage. Brain Research, 533: Lopes, M.B., Bogaev, C.A., Gonias, S.L. and Vandenberg,
315–320. S.R. (1994). Expression of α2-macroglobulin receptor/
Kosik, K.S., Joachim, C.L. and Selkoe, D.J. (1986). low density lipoprotein receptor-related protein
Microtubule-associated protein tau (tau) is a major is increased in reactive and neoplastic glial cells.
antigenic component of paired helical filaments Federation of European Biochemical Societies Letters,
in Alzheimer disease. Proceedings of the National 338: 301–305.
Academy of Sciences of the United States of America, Lorenzo, A. and Yanker, B.A. (1994). β-Amyloid
83: 4044–4048. neurotoxicity requires fibril formation and is inhibited
Kwok, J.B.J., Li, Q.X., Hallup, M. et al. (2000). Novel Leu by Congo red. Proceedings of the National Academy
723 pro amyloid precursor protein mutation increases of Sciences of the United States of America, 91:
amyloid β42 (43) peptide levels and induces apoptosis. 12243–12247.
Annals of Neurology, 47: 249–253. Lovestone, S., Reynolds, C., Latimer, D. et al. (1994).
Lahiri, D.K., Nall, C. and Farlow, M. (1992). The cholinergic Alzheimer’s disease-like phosphorylation of the
agonist carbachol reduces intracellular β-amyloid microtubule-associated protein tau by glycogen
precursor protein in PC 12 and C6 cells. Biochemistry synthase kinase-3 in transfected mammalian cells.
International, 28: 853–860. Current Biology, 4: 1077–1086.
Lambert, J.C., Pasquier, F., Cottel, D. et al. (1998). A new MacLullich, A.M., Ferguson, K.J., Reid, L.M. et al. (2012).
polymorphism in the ApoE promoter associated with 11β-hydroxysteroid dehydrogenase type 1, brain
risk of developing Alzheimer’s dementia. Human atrophy and cognitive decline. Neurobiology of Aging,
Molecular Genetics, 7: 533–540. 33 (1): 207.
Lammich, S., Kojro, E., Postina, R. et al. (1999). Mahley, R.W. (1988). Apoliprotein E: Cholesterol transport
Constitutive and regulated α-secretase cleavage of protein with expanding role in cell biology. Science,
Alzheimer’s amyloid precursor protein by a disintegrin 240: 622–630.
metalloprotease. Proceedings of the National Mark, R.J., Blanc, E.M. and Mattson, M.P. (1996). Amyloid
Academy of Sciences of the United States of America, β-peptide and oxidative cellular injury in Alzheimer’s
96: 3922–3927. disease. Molecular Neurobiology, 12: 211–224.
Lannfelt, L., Blennow, K., Zetterberg, H. et al. (2008). Masters, C.L., Bateman, R., Blennow, K. et al. (2015).
Safety, efficacy, and biomarker findings of PBT2 Alzheimer’s disease. Nature Reviews Disease Primers,
in targeting Abeta as a modifying therapy for Oct 15 (1): 15056.
Alzheimer’s disease: A phase IIa, double-blind, Masters, C.L., Simms, G., Weinman, N.A. et al. (1985).
randomised, placebo-controlled trial. Lancet Amyloid plaque core protein in Alzheimer disease
Neurology, 7 (9): 779–786. and Down syndrome. Proceedings of the National
The central role of Aβ amyloid and tau in the pathogenesis of Alzheimer’s disease 505

Academy of Sciences of the United States of America, Olichney, J.M., Hansen, L.A., Galasko, D. et al. (1996). The
82: 4245–4249. apolipoprotein E ε4 allele is associated with increased
Mattson, M.P., Cheng, B., Culwell, A.R. et al. neuritic plaques and cerebral amyloid angiopathy
(1993). Evidence for excitoprotective and in Alzheimer’s disease and Lewy body variant.
intraneuronal calcium-regulating roles for secreted Neurology, 47: 190–196.
forms of the β-amyloid precursor protein. Neuron, Payami, H., Montee, K.R., Kaye, J.A. et al. (1994).
10: 243–254. Alzheimer’s disease, apolipoprotein e4, and gender.
Mawuenyega, K.G., Sigurdson, W., Ovod, V. et al. (2010). Journal of the American Medical Association, 271:
Decreased clearance of CNS β-amyloid in Alzheimer’s 1316–1317.
disease. Science, 330: 1774. Phiel, C.J., Wilson, C.A., Lee, V.M. and Klein, P.S. (2003).
McKee, A.C., Kosik, K.S. and Kowall, N.W. (1991). Neuritic GSK-3alpha regulates production of Alzheimer’s
pathology and dementia in Alzheimer’s disease. disease amyloid-beta peptides. Nature, 423 (6938):
Annals of Neurology, 30: 156–165. 435–439.
McLean, C.A., Cherney, R.A., Fraser, F.W. et al. (1999). Poirier, J. (1994). Apolipoprotein E in CNS models of
Soluble pool of Aβ as a determinant of severity of CNS injury and in Alzheimer’s disease. Trends in
neurodegeneration in Alzheimer’s disease. Annals of Neuroscience, 17: 525–530.
Neurology, 46: 860–866. Poirier, J., Davignon, J., Bouthillier, D. et al. (1993).
Meda, L., Baron, P. and Scarlato, G. (2001). Glial activation Apolipoprotein E polymorphism and Alzheimer’s
in Alzheimer’s disease: The role of Abeta and its disease. Lancet, 342: 697–699.
associated proteins. Neurobiology of Aging, 22 (6): Polvikoski, T., Sulkava, R., Haltia, M. et al. (1995).
885–893. Apolipoprotein E, dementia and cortical deposition
Milward, E., Papadopoulos, R., Fuller, S.J. et al. (1992). of beta-amyloid protein. New England Journal of
The amyloid protein precursor of Alzheimer’s Medicine, 333: 1242–1247.
disease is a mediator of the effects of nerve Rebeck, G.W., Reiter, J.S., Strickland, D.K. and Hyman,
growth factor on neurite outgrowth. Neuron, B.T. (1993). Apolipoprotein E in sporadic Alzheimer’s
9: 129–137. disease: Allelic variation and receptor interactions.
Morris, J.C., McKeel, D.W., Storandt, M. et al. (1991). Very Neuron, 11: 575–580.
mild Alzheimer’s disease: Informant-based clinical Reddy, P.H. and Beal, M.F. (2008). Amyloid beta,
psychometric and pathologic distinction from normal mitochondrial dysfunction and synaptic damage:
aging. Neurology, 41: 469–478. Implications for cognitive decline in aging and
Myers, R., Schaeter, E.J., Wilson, P.W. et al. (1996). Alzheimer’s disease. Trends in Molecular Medicine,
Apolipoprotein E4 association with dementia in a 14 (2): 45–53.
population based study: The Framingham study. Reitz, C., Jun, G., Naj, A. et al. (2013). Variants in the ATP-
Neurology, 46: 673–677. binding cassette transporter (ABCA7), apolipoprotein
Narita, M., Holtzman, D.M., Schwartz, A.L. and Bu, E ϵ4, and the risk of late-onset Alzheimer disease in
G.J. (1997). α2-Macroglobulin complexes with and African Americans. Journal of the American Medical
mediates the endocytosis of beta-amyloid peptide Association, 309 (14): 1483–1492.
via cell surface low-density lipoprotein receptor- Rhein, V. and Eckert, A. (2007). Effects of Alzheimer’s
related protein. Journal of Neurochemistry, 69: amyloid-beta and tau protein on mitochondrial
1904–1911. function—Role of glucose metabolism and insulin
National Institute on Aging/Alzheimer’s Association signalling. Archives of Physiology and Biochemistry,
Working Group. (1996). Apolipoprotein E genotyping 113 (3): 131–141.
in Alzheimer’s disease. Lancet, 347: 1091–1095. Ritchie, C.W., Ames, D., Clayton, T. and Lai, R. (2004).
Naylor, R., Hill, A.F. and Barnham, K.J. (2008). Is covalently Metaanalysis of randomized trials of the efficacy and
crosslinked Abeta responsible for synaptotoxicity in safety of donepezil, galantamine, and rivastigmine for
Alzheimer’s disease? Current Alzheimer Research, 5 (6): the treatment of Alzheimer disease. American Journal
533–539. of Geriatric Psychiatry, 12 (4): 358–369.
Nicoll, J., Roberts, G.W. and Graham, D. (1995). Ritchie, C.W., Bush, A.I., Mackinnon, A. et al. (2003).
Apolipoprotein E epsilon 4 allele is associated with Metal-protein attenuation with iodochlorhydroxyquin
deposition of amyloid following head injury. Nature (clioquinol) targeting A beta amyloid deposition
Medicine, 1: 135–137. and toxicity in Alzheimer disease—A pilot
Nitsch, R.M., Slack, B.E., Wurtman, R.J. and Growdon, phase 2 clinical trial. Archives of Neurology, 60
J.H. (1992). Release of Alzheimer amyloid (12): 1685–1691.
precursor derivatives stimulated by activation Rogaev, E.I. (1998). Presenilins—Discovery and
of muscarinic acetylcholine receptors. Science, characterization of the genes of Alzheimer’s disease.
258: 304–307. Molecular Biology, 32: 58–69.
506 Dementia

Rogers, J., Cooper, N.R., Webster, S. et al. (1992). Smith, D.G., Cappai, R. and Barnham, K.J. (2007). The
Complement activation by β-amyloid in Alzheimer’s redox chemistry of the Alzheimer's disease amyloid
disease. Proceedings of the National Academy beta peptide. Biochim Biophys Acta, 1768 (8):
of Sciences of the United States of America, 1976–1990.
89: 10016–10020. Struhl, G. and Greenwald, I. (1999). Presenilin is required
Roh, J.H., Huang, Y., Bero, A.W. et al. (2012). Disruption for activity and nuclear access of notch in Drosophila.
of the sleep-wake cycle and diurnal fluctuation Nature, 398: 522–524.
of beta-amyloid in mice with Alzheimer’s disease Suzuki, N., Iwatsubo, T., Odaka, A. et al. (1994). High
pathology. Science Translational Medicine, 4 (150): tissue content of soluble beta 1–40 is linked to
150ra122. cerebral amyloid angiopathy. American Journal of
Roperch, J.P., Alvaro, V., Prieur, S. et al. (1998). Inhibition Pathology, 145: 452–460.
of presenilin 1 expression is promoted by p53 Szaruga, M., Veugelen, S., Benurwar, M. et al. (2015).
and p21waf-1 and results in apoptosis and tumor Qualitative changes in human gamma-secretase
­suppression. Nature Medicine, 4: 835–838. underlie familial Alzheimer’s disease. Journal of
Sano, M., Ernesto, C., Thomas, R.G. et al. (1997). A Experimental Medicine, 212: 2003–2013.
controlled trial of selegiline, alpha-tocopherol, or both Tan, M.S., Yu, J.T. and Tan, L. (2013). Bridging integrator 1
as treatment for Alzheimer’s disease. New England (BIN1): Form, function and Alzheimer’s disease. Trends
Journal of Medicine, 336: 1216–1222. in Molecular Medicine, 19 (10): 594–603.
Saunders, A.M., Strittmatter, W.J., Schmechel, D. et al. Tanaka, N., Abe-Dohmae, S., Iwamoto, N. et al. (2011). Roles
(1993). Association of apolipoprotein E allele ϵ4 with of ATP-binding cassette transporter A7 in cholesterol
late‐onset familial and sporadic Alzheimer's disease. homeostasis and host defense system. Journal of
Neurology, 43 (8): 1467–1467. Atherosclerosis and Thrombosis, 18 (4): 274–281.
Scheuner, D., Eckman, C., Jensen, M. et al. (1996). Secreted Tanzi, R.E., Gusella, J.F., Watkins, P.C. et al. (1987).
amyloid β-protein similar to that in the senile plaques of Amyloid β protein gene: cDNA, mRNA distribution
Alzheimer’s disease is increased in vivo by the presenilin and genetic linkage near the Alzheimer locus. Science,
1 and 2 and APP mutations linked to familial Alzheimer’s 235: 880–884.
disease. Nature Medicine, 2: 864–870. Teller, J.K., Russo, C., DeBusk, L.M. et al. (1996). Presence
Schmechel, D.E., Saunders, A.M., Strittmatter, of soluble amyloid β-peptide precedes amyloid plaque
W.J. et al. (1993). Increased amyloid β-peptide formation in Down’s syndrome. Nature Medicine,
deposition in cerebral cortex as a consequence of 2: 93–95.
apolipoprotein E genotype in late-onset Alzheimer’s Terry, R.D., Masliah, E., Salmon, D.P. et al. (1991). Physical
disease. Proceedings of the National Academy basis of cognitive alterations in Alzheimer’s disease:
of Sciences of the United States of America, 90: Synapse loss is the major correlate of cognitive
9649–9653. ­impairment. Annals of Neurology, 30: 572–580.
Shearman, M.S., Ragan, C.I. and Iversen, L.L. (1994). Thambisetty, M., An, Y., Nalls, M. et al. (2013). Effect of
Inhibition of PC12 cell redox activity is a specific, complement CR1 on brain amyloid burden during
early indicator of the mechanism of β-amyloid- aging and its modification by APOE genotype.
mediated cell death. Proceedings of the National Biological Psychiatry, 73 (5): 422–428.
Academy of Sciences of the United States of America, Vassar, R., Kovacs, D.M., Yan, R. and Wong, P.C. (2009).
91: 1470–1474. The beta-secretase enzyme BACE in health and
Shankar, G.M. and Walsh, D.M. (2009). Alzheimer’s Alzheimer’s disease: Regulation, cell biology, function,
disease: Synaptic dysfunction and Aβ. Molecular and therapeutic potential. Journal of Neuroscience,
Neurodegeneration, 4: 48. 29 (41): 12787–12794.
Sinha, S. and Lieberburg, I. (1992). Normal metabolism Vasto, S., Mocchegiani, E., Malavolta, M. et al. (2007). Zinc
of the amyloid precursor protein (APP). and inflammatory/immune response in aging. Annals
Neurodegeneration, 1: 169–175. of New York Academy of Science, 1100: 111–122.
Slooter, A.J.C., Tang, M.X., van Duijn, C.M. et al. (1997). Villemagne, V.L., Burnham, S., Bourgeat, P. et al; for
Apolipoprotein E ε4 and the risk of dementia the Australian Imaging Biomarkers and Lifestyle
with stroke (a population based investigation). (AIBL) Research Group. (2013). Amyloid β deposition,
Journal of the American Medical Association, ­neurodegeneration and cognitive decline in sporadic
277: 818–821. Alzheimer’s disease: A prospective cohort study. The
Small, D.H., Nurcombe, V., Reed, G. et al. (1994). Lancet Neurology, 12: 357–367.
A ­heparin-binding domain in the amyloid protein Wisniewski, T., Golabek, A., Matsubara, E. et al. (1993).
­precursor of Alzheimer’s disease is involved in Apolipoprotein E: Binding to soluble Alzheimer’s
the ­regulation of neurite outgrowth. Journal of β-amyloid. Biochemical and Biophysical Research
Neuroscience, 14: 2117–2127. Communications, 192: 359–365.
The central role of Aβ amyloid and tau in the pathogenesis of Alzheimer’s disease 507

Whitson, J.S. and Appel, S.H. (1995). Neurotoxicity axonal microtubule associated protein tau
of Aβ amyloid protein in vitro is not altered by (tau). Proceedings of the National Academy of
calcium channel blockade. Neurobiology of Aging, Sciences of the United States of America, 83:
16: 5–10. 4040–4043.
Wolfe, M.S., Xia, W., Ostaszewski, B.L. et al. (1999). Two Yankner, B.A. (1996). Mechanisms of neuronal
transmembrane aspartates in presenilin-1 required for degeneration in Alzheimer’s disease. Neurology,
presenilin endoproteolysis and γ-secretase activity. 16: 921–932.
Nature, 398: 513–517. Ye, Y., Lukinova, N. and Fortini, M.E. (1999).
Wood, J.G., Mirra, S.S., Pollock, N.J. and Binder, Neurogenic phenotypes and altered Notch
L.I. (1986). Neurofibrillary tangles of Alzheimer ­processing in Drosophila presenilin mutants.
disease share antigenic determinants with the Nature, 398: 525–529.
49
Inflammation in Alzheimer’s disease

CLIVE HOLMES

49.1 INTRODUCTION 49.2 PRECLINICAL STUDIES

In the past, the role of neuroinflammation in Alzheimer’s 49.2.1 CNS RESIDENT CELLS


disease (AD) was considered to be a simple response to the
well-established neuropathological features (e.g. the neu- 49.2.1.1 Microglia
ritic plaques or tangles) of the disease. However, emerg-
Microglia are unique among the major cell types of the
ing evidence now shows it is a major contributor to the
CNS in that they are not derived from the neuroectoderm
progression and development of the disease. Indeed, both
(Chan, et al., 2007). These resident CNS cells were originally
preclinical and clinical research support an early and sub-
derived from myeloid precursors and are representatives of
stantial involvement of neuroinflammation in AD patho-
the early migration of monocytes from the periphery into
genesis that changes in character as the disease progresses.
the brain (Ginhoux et al., 2010).
The term ‘neuroinflammation’, in its broadest sense,
Microglia are ubiquitously distributed throughout the
encompasses any inflammatory process, whether acute or
brain and constantly survey their environment for the
chronic, involving the nervous system. Depending on the presence of pathogens, protein aggregates and evidence of
nature of the inflammatory process diverse cell types may neuronal cell death or synaptic damage. Detection of these
be involved. The central nervous system (CNS) resident triggers is mediated by receptors that recognize pathogen-
cells (microglia and astrocytes) are a major component of associated molecular patterns (PAMPs), danger-associated
this inflammatory response. However, in some circum- molecular patterns (DAMPs) and other cell surface recep-
stances, e.g. where the blood–brain barrier (BBB) is dam- tors including cluster of differentiation (CD) 33, CD36,
aged or in areas surrounding the vasculature of the brain, CD40 and toll-like (TL) receptors (e.g. TLR3, TLR4 and
other peripherally derived cells (neutrophils, lymphocytes, TLR6) that drive the activation of these microglial cells
plasma cells, macrophages and monocytes) may also be (Bianchi, 2007; Medzhitov, 2007; Kono and Rock, 2008;
involved. In AD, the key cellular players are thought to be Stewart et al., 2010).
the CNS resident cells with its key mediators being cyto- Activation of microglia to these triggers is a complex
kine proteins. However, there is also a developing interest process and our understanding is largely based on mod-
in the potential role for peripherally derived cells and other els of activated macrophages outside the CNS. Here acti-
mediators of inflammation. Furthermore, there is increas- vated macrophages are considered to be of two broad
ing recognition that neuroinflammatory processes in the phenotypes. The first phenotype M1 or classical activation
AD brain are markedly influenced by inflammation that is defined by the response of macrophages to challenge
occurs outside the CNS. A greater understanding of the by the cytokine interferon-γ, a cytokine usually associ-
central and peripheral inflammatory processes involved in ated with host defence to intracellular pathogens. M1 is
AD will undoubtedly enable the development of clinical pro-inflammatory and is characterized by increases in
tools allowing the early detection of the disease but will, pro-inflammatory cytokines, including interleukin (IL)-1,
most importantly, lead to new preventative strategies and IL-6, IL-12 and tumour necrosis factor (TNF) α, and is
treatment approaches. associated with tissue damage caused by increases in nitric

508
Inflammation in Alzheimer’s disease 509

oxide and reactive oxygen species. The second major phe- 49.2.1.2 Astrocytes
notype M2 or alternative activation phenotype is defined
by the response to the cytokine IL-4, a cytokine associ- Astrocytes, unlike microglial cells, are ectodermally
ated with the host defence to extracellular pathogens and derived (Campbell and Williams, 1978). Their role in medi-
is associated with the upregulation of the mannose recep- ating CNS inflammation has been relatively neglected.
tor that binds to the surface of bacteria and fungi enabling They have an important role in innate immunity, including
their phagocytosis. M2 is characterized by secretion of cytokine production, complement and nitric oxide produc-
anti-inflammatory cytokines IL-4, IL-10, IL-13 and trans- tion (Sofroniew and Vinters, 2010). In AD astrocytes, like
forming growth factor (TGF)-β and is also associated with microglia, Aβ plaques are found in a hypertrophic reactive
tissue repair and remodelling. A third type of activation, state and they also have a role in the internalization and
acquired deactivation (considered a subtype of M2), is degradation of fibrillar Aβ (Medeiros and LaFerla, 2013).
defined by the response to the cytokine IL-10, a cytokine In addition, astrocytes have a role in clearance of soluble
associated with the presence of apoptotic cells. Acquired Aβ from the parenchyma to cerebral blood vessels, a pro-
deactivation is associated with removal of apoptotic cells, cess that requires the presence of apolipoprotein E (ApoE)
a strong immunosuppressive profile and by an increase in (Koistinaho et al., 2004). In addition, astrocytes are central
the cytokine TGF-β (Colton, 2009; Boche et al., 2013). to the maintenance of synaptic transmission and because of
Against this complex background is the finding that their close contact with neurones and cerebral blood vessels
microglial cells show enhanced sensitivity to inflammatory (the neurovascular unit) they can also act to modify BBB
stimuli with ageing with an upregulation of a range of cell permeability (Farina et al., 2007).
surface receptors and an exaggerated microglial response
to a second inflammatory stimulus. This phenomenon is 49.2.2 SYSTEMIC INFLAMMATION
called priming and its exact causes are unknown. It might AND ITS EFFECTS ON CNS
be caused by microglial senescence or by prolonged expo- INFLAMMATION
sure to the aged neuronal environment including the pres-
ence of amyloid beta (Aβ) protein. The result is that in aged In the past, the parenchyma of the CNS was considered
animal or AD mouse models, priming results in microglial an ‘immunologically privileged site’ because the BBB was
cells show that a markedly increased production of pro- thought to prevent the entry, or exit, of many molecules,
inflammatory cytokines, enhanced phagocytic activity including antibodies from the periphery and was largely
and an increased production of reactive oxygen species by devoid of macrophages, neutrophils and lymphocytes. This
microglia follow this second stimuli. Thus, priming results historical concept has led to research on CNS inflamma-
in a microglial state that are once activated cut across the tion being viewed in isolation and independent of systemic
simple classification of M1 or M2 state by having elements inflammation occurring outside the CNS. However, com-
of both (Perry et al., 2010; Perry and Holmes, 2014). munication of the presence of systemic inflammation to
It is clear from the above description that the activation the brain is clearly supported by the constellation of cen-
of microglial cells can have both beneficial and detrimental trally derived symptoms that occur in animals following an
effects. Classical activation of microglia (e.g. following trau- acute systemic bacterial or viral infection. This syndrome
matic brain injury and stroke) is associated with persistent is known as ‘sickness behaviour’ and includes anorexia,
microglial activation, neuronal damage and reduced phago- depression, somnolence, decreased social interaction,
cytic removal of Aβ and detrimental outcomes on disease pro- decreased concentration and fever and is an evolutionary
gression. However, activation of the alternative pathway might conserved, homeostatic mechanism that allows the body to
be considered to have potentially positive effects by increasing adapt to an insult (Hart, 1988). The state of the BBB in AD is
phagocytosis of Aβ (Koshinaga et al., 2000; Gentleman et al., still uncertain, and although controversial, it is possible that
2004; Tajiri et al., 2013). Indeed, cellular studies have shown there may, in limited circumstances, be some direct entry of
that microglia bind to soluble Aβ oligomers and Aβ fibrils immune cells (monocytes, macrophages) from the periph-
through the cell surface CD33 receptors (Griciuc et al., 2013). ery into the brain. Thus, it is known that sepsis (Lee and
This results in activation of microglia with extracellular enzy- Slutsky, 2010) and persistent peripheral inflammatory stim-
matic degradation of soluble Aβ and the phagocytosis of Aβ uli may compromise the BBB because of a breakdown of
fibrils and degradation in the endolysosomal pathway. Indeed, the intercellular tight junctions caused by lipopolysaccha-
in late-onset AD, inefficient clearance of Aβ by microglia ride (LPS) and peptidoglycans (Nau et al., 2010). However,
because of defects in the alternative pathway is thought to lead there are a number of other communication routes from the
to the accumulation of Aβ. This inefficient clearance is thought periphery to the brain that occur despite the presence of an
to be caused in part by downregulation of CD33 receptors. In intact BBB. These include the following:
support of this hypothesis, peripheral macrophages isolated
from subjects carrying the single nucleotide polymorphism 1. Stimulation of peripheral nerves (e.g. the vagus) by
(SNP) in CD33 and established as a risk factor for the develop- cytokines and prostaglandin that signal to the medulla
ment of AD have been shown to have reduced Aβ phagocyto- oblongata and are relayed to the hypothalamus
sis (Bradshaw et al., 2013). (Ek et al., 1998; Dantzer et al., 2000).
510 Dementia

2. Direct actions of LPS or pro-inflammatory cytokines c­ oncentrations of TNF-α in mouse models of AD (Jin et al.,
at brain areas lacking a BBB, e.g. the circumventricu- 2008). However, although a pro-inflammatory cytokine
lar organs (Blatteis et al., 1983). ­expression drive may drive the development of the disease,
3. Direct action of LPS or pro-inflammatory cytokines of once there is appreciable cell death, mouse models show
perivascular macrophages in the neurovascular unit of that the inflammatory response more closely resembles an
the BBB (Matsumura and Kobayashi, 2004). acquired deactivation state becoming more muted with
active apoptosis (Streit et al., 2004; Streit, 2006).
In animals, unless the peripheral inflammatory event is Consistent with the priming hypothesis, in a mouse APP/
extreme, sickness behaviour is a relatively benign and tran- presenilin (PS)1 model of AD where microglial are primed
sient phenomenon (Perry, 2010). Damage limitation in the by the presence of Aβ, the additional transgenic expression
brain is in part due to the wide number of regulatory mecha- of a second pro-inflammatory stimulus (IL-1β) leads to a
nisms that reduce the pro-inflammatory response to an acute robust inflammatory state and a reduction of amyloid plaque
challenge within the brain. Thus, following a peripheral sig- pathology (Ghosh et al., 2013). In another study, expression
nal the pro-inflammatory response in the brain appears to be of interferon-γ in the brain of another amyloid mouse
suppressed by the increased production of a large variety of model showed the ability of this pro-inflammatory cytokine
proteins from microglial cells including anti-inflammatory to enhance clearance of amyloid plaques, with a widespread
cytokines IL-10; TGF-β and other cytokine signalling protein increase in astrogliosis and microgliosis (Chakrabarty et
suppressors (Rivest, 2009). However, in circumstances where al., 2010a, 2010b). Additionally, these mice had decreased
microglial cells are primed the response of these cells to even concentrations of soluble Aβ and Aβ plaque burden, with-
a very modest peripheral inflammatory event is very different. out altered APP processing. Similar results were obtained
Here, the presence of even modest peripheral inflammation following the expression of the pro-inflammatory cytokines
can switch the ­protective downregulated state to a damag- IL-6 and TNF-α (Chakrabarty et al., 2010a, 2010b, 2011)
ing phenotype with increased pro-inflammatory cytokine and conversely, the expression of the anti-inflammatory
productive and oxidative stress and neuronal damage (Perry cytokine IL-4 resulted in an exacerbation of Aβ deposition
and Holmes, 2014). This hypothesis is supported by animal (Chakrabarty et al., 2012).
studies showing an exaggerated inflammatory and oxidative In addition to their direct actions via surface receptors,
stress response to peripheral stimuli in aged mice (Godbout cytokines also stimulate inducible nitric oxide synthase
and Johnson, 2009), increased concentrations of IL-1β in the (iNOS) in microglia and astroglia cells, producing high
CNS and neuronal apoptosis in the ME7 prion mouse after concentrations of nitric oxide that is toxic to neurones,
peripheral challenge with the bacterial mimic LPS or the viral whereas genetic knockouts of iNOS are protective in mouse
mimic polyinosinic–polycytidylic acid (Cunningham et al., models of AD (Nathan et al., 2005). Likewise, nicotinamide
2005, 2009; Field et al., 2010). adenine dinucleotide phosphate (NADPH) oxidase is highly
expressed by microglia and rapidly activated by inflamma-
49.2.3 MOLECULAR MEDIATORS OF CNS tory stimuli such as Aβ, resulting in production of hydro-
INFLAMMATION gen peroxide, which further promotes microglia activation
(Jekabsone et al., 2006; Choi et al., 2012).
Both astrocytes and microglial cells are sources of a variety of Chemokines have a role to play in microglial migration to
molecules that orchestrate the inflammatory response. These areas of neuroinflammation, thereby potentially enhancing
include cytokines, chemokines, nitric oxide, hydrogen perox- local inflammation in AD (Savarin-Vuaillat and Ransohoff,
ide, complement and antimicrobial peptides (AMPS). 2007). In AD, upregulation of the chemokines CCL2, CCR3,
Cytokines in the brain are produced largely by microglia CCL4 and CCR5 has been reported in reactive microglia
and astrocytes. There is increasing evidence from animal and astrocytes (Ishizuka et al., 1997; Xia et al., 1998). In
studies to suggest that the development of AD is preceded in vitro studies of human post-mortem tissue, Aβ has been
by a relative increase in the pro-inflammatory cytokine shown to lead to the generation of CXCL8 (also known as
response (M1), e.g. TNF-α, IL-6 and IL-1β, and a reduced IL8), CCL2, CCL3 in human macrophages and astrocytes
anti-inflammatory cytokine response (M2), e.g. IL-4, IL-10 (Lue et al., 2001; Smits et al., 2002).
and IL-13. Thus, both cellular and animal studies of amy- The complement system is a major constituent of the innate
loid precursor protein (APP) transgenic mice show that immune system, mainly involved in the defence against
pro-inflammatory cytokine expression increases with pathogens. Activation of the proteolytic complement cas-
increasing Aβ levels (Patel et al., 2005). Several other inter- cade results in opsonization and in lysis of microorganisms.
actions also suggest an M1-like activation state to exist in In the brain, the major cells that contribute to the produc-
early AD. For example, in neurone–microglia co-cultures, tion of proteins of the complement system are microglia and,
the synergistic action of Aβ with either interferon-γ or to a lesser extent, astrocytes (Veerhuis et al., 2011). Activated
CD40 ligand triggers TNF-α secretion and production of factors of the complement system are associated with Aβ
neurotoxic reactive oxygen species (Meda et al., 1995; Tan deposits (Strohmeyer et al., 2002) and a number of genetic
et al., 1999, 2002). Additionally, the innate immune r­ eceptor variants of complement are associated with the develop-
Toll-like r­eceptor 4 (TLR4) is responsible for increased ment of AD providing strong evidence for the importance
Inflammation in Alzheimer’s disease 511

of the complement system in disease pathogenesis (Harold or years following traumatic brain injury (Koshinaga
et al., 2009; Lambert et al., 2009). et al., 2000; Ramlackhansingh et al., 2011) and several stud-
AMPS are produced by a large number of cells includ- ies have established traumatic brain injury as a risk factor
ing microglia and other phagocytes and endothelial cells in for development of AD (Sivanandam and Thakur, 2012).
the defence against pathogens (Lupetti et al., 2003). AMPS Likewise, although different mechanisms are involved in
are small proteins, usually less than 50 amino acids long, the pathogenesis of stroke, there is increasing evidence that
that interact with the outer membrane of pathogens lead- the pro-inflammatory response to the damage caused by the
ing to pore formation and the destabilization of the mem- stroke accounts for its progression (Feuerstein et al., 1998).
brane (Cudic and Otvos, 2002; Radek and Gallo, 2007). Again, stroke is a well-established risk factor for the devel-
Oligomerization of AMPS is a key to the targeting and per- opment of AD (Honig et al., 2003).
meabilization of membranes (Kourie and Shorthouse, 2000) A range of inflammatory conditions outside the CNS
with non-oligomerized fibrils having little antimicrobial are also known to act as risk factors for the development
properties. AMPS also trigger the upregulation of TNF-α of AD. Delirium, a condition largely attributed to the pres-
and CXCL8. A number of cerebral AMPS are known to exist ence of a variety of acute systemic infections, is associ-
including neuro-AMP but it has recently been recognized ated with an increased risk of developing dementia. Thus,
that Aβ1–42 is an AMP (Soscia et al., 2010). Thus, it has been the increased risk of developing dementia in cognitively
hypothesized that the development of Aβ deposits in AD intact individuals following a delirium is substantial with
may be initiated by pathogens originating in the periphery a cumulative incidence of 55% after a 1-year follow-up
and entering the brain following altered BBB permeability (Rahkonen et al., 2000). In addition, the risk of develop-
(Welling et al., 2015). Whether the signalling mechanisms ing AD appears increased following the development of
communicating the presence of systemic inflammation to an acute infection in the absence of an obvious delirium.
the brain are also associated with increased brain AMPS are Thus, in a retrospective general practitioner database the
not known, but animal studies have shown that peripheral presence of one or more infections over a 5-year follow-
infections in animal with an intact BBB are associated with up period increased the odds of developing AD by around
the deposition of amyloid (Krstic et al., 2012). twofold with risk increasing with age (Dunn et al., 2005).
In addition to acute infections there is also increasing evi-
dence for a role for low-grade chronic peripheral infections
being associated with increased risk. Thus, periodontitis,
49.3 CLINICAL STUDIES a condition caused by chronic infection with periodontal
bacteria has been identified as a potential risk factor for
49.3.1 RISK FACTORS the development of AD (Stein et al., 2007) with research
showing that both elevated levels of antibodies against
49.3.1.1 The ageing inflammatory periodontal bacteria and raised peripheral TNF-α are
environment associated with the disease (Kamer et al., 2009). Likewise,
although not all studies have been consistent (Shiota et al.,
Ageing is the major risk factor for the development of AD. 2011), the gut bacterium Helicobacter pylori has also been
One possible explanation for this is the observed change in identified as a possible risk factor for the development
the regulation of inflammatory pathways as we age, a con- of AD (Kountouras et al., 2006; Shiota et al., 2011). In
cept known as inflammaging (Franceschi et al., 2007). Thus, addition to infective agents, aseptic chronic inflamma-
as we get older there is a low-grade chronic upregulation of tory conditions including atherosclerosis (Casserly and
the pro-inflammatory response and a relative decline in Topol, 2004), diabetes (Donath and Shoelson, 2011) and
adaptive immunity and the anti-inflammatory cell response depressive illness (Andersen et al., 2005) have a robust
(Franceschi et al., 2007). The reasons for inflammaging are epidemiological basis for being proposed as risk factors in
thought to be due to various factors that increase the imbal- the development of AD. For all of these risk factors, the
ance between pro-inflammatory and anti-inflammatory individual attributable risk is likely to be small (Launer
networks. et al., 1999). However, the combined cumulative effects
Possibly the most important group of inflammaging over time might be considerable.
factors is exposure to an increased range of inflamma- Importantly, there are a number of modifying factors
tory diseases or events with age. Thus, age can be seen as a that can influence the onset of inflammaging. One impor-
proxy for lifelong exposure to these inflammatory events. tant modifying factor is age-related endocrine dyscrasia.
These events include pro-inflammatory conditions such Thus, the loss of sex steroids and associated elevation of
as recurrent acute or chronic inflammatory diseases that gonadotrophins as we age is associated with an increase
directly or indirectly affect the brain from the systemic in the pro-inflammatory state and the development of AD
environment. pathology (Clark and Atwood, 2011; Butchart et al., 2013).
Two major conditions, traumatic brain injury and stroke, Likewise, lifestyle factors may also modify inflammaging
cause direct CNS inflammation. Human studies have and delay the onset of AD, including exercise and calorific
shown that a pro-inflammatory state can persist for months restriction (Ngandu et al., 2015).
512 Dementia

49.3.1.2 Genetic factors component of early-onset AD, more recent studies have now
identified inflammatory pathways as a major component of
Following ageing, genetic inheritance is considered the sec- late-onset AD.
ond most important risk factor for the development of AD.
In a small number of individuals, less than 0.1% of the 49.3.2 BLOOD AND CEREBROSPINAL
total AD population, mutations in one of three genes, pre-
FLUID (CSF) MARKERS
senilin 1, presenilin 2 and APP, or a duplication of the APP
gene (Campion et al., 1999; Rovelet-Lecrux et al., 2006) is A number of studies have tried to establish differences
directly responsible for the development of early-onset AD in serum or plasma markers of inflammation between
by altering amyloid processing so that Aβ deposition is AD populations and aged matched control groups in
greatly enhanced (Hardy, 2006). However, the majority of cross-sectional studies. A meta-analysis of these studies
the cases of AD are of late onset. Genome-wide association (Swardfager et al., 2010) suggests that overall there are
studies (GWAS) have now established that, in addition to the increases in pro-inflammatory cytokines in AD compared
well-established genetic risk factor ApoE ε4, a large number with control groups, although clearly cross-sectional
of less common genetic polymorphisms are also associated studies cannot establish whether this is cause or effect.
with increased risk of developing late-onset AD. While these More convincing data come from longitudinal studies.
new risk alleles have a much smaller individual effect on AD Thus, C-reactive protein (CRP) and IL-6 have been found
susceptibility than ApoE ε4 there is good evidence to suggest to be elevated 5 years before the clinical onset of dementia
that, in combination, they have a substantial impact on AD (Engelhart et al., 2004; Tilvis et al., 2004; Kuo et al., 2005).
predisposition. Thus, estimates of the population-attribut- Indeed, a raised CRP in midlife has been associated with
able fractions for these individual new loci are smaller than a threefold increased risk of developing AD up to 25 years
for ApoE ε4 (in the range 1%–8.1% c.f. 27.3%) but the cumu- later (Laurin et al., 2009) and has been shown to be raised
lative population-attributable fraction effect is 31.7% and is in prodromal AD (Hu et al., 2012). Another study has found
therefore comparable (Lambert et al., 2013). that higher levels of serum soluble TNF receptors (TNFR1
Of the genetic variants identified by GWAS the immune and TNFR2) in mild cognitive impairment (MCI) subjects
and complement system/inflammatory response pathway is are also associated with an increase in the development of
the most substantial accounting for 11 of the 22 genes iden- dementia over a 4- to 6-year follow-up period (Buchhave
tified to date. Thus the first two large-scale GWAS (Harold et al., 2010). Furthermore, a proteomics study that had
et al., 2009; Lambert et al., 2009) both identified poly- not a priori identified inflammatory protein candidates
morphisms in the genes encoding complement receptor 1 as potential markers of disease development reported the
(CR1) and clusterin. CR1 has binding sites for complement ­presence of a number of plasma inflammatory proteins,
factors C3b and C4b. Like ApoE, clusterin is involved in including complement factors and clusterin to be associated
lipid transport, but it has also been hypothesized to influ- with hippocampal atrophy and clinical progression in MCI
ence receptor-mediated clearance of Aβ from the brain by and AD (Thambisetty and Lovestone, 2010). In another AD
microglial endocytosis (Nuutinen et al., 2009). Two sub- cohort, evidence of acute and chronic systemic inflamma-
sequent studies (Hollingworth et al., 2011; Naj et al., 2011) tory diseases was associated with raised serum TNF-α levels
identified six additional genes (CD33, the MS4A6–MS4A4 and an exacerbation of sickness behaviour-like symptoms
cluster, ABCA7, CD2AP, BIN1 and EPHA1), the products and increased cognitive decline (Holmes et al., 2009, 2011).
of which are all postulated to be involved in immune sys- Several studies have also examined concentrations of
tem activation. A further large, two-stage meta-analysis of cytokines in the CSF of patients with MCI and AD. Here
AD (Lambert et al., 2013) has identified three further genes increased concentrations of the pro-inflammatory cytokine
involved in inflammatory processes including HLA-DRB5- TNF-α and decreased levels of the anti-inflammatory cyto-
CDRB1, INPP5D and MEF2C. kine TGF-β have been shown to be risk factors for increased
More recently, a rare genetic variant involved in innate rates of cognitive decline as well as conversion of MCI to
immunity with an effect size comparable to ApoE ε4 has AD (Tarkowski et al., 2003; Galimberti et al., 2008).
also been identified. Thus, two independent data sets have
identified a rare missense mutation in triggering receptor 49.3.3 IMAGING STUDIES
expressed on myeloid cells 2 (TREM2), which gives rise
to a threefold increase in the risk of AD (Guerreiro et al., Most imaging studies of inflammation have focused on the
2013; Jonsson et al., 2013). TREM2 is highly expressed on direct examination of microglial activation using transloca-
microglial cells and its expression is thought to suppress tor protein (TSPO) ligands. Cross-sectional positron emis-
pro-inflammatory cytokine production (Neumann and sion tomography (PET) studies support evidence of an up
Takahashi, 2007) suggesting that these variants confer a to 50% increased binding of 11C-PK11195 in the frontal and
heightened risk of AD by increasing the pro-inflammatory temporal cortex of AD (Diorio et al., 1991; Cagnin et al., 2001;
response. Edison et al., 2008; Okello et al., 2009; Venneti et al., 2009;
Thus, while early genetic studies found variation in Wiley et al., 2009) and an increase uptake of up to 33% of
genes affecting amyloid metabolism as being the key genetic 11C-DAA1106 in patients with AD (Yasuno et al., 2012). AD
Inflammation in Alzheimer’s disease 513

studies that have examined both microglial and Aβ markers clinical outcomes, but it has yet to be replicated (Butchart
have found significant correlations between cognitive scores et al., 2015).
and both TSPO ligands 11C-PK11195 and 11C-PBR28 (Kreisl Epidemiological studies of the protective effects of
et al., 2013) but not with the amyloid ligand Pittsburgh com- NSAIDs are more positive. Thus, the incidence of AD in
pound B (11C-PIB) binding (Edison et al., 2008; Yokokura cohorts of older people taking NSAIDs has been examined
et al., 2011) supporting the view that microglial activation in several large prospective studies (McGeer and McGeer,
rather than Aβ alone may be a key change leading to neuro- 2007). The largest of these prospective studies, the Baltimore
degeneration. The lack of correlation of 11C-PIB binding and Longitudinal Study of Aging, found a relative risk of AD
cognitive scores together with the failure to find a strong of 0.35 for 10 years of NSAID use (Stewart et al., 1997). A
relationship between the 11C-PK11195 microglial activation meta-analysis of case–control studies found that regular
marker and 11C-PIB binding (Edison et al., 2008; Okello NSAID use was associated with a twofold reduction in the
et al., 2009; Wiley et al., 2009; Yokokura et al., 2011) is sup- odds of developing AD (odds ratio [OR] = 0.5; p = 0.0002)
portive of the hypothesis that microglial activation follow- (McGeer et al., 1996) and, not included in this analysis,
ing the initial priming of microglial is independent of the the largest case–control study to date (49,349 cases and
continued presence of Aβ and may explain why removal of 196,850 matched controls) (Vlad et al., 2008) also showed
Aβ in established AD does not influence cognitive decline a significant reduction in odds of developing AD after
(Holmes et al., 2008). 5 years of regular use, with a combined OR of 0.76. However,
In MCI, the detection of inflammation is mixed. Thus, randomized placebo control trials examining the possible
one study using 11C-PK11195 detected inflammation in 40% protective effects seen in these epidemiological studies
of amnestic cases (Okello et al., 2009). An MCI study using are more mixed. Thus a large trial of rofecoxib examin-
11C-DAA1106 showed a 30% increase in uptake in the lat- ing conversion of MCI subjects to AD found an increased
eral temporal cortex compared with controls (Yasuno et al., risk of conversion in the MCI-treated group (Thal et al.,
2012). In this study, around 70% patients with MCI with 2005). Likewise a large randomized study of the NSAIDs
high 11C-DAA1106 uptake progressed to dementia during a naproxen and celecoxib in asymptomatic individuals with a
2-year follow-up period. family history of AD initially reported an increased risk of
Few studies have examined the effects of inflammation increased cognitive decline or development of AD for both
on other brain imaging markers in AD. However, interest- drugs (ADAPT Research Group et al., 2007). However, a
ingly one study has shown increased fibrillar Aβ deposi- longer-term follow-up of these patients suggests that care-
tion as shown by 11C-PIB in the brains of subjects carrying ful selection of asymptomatic patients and the choice of
the rs3865444 allele in the CD33 AD susceptibility locus the specific NSAID are important (Breitner et al., 2011).
(Bradshaw et al., 2013). Thus, the early detrimental effects were mostly in a small
group of patients with early cognitive impairment and
naproxen seemed thereafter to be protective in patients
for up to 4 years in those who had been asymptomatic at
49.4 THERAPEUTIC STRATEGIES baseline (Breitner et al., 2011; Alzheimer’s Disease Anti-
inflammatory Prevention Trial Research Group, 2013).
Given the increasing evidence for a role for inflammation in Although speculative, the reasons for these mixed find-
the development and progression of AD it is disappointing ings are likely to be found in the variability in the inflam-
to see mixed findings from a wide range of treatment stud- matory state as the disease progresses and in the ability of
ies aimed at modulating inflammatory pathways. agents to target the key pathways. Thus, early interventions
In AD, small early trials with indomethacin suggested prior to development of Aβ may benefit from approaches
some evidence of reduced cognitive decline (Rogers et al., aimed at dampening down the largely peripherally driven
1993). However, this study was not replicated in a later M1 activation state seen in inflammaging and thus reduc-
follow-up study (de Jong et al., 2008). Large-scale studies ing the drive towards Aβ deposition. However, once Aβ is
of other nonsteroidal anti-inflammatory drugs (NSAIDs) deposited and microglial cells are primed the high sensitiv-
including naproxen (Aisen et al., 2003) and rofecoxib ity of these cells to central and systemic pro-inflammatory
(Reines et al., 2004) in AD have also been unsuccessful. signals and the damaging nature of the activated primed
Randomized trials with a range of other anti-inflammatory microglial cell response is likely to require a peripheral
drugs, including prednisolone (Aisen et al., 2000), hydroxy- and/or centrally targeted robust suppression of the pro-
chloroquine (Van Gool et al., 2001), simvastatin (Simons inflammatory pathway to reduce neuronal damage at the
et al., 2002), atorvastatin (Sparks et al., 2005; Feldman potential expense of reducing Aβ phagocytosis. Finally, in
et al., 2010), aspirin (Bentham et al., 2008) and rosiglitazone the later stages of AD, when the inflammatory state more
(Harrington et al., 2011) have also shown no clinically sig- closely resembles an acquired deactivation state interven-
nificant changes in primary cognitive outcomes in patients tions aimed at reducing the pro-inflammatory drive or
with AD. More recently, a small study of AD subjects using promoting an anti-inflammatory drive are less likely to be
the TNF-α inhibiting agent etanercept in AD does show beneficial and other approaches, e.g. anti-apoptotic drugs,
some evidence of a reduction of decline on a number of may be needed.
514 Dementia

REFERENCES placebo-controlled, double-blind, phase 2 trial.


Neurology, 84 (21): 2161–2168.
ADAPT Research Group, Lyketsos, C.G., Breitner, J.C. Cagnin, A., Brooks, D.J., Kennedy, A.M. et al. (2001).
et al. (2007). Naproxen and celecoxib do not prevent In-vivo measurement of activated microglia in demen-
AD in early results from a randomized controlled trial. tia. Lancet, 358 (9280): 461–467.
Neurology, 68 (21): 1800–1808. Campbell, G.L. and Williams, M.P. (1978). In vitro growth
Aisen, P.S., Davis, K.L., Berg, J. D. et al. (2000). A ran- of glial cell-enriched and depleted populations from
domized controlled trial of prednisone in Alzheimer’s mouse cerebellum. Brain Research, 156 (2): 227–239.
disease. Alzheimer’s Disease Cooperative Study. Campion, D., Dumanchin, C., Hannequin, D. et al. (1999).
Neurology, 54: 588–593. Early-onset autosomal dominant Alzheimer disease:
Aisen, P.S., Schafer, K.A., Grundman, M. et al. (2003). Prevalence, genetic heterogeneity, and mutation spec-
Effects of rofecoxib or naproxen vs placebo on trum. The American Journal of Human Genetics, 65 (3):
Alzheimer disease progression: A randomized con- 664–670.
trolled trial. JAMA: The Journal of the American Casserly, I. and Topol, E. (2004). Convergence of athero-
Medical Association, 289: 2819–2826. sclerosis and Alzheimer’s disease: Inflammation, cho-
Alzheimer’s Disease Anti-inflammatory Prevention Trial lesterol, and misfolded proteins. Lancet, 363 (9415):
Research Group (2013). Results of a follow-up study to 1139–1146.
the randomized Alzheimer’s Disease Anti-inflammatory Chakrabarty, P., Ceballos-Diaz, C., Beccard, A. et al.
Prevention Trial (ADAPT). Alzheimer’s & Dementia: The (2010a). IFN-gamma promotes complement expression
Journal of the Alzheimer’s Association, 9 (6): 714–723. and attenuates amyloid plaque deposition in amyloid
Andersen, K., Lolk, A., Kragh-Sorensen, P. et al. (2005). beta precursor protein transgenic mice. The Journal of
Depression and the risk of Alzheimer disease. Immunology, 184 (9): 5333–5343.
Epidemiology, 16 (2): 233–238. Chakrabarty, P., Herring, A., Ceballos-Diaz, C. et al. (2011).
Bentham, P., Gray, R., Sellwood, E. et al. (2008). Aspirin in Hippocampal expression of murine TNFalpha results
Alzheimer’s disease (AD2000): A randomised open- in attenuation of amyloid deposition in vivo. Molecular
label trial. Lancet Neurology, 7 (1): 41–49. Neurodegeneration, 6 (1): 1–10.
Bianchi, M.E. (2007). DAMPs, PAMPs and alarmins: All Chakrabarty, P., Jansen-West, K., Beccard, A. et al.
we need to know about danger. Journal of Leukocyte (2010b). Massive gliosis induced by interleukin-6 sup-
Biology, 81 (1): 1–5. presses Abeta deposition in vivo: Evidence against
Blatteis, C.M., Bealer, S.L., Hunter, W.S. et al. (1983). inflammation as a driving force for amyloid deposition.
Suppression of fever after lesions of the anteroventral The FASEB Journal, 24 (2): 548–559.
third ventricle in guinea pigs. Brain Research Bulletin, Chakrabarty, P., Tianbai, L., Herring, A. et al. (2012).
11 (5): 519–526. Hippocampal expression of murine IL-4 results in
Boche, D., Perry, V.H. and Nicoll, J.A. (2013). Review: exacerbation of amyloid deposition. Molecular
Activation patterns of microglia and their identifica- Neurodegeneration, 7: 36.
tion in the human brain. Neuropathology and Applied Chan, W.Y., Kohsaka, S. and Rezaie, P. (2007). The origin
Neurobiology, 39 (1): 3–18. and cell lineage of microglia: New concepts. Brain
Bradshaw, E.M., Chibnik, L.B., Keenan, B.T. et al. (2013). Research Review, 53 (2): 344–354.
CD33 Alzheimer’s disease locus: Altered monocyte Choi, S.H., Aid, S., Kim, H.W. et al. (2012). Inhibition of
function and amyloid biology. Nature Neuroscience, 16 NADPH oxidase promotes alternative and anti-inflam-
(7): 848–850. matory microglial activation during neuroinflammation.
Breitner, J.C., Baker, L.D., Montine, T.J. et al. (2011). Journal of Neurochemistry, 120 (2): 292–301.
Extended results of the Alzheimer’s disease anti-inflam- Clark, I.A. and Atwood, C.S. (2011). Is TNF a link between
matory prevention trial. Alzheimer’s & Dementia: The aging-related reproductive endocrine dyscrasia and
Journal of the Alzheimer’s Association, 7 (4): 402–411. Alzheimer’s disease? Journal of Alzheimers Disease, 27
Buchhave, P., Zetterberg, H., Blennow, K. et al. (2010). (4): 691–699.
Soluble TNF receptors are associated with Abeta Colton, C.A. (2009). Heterogeneity of microglial activation
metabolism and conversion to dementia in subjects in the innate immune response in the brain. Journal of
with mild cognitive impairment. Neurobiology of Neuroimmune Pharmacology, 4 (4): 399–418.
Aging, 31 (11): 1877–1884. Cudic, M. and Otvos, L. Jr. (2002). Intracellular targets
Butchart, J., Birch, B., Bassily, R. et al. (2013). Male sex of antibacterial peptides. Current Drug Targets, 3 (2):
hormones and systemic inflammation in Alzheimer 101–106.
disease. Alzheimer Disease and Associated Disorders, Cunningham, C., Campion, S., Lunnon, K. et al. (2009).
27 (2): 153–156. Systemic inflammation induces acute behavioral and
Butchart, J., Brook, L., Hopkins, V. et al. (2015). cognitive changes and accelerates neurodegenerative
Etanercept in Alzheimer disease: A randomized, disease. Biological Psychiatry, 65 (4): 304–312.
Inflammation in Alzheimer’s disease 515

Cunningham, C., Wilcockson, D.C., Campion, S. et al. in humans. Mechanisms of Ageing and Development,
(2005). Central and systemic endotoxin challenges 128 (1): 92–105.
exacerbate the local inflammatory response and Galimberti, D., Fenoglio, C. and Scarpini, E. (2008).
increase neuronal death during chronic neurodegen- Inflammation in neurodegenerative disorders: Friend
eration. Journal of Neuroscience, 25 (40): 9275–9284. or foe? Current Aging Science, 1 (1): 30–41.
Dantzer, R., Konsman, J.P., Bluthe, R.M. and Kelley, K.W. Gentleman, S.M., Leclercq, P.D., Moyes, L. et al. (2004).
(2000). Neural and humoral pathways of communica- Long-term intracerebral inflammatory response after
tion from the immune system to the brain: Parallel traumatic brain injury. Forensic Science International,
or convergent? Autonomic Neuroscience, 85 (1–3): 146 (2–3): 97–104.
60–65. Ghosh, S., Wu, M.D., Shaftel, S.S. et al. (2013). Sustained
de Jong, D., Jansen, R., Hoefnagels, W. et al. (2008). No interleukin-1beta overexpression exacerbates tau
effect of one-year treatment with indomethacin on pathology despite reduced amyloid burden in an
Alzheimer’s disease progression: A randomized con- Alzheimer’s mouse model. Journal of Neuroscience, 33
trolled trial. PLoS ONE, 3 (1): e1475. (11): 5053–5064.
Diorio, D., Welner, S.A., Butterworth, R.F. et al. (1991). Ginhoux, F., Greter, M., Leboeuf, M. et al. (2010). Fate map-
Peripheral benzodiazepine binding sites in Alzheimer’s ping analysis reveals that adult microglia derive from
disease frontal and temporal cortex. Neurobiology of primitive macrophages. Science, 330 (6005): 841–845.
Aging, 12 (3): 255–258. Godbout, J.P. and Johnson, R.W. (2009). Age and neuroin-
Donath, M.Y. and Shoelson, S.E. (2011). Type 2 diabetes as flammation: A lifetime of psychoneuroimmune con-
an inflammatory disease. Nature Reviews Immunology, sequences. Immunology and Allergy Clinics of North
11 (2): 98–107. America, 29 (2): 321–337.
Dunn, N., Mullee, M., Perry, V.H. and Holmes, C. (2005). Griciuc, A., Serrano-Pozo, A., Parrado, A.R. et al. (2013).
Association between dementia and infectious disease: Alzheimer’s disease risk gene CD33 inhibits microglial
Evidence from a case-control study. Alzheimer Disease uptake of amyloid beta. Neuron, 78 (4): 631–643.
and Associated Disorders, 19 (2): 91–94. Guerreiro, R., Wojtas, A., Bras, J. et al. (2013). TREM2 vari-
Edison, P., Archer, H.A., Gerhard, A. et al. (2008). ants in Alzheimer’s disease. New England Journal of
Microglia, amyloid, and cognition in Alzheimer’s dis- Medicine, 368 (2): 117–127.
ease: An [11C](R)PK11195-PET and [11C]PIB-PET study. Hardy, J. (2006). Alzheimer’s disease: The amyloid cas-
Neurobiology Disease, 32 (3): 412–419. cade hypothesis: An update and reappraisal. Journal
Ek, M., Kurosawa, M., Lundeberg, T. and Ericsson, A. of Alzheimers Disease, 9 (Suppl. 3): 151–153.
(1998). Activation of vagal afferents after intravenous Harold, D., Abraham, R., Hollingworth, P. et al. (2009).
injection of interleukin-1beta: Role of endogenous Genome-wide association study identifies variants at
prostaglandins. Journal of Neuroscience, 18 (22): CLU and PICALM associated with Alzheimer’s disease.
9471–9479. Nature Genetics, 41 (10): 1088–1093.
Engelhart, M.J., Geerlings, M.I., Meijer, J. et al. (2004). Harrington, C., Sawchak, S., Chiang, C. et al. (2011).
Inflammatory proteins in plasma and the risk Rosiglitazone does not improve cognition or global
of dementia: The Rotterdam Study. Archives of function when used as adjunctive therapy to AChE
Neurology, 61 (5): 668–672. inhibitors in mild-to-moderate Alzheimer’s disease:
Farina, C., Aloisi, F. and Meinl, E. (2007). Astrocytes are Two phase 3 studies. Current Alzheimer Research, 8
active players in cerebral innate immunity. Trends in (5): 592–606.
Immunology, 28 (3): 138–145. Hart, B.L. (1988). Biological basis of the behavior of sick
Feldman, H.H., Doody, R.S., Kivipelto, M. et al. (2010). animals. Neuroscience and Biobehavioral Review, 12
Randomized controlled trial of atorvastatin in mild to (2): 123–137.
moderate Alzheimer disease: LEADe. Neurology, 74 Hollingworth, P., Harold, D., Sims, R. et al. (2011). Common
(12): 956–964. variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33
Feuerstein, G.Z., Wang, X. and Barone, F.C. (1998). The and CD2AP are associated with Alzheimer’s disease.
role of cytokines in the neuropathology of stroke Nature Genetics, 43 (5): 429–435.
and neurotrauma. Neuroimmunomodulation, 5 (3–4): Holmes, C., Boche, D., Wilkinson, D. et al. (2008). Long-
143–159. term effects of Abeta42 immunisation in Alzheimer’s
Field, R., Campion, S., Warren, C. et al. (2010). Systemic disease: Follow-up of a randomised, placebo-con-
challenge with the TLR3 agonist poly I:C induces trolled phase I trial. Lancet, 372 (9634): 216–223.
amplified IFNalpha/beta and IL-1beta responses in the Holmes, C., Cunningham, C., Zotova, E. et al. (2009).
diseased brain and exacerbates chronic neurodegen- Systemic inflammation and disease progression in
eration. Brain Behavior and Immunity, 24 (6): 996–1007. Alzheimer disease. Neurology, 73 (10): 768–774.
Franceschi, C., Capri, M., Monti, D. et al. (2007). Holmes, C., Cunningham, C., Zotova, E. et al. (2011).
Inflammaging and anti-inflammaging: A systemic per- Proinflammatory cytokines, sickness behavior, and
spective on aging and longevity emerged from studies Alzheimer disease. Neurology, 77 (3): 212–218.
516 Dementia

Honig, L.S., Tang, M.X., Albert, S. et al. (2003). Stroke and depression in th∆e general population: Systematic
the risk of Alzheimer disease. Archives of Neurology, review and meta-analysis. Lancet Neurology, 4 (6):
60 (12): 1707–1712. 371–380.
Hu, W.T., Holtzman, D.M., Fagan, A.M. et al. (2012). Lambert, J.C., Heath, S., Even, G. et al. (2009). Genome-
Plasma multianalyte profiling in mild cognitive impair- wide association study identifies variants at CLU and
ment and Alzheimer disease. Neurology, 79 (9): CR1 associated with Alzheimer’s disease. Nature
897–905. Genetics, 41 (10): 1094–1099.
Ishizuka, K., Kimura, T., Igata-yi, R. et al. (1997). Lambert, J.C., Ibrahim-Verbaas, C.A., Harold, D. et al.
Identification of monocyte chemoattractant protein-1 (2013). Meta-analysis of 74,046 individuals identifies 11
in senile plaques and reactive microglia of Alzheimer’s new susceptibility loci for Alzheimer’s disease. Nature
disease. Psychiatry and Clinical Neuroscience, 51 (3): Genetics, 45 (12): 1452–1458.
135–138. Launer, L.J., Andersen, K., Dewey, M.E. et al. (1999).
Jekabsone, A., Mander, P.K., Tickler, A. et al. (2006). Rates and risk factors for dementia and Alzheimer’s
Fibrillar beta-amyloid peptide Abeta1-40 activates disease: Results from EURODEM pooled analyses.
microglial proliferation via stimulating TNF-alpha EURODEM Incidence Research Group and Work
release and H2O2 derived from NADPH oxidase: A cell Groups. European Studies of Dementia. Neurology, 52
culture study. Journal of Neuroinflammation, 3: 24. (1): 78–84.
Jin, J.J., Kim, H.D., Maxwell, J.A. et al. (2008). Toll-like Laurin, D., David Curb, J., Masaki, K.H. et al. (2009).
receptor 4-dependent upregulation of cytokines in Midlife C-reactive protein and risk of cognitive decline:
a transgenic mouse model of Alzheimer’s disease. A 31-year follow-up. Neurobiology Aging, 30 (11):
Journal of Neuroinflammation, 5: 23. 1724–1727.
Jonsson, T., Stefansson, H., Steinberg, S. et al. Lee, W.L. and Slutsky, A.S. (2010). Sepsis and endothelial
(2013). Variant of TREM2 associated with the risk permeability. New England Journal of Medicine, 363
of Alzheimer’s disease. New England Journal of (7): 689–691.
Medicine, 368 (2): 107–116. Lue, L.F., Walker, D.G. and Rogers, J. (2001). Modeling
Kamer, A.R., Craig, R.G., Pirraglia, E. et al. (2009). TNF-alpha microglial activation in Alzheimer’s disease with human
and antibodies to periodontal bacteria discriminate postmortem microglial cultures. Neurobiology of
between Alzheimer’s disease patients and normal sub- Aging, 22 (6): 945–956.
jects. Journal of Neuroimmunology, 216 (1–2): 92–97. Lupetti, A., Nibbering, P.H., Welling, M.M. and Pauwels,
Koistinaho, M., Lin, S., Wu, X. et al. (2004). Apolipoprotein E.K. (2003). Radiopharmaceuticals: New antimicrobial
E promotes astrocyte colocalization and degrada- agents. Trends Biotechnology, 21 (2): 70–73.
tion of deposited amyloid-beta peptides. Nature of Matsumura, K. and Kobayashi, S. (2004). Signaling the
Medicine, 10 (7): 719–726. brain in inflammation: The role of endothelial cells.
Kono, H. and Rock, K.L. (2008). How dying cells alert Frontiers in Bioscience, 9: 2819–2826.
the immune system to danger. Nature Reviews McGeer, P.L. and McGeer, E.G. (2007). NSAIDs and
Immunology, 8 (4): 279–289. Alzheimer disease: Epidemiological, animal model
Koshinaga, M., Katayama, Y., Fukushima, M. et al. (2000). and clinical studies. Neurobiology of Aging, 28 (5):
Rapid and widespread microglial activation induced 639–647.
by traumatic brain injury in rat brain slices. Journal of McGeer, P.L., Schulzer, M. and McGeer, E.G. (1996).
Neurotrauma, 17 (3): 185–192. Arthritis and anti-inflammatory agents as possible pro-
Kountouras, J., Tsolaki, M., Gavalas, E. et al. (2006). tective factors for Alzheimer’s disease: A review of 17
Relationship between Helicobacter pylori infection epidemiologic studies. Neurology, 47 (2): 425–432.
and Alzheimer disease. Neurology, 66 (6): 938–940. Meda, L., Cassatella, M.A., Szendrei, G.I. et al. (1995).
Kourie, J.I. and Shorthouse, A.A. (2000). Properties of Activation of microglial cells by beta-amyloid protein
cytotoxic peptide-formed ion channels. American and interferon-gamma. Nature, 374 (6523): 647–650.
Journal of Physiology Cell Physiology, 278 (6): Medeiros, R. and LaFerla, F.M. (2013). Astrocytes:
C1063–C1087. Conductors of the Alzheimer disease neuroinflamma-
Kreisl, W.C., Lyoo, C.H., McGwier, M. et al. (2013). In vivo tory symphony. Experimental Neurology, 239: 133–138.
radioligand binding to translocator protein correlates Medzhitov, R. (2007). Recognition of microorganisms and
with severity of Alzheimer’s disease. Brain, 136 (Pt 7): activation of the immune response. Nature, 449 (7164):
2228–2238. 819–826.
Krstic, D., Madhusudan, A., Doehner, J. et al. (2012). Naj, A.C., Jun, G., Beecham, G.W. et al. (2011). Common
Systemic immune challenges trigger and drive variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1
Alzheimer-like neuropathology in mice. Journal of are associated with late-onset Alzheimer’s disease.
Neuroinflammation, 9: 151. Nature Genetics, 43 (5): 436–441.
Kuo, H.K., Yen, C.J., Chang, C.H. et al. (2005). Relation of Nathan, C., Calingasan, N., Nezezon, J. et al. (2005).
C-reactive protein to stroke, cognitive disorders, and Protection from Alzheimer’s-like disease in the mouse
Inflammation in Alzheimer’s disease 517

by genetic ablation of inducible nitric oxide syn- Rogers, J., Kirby, L.C., Hempelman, S.R. et al. (1993).
thase. The Journal of Experimental Medicine, 202 (9): Clinical trial of indomethacin in Alzheimer’s disease.
1163–1169. Neurology, 43 (8): 1609–1611.
Nau, R., Sorgel, F. and Eiffert, H. (2010). Penetration of Rovelet-Lecrux, A., Hannequin, D., Raux, G. et al. (2006).
drugs through the blood–cerebrospinal fluid/blood– APP locus duplication causes autosomal dominant
brain barrier for treatment of central nervous system early-onset Alzheimer disease with cerebral amyloid
infections. Clinical Microbiology Reviews, 23 (4): angiopathy. Nature Genetics, 38 (1): 24–26.
858–883. Savarin-Vuaillat, C. and Ransohoff, R.M. (2007).
Neumann, H. and Takahashi, K. (2007). Essential role Chemokines and chemokine receptors in neurological
of the microglial triggering receptor expressed on disease: Raise, retain, or reduce? Neurotherapeutics, 4
myeloid cells-2 (TREM2) for central nervous tissue (4): 590–601.
immune homeostasis. Journal of Neuroimmunology, Shiota, S., Murakami, K., Yoshiiwa, A. et al. (2011). The
184 (1–2): 92–99. relationship between Helicobacter pylori infection and
Ngandu, T., Lehtisalo, J., Solomon, A. et al. (2015). A Alzheimer’s disease in Japan. Journal of Neurology,
2 year multidomain intervention of diet, exercise, 258 (8): 1460–1463.
cognitive training, and vascular risk monitoring versus Simons, M., Schwarzler, F., Lutjohann, D. et al. (2002).
control to prevent cognitive decline in at-risk elderly Treatment with simvastatin in normocholesterolemic
people (FINGER): A randomised controlled trial. patients with Alzheimer’s disease: A 26-week random-
Lancet, 385 (9984): 2255–2263. ized, placebo-controlled, double-blind trial. Annals of
Nuutinen, T., Suuronen, T., Kauppinen, A. and Salminen, Neurology, 52 (3): 346–350.
A. (2009). Clusterin: A forgotten player in Alzheimer’s Sivanandam, T.M. and Thakur, M.K. (2012). Traumatic
disease. Brain Research Reviews, 61 (2): 89–104. brain injury: A risk factor for Alzheimer’s disease.
Okello, A., Edison, P., Archer, H.A. et al. (2009). Microglial Neuroscience Biobehavioral Reviews, 36 (5): 1376–
activation and amyloid deposition in mild cognitive 1381. Smits, H.A., Rijsmus, A., van Loon, J.H. et al.
impairment: A PET study. Neurology, 72 (1): 56–62. (2002). Amyloid-beta-induced chemokine produc-
Patel, N.S., Paris, D., Mathura, V. et al. (2005). tion in primary human macrophages and astrocytes.
Inflammatory cytokine levels correlate with amyloid Journal of Neuroimmunology, 127 (1–2): 160–168.
load in transgenic mouse models of Alzheimer’s dis- Sofroniew, M.V. and Vinters, H.V. (2010). Astrocytes:
ease. Journal of Neuroinflammation, 2 (1): 9. Biology and pathology. Acta Neuropathologica, 119
Perry, V.H. (2010). Contribution of systemic inflammation (1): 7–35.
to chronic neurodegeneration. Acta Neuropathologica, Soscia, S.J., Kirby, J.E., Washicosky, K.J. et al. (2010). The
120 (3): 277–286. Alzheimer’s disease-associated amyloid beta-protein is
Perry, V.H. and Holmes, C. (2014). Microglial prim- an antimicrobial peptide. PLoS ONE, 5 (3): e9505.
ing in neurodegenerative disease. Nature Reviews Sparks, D.L., Sabbagh, M.N., Connor, D.J. et al. (2005).
Neurology, 10 (4): 217–224. Atorvastatin for the treatment of mild to moderate
Perry, V.H., Nicoll, J.A. and Holmes, C. (2010). Microglia Alzheimer disease: Preliminary results. Archives of
in neurodegenerative disease. Nature Reviews Neurology, 62 (5): 753–757.
Neurology, 6 (4): 193–201. Stein, P.S., Desrosiers, M., Donegan, S.J. et al. (2007).
Radek, K. and Gallo, R. (2007). Antimicrobial peptides: Tooth loss, dementia and neuropathology in the Nun
Natural effectors of the innate immune system. study. The Journal of the American Dental Association,
Seminars in Immunopathology, 29 (1): 27–43. 138 (10): 1314–1322
Rahkonen, T., Luukkainen-Markkula, R., Paanila, S. et al. Stewart, W.F., Kawas, C., Corrada, M. and Metter, E.J.
(2000). Delirium episode as a sign of undetected (1997). Risk of Alzheimer’s disease and duration of
dementia among community dwelling elderly subjects: NSAID use. Neurology, 48 (3): 626–632.
A 2 year follow up study. Journal of Neurology and Stewart, C.R., Stuart, L.M., Wilkinson, K. et al. (2010).
Neurosurgery and Psychiatry, 69 (4): 519–521. CD36 ligands promote sterile inflammation through
Ramlackhansingh, A.F., Brooks, D.J., Greenwood, R.J. assembly of a Toll-like receptor 4 and 6 heterodimer.
et al. (2011). Inflammation after trauma: Microglial acti- Nature Immunology, 11 (2): 155–161.
vation and traumatic brain injury. Annals of Neurology, Streit, W.J. (2006). Microglial senescence: Does the brain’s
70 (3): 374–383. immune system have an expiration date? Trends
Reines, S.A., Block, G.A., Morris, J.C. et al. (2004). Neuroscience, 29 (9): 506–510.
Rofecoxib: No effect on Alzheimer’s disease in Streit, W.J., Sammons, N.W., Kuhns, A.J. and Sparks, D.L.
a 1-year, randomized, blinded, controlled study. (2004). Dystrophic microglia in the aging human brain.
Neurology, 62 (1): 66–71. Glia, 45 (2): 208–212.
Rivest, S. (2009). Regulation of innate immune responses Strohmeyer, R., Ramirez, M., Cole, G.J. et al. (2002).
in the brain. Nature Reviews of Immunology, 9 (6): Association of factor H of the alternative path-
429–439. way of complement with agrin and complement
518 Dementia

receptor 3 in the Alzheimer’s disease brain. Journal of Veerhuis, R., Nielsen, H.M. and Tenner, A.J. (2011).
Neuroimmunology, 131 (1–2): 135–146. Complement in the brain. Molecular Immunology, 48
Swardfager, W., Lanctot, K., Rothenburg, L. et al. (2010). (14): 1592–1603.
A meta-analysis of cytokines in Alzheimer’s disease. Venneti, S., Lopresti, B.J., Wang, G. et al. (2009). PK11195
Biological Psychiatry, 68 (10): 930–941. labels activated microglia in Alzheimer’s disease and
Tajiri, N., Kellogg, S.L., Shimizu, T. et al. (2013). Traumatic in vivo in a mouse model using PET. Neurobiology of
brain injury precipitates cognitive impairment and Aging, 30 (8): 1217–1226.
extracellular Abeta aggregation in Alzheimer’s disease Vlad, S.C., Miller, D.R., Kowall, N.W. and Felson, D.T.
transgenic mice. PLoS ONE, 8 (11): e78851. (2008). Protective effects of NSAIDs on the devel-
Tan, J., Town, T., Crawford, F. et al. (2002). Role of CD40 opment of Alzheimer disease. Neurology, 70 (19):
ligand in amyloidosis in transgenic Alzheimer’s mice. 1672–1677.
Nature Neuroscience, 5 (12): 1288–1293. Welling, M.M., Nabuurs, R.J. and van der Weerd, L. (2015).
Tan, J., Town, T., Paris, D. et al. (1999). Icroglial activation Potential role of antimicrobial peptides in the early
resulting from CD40–CD40L interaction after beta- onset of Alzheimer’s disease. Alzheimer’s & Dementia:
amyloid stimulation. Science, 286 (5448): 2352–2355. The Journal of the Alzheimer’s Association, 11 (1):
Tarkowski, E., Andreasen, N., Tarkowski, A. and Blennow, 51–57.
K. (2003). Intrathecal inflammation precedes devel- Wiley, C.A., Lopresti, B.J., Venneti, S. et al. (2009). Carbon
opment of Alzheimer’s disease. Journal Neurology 11-labeled Pittsburgh Compound B and carbon
Neurosurgery and Psychiatry, 74 (9): 1200–1205. 11-labeled (R)-PK11195 positron emission tomographic
Thal, L.J., Ferris, S.H., Kirby, L. et al. (2005). A randomized, imaging in Alzheimer disease. Archives of Neurology,
double-blind, study of rofecoxib in patients with mild 66: 60–67.
cognitive impairment. Neuropsychopharmacology, 30 Xia, M.Q., Qin, S.X., Wu, L.J. et al. (1998).
(6): 1204–1215. Immunohistochemical study of the beta-chemokine
Thambisetty, M. and Lovestone, S. (2010). Blood-based receptors CCR3 and CCR5 and their ligands in normal
biomarkers of Alzheimer’s disease: Challenging but and Alzheimer’s disease brains. American Journal of
feasible. Biomarkers in Medicine, 4 (1): 65–79. Pathology, 153: 31–37.
Tilvis, R.S., Kahonen-Vare, M.H., Jolkkonen, J. et al. Yasuno, F., Kosaka, J., Ota, M. et al. (2012). Increased
(2004). Predictors of cognitive decline and mortality binding of peripheral benzodiazepine receptor in mild
of aged people over a 10-year period. The Journals of cognitive impairment-dementia converters measured
Gerontology Series A: Biological Sciences and Medical by positron emission tomography with [11 C]DAA1106.
Sciences, 59 (3): 268–274. Psychiatry Research Neuroimaging, 203: 67–74.
Van Gool, W.A., Weinstein, H.C., Scheltens, P. and Yokokura, M., Mori, N., Yagi, S. et al. (2011). In vivo
Walstra, G.J. (2001). Effect of hydroxychloroquine on changes in microglial activation and amyloid deposits
progression of dementia in early Alzheimer’s disease: in brain regions with hypometabolism in Alzheimer’s
An 18-month randomised, double-blind, placebo-con- disease. European Journal of Nuclear Medicine and
trolled study. Lancet, 358 (9280): 455–460. Molecular Imaging, 38: 343–351.
50
Genetics of Alzheimer’s disease

MARGIE SMITH

family members having AD. However, both familial


50.1 INTRODUCTION and non-­familial cases of AD have the same clinical and
pathological presentations. Familial and non-familial AD
The genetic architecture of Alzheimer’s disease (AD) remains ­differ in relation to family history and molecular analysis.
elusive despite the success in identifying patients with rare The molecular and genetic basis of AD has been ­studied
autosomal dominant early-onset AD (EOAD). Mutations Rosenberg et al. (2000), Sleegers and Van Duijn (2001),
in three genes, the amyloid protein precursor (APP or AD1) Nussbaum and Ellis (2003), Goedert and Spillantini (2006)
and the presenilin genes (PSEN1 or AD3 and PSEN2 or and Roses and Saunders (2006).
AD4), located on chromosomes 21, 14 and 1, respectively, A diagnosis of late-onset familial AD (FAD) is estab-
exert their effect in a fully penetrant manner. lished with multiple cases of AD with an age of onset after
The gene that encodes apolipoprotein E (ApoE or AD2) the age of 60–65 years. Molecular genetic investigations
is the only genetic variant, which consistently influences support the concept that AD2 is a complex disorder that
disease risk and it is reported in association with late-onset may involve a large number of susceptibility genes. Bertram
AD (LOAD). Contrastingly, susceptibility for LOAD shows et al. (2008) reported the findings of meta-analysis of this
a less obvious familial aggregation and is, therefore, likely to data.
be governed by a number of risk alleles located on a number
of different genes. Continued research in this complex area
of the genes and proteins involved in AD will help establish 50.2.3 ApoE GENETICS AND LOAD
a logical basis for therapeutics and management.
The associations of the ApoE genotypes ε2/ε4, ε3/ε4 and ε4/
ε4 with LOAD have been verified in a number of studies
(Jarvik et al., 1996; Khachaturian et al., 2004; Martins et
50.2 HERITABLE CAUSES OF AD al., 2005).

50.2.1 CHROMOSOMAL ●● The ApoE ε4 allele shows the strongest association in


AD compared with normal population. The ε4 allele
All individuals with Down’s syndrome (trisomy 21) will occurs in 19% of FAD cases versus 1% found in the
develop AD after the age of 40 due to the lifelong overex- normal control population.
pression of APP located on chromosome 21, which encodes ●● The presence of the ApoE ε4/ε4 genotype in a patient
the amyloid precursor protein. The neuropathologic hall- with a clinical diagnosis of AD elevates the diagnostic
marks of AD presumably are due to the APP gene. certainty to 97%.
It was the observation that all individuals with Down’s ●● The ApoE genotype is not specific for AD since 42% of
syndrome develop AD that implied a relationship between persons with AD do not harbour an ApoE ε4 allele. In
chromosome 21 and AD pathology (Mann et al., 1984, 1989). summary, the absence of an ApoE ε4 allele does not rule
out the diagnosis of AD (Mayeux et al., 1998).
50.2.2 LATE-ONSET FAMILIAL AD (AD2) ●● The ApoE ε4 allele is associated with late-onset FAD
with those individuals harbouring an ε4 allele being
Molecular genetic investigations have revealed that 25% at a greater risk of developing AD at an earlier age
of LOAD is familial, which is defined as two or more (Khachaturian et al., 2004).
519
520 Dementia

●● The estimated lifetime risks for developing AD based of the 11 newly identified loci was significant since it rein-
on ApoE genotype and gender have been determined forced the importance of possible pathways thought to be
(Breitner et al., 2009). involved in the pathogenesis of AD.
●● The clinical utility of ApoE genotyping remains The candidate genes are involved in pathways that have
unclear with the presence of either one or two copies been reported to be enriched in AD, specifically inflamma-
of the ε4 allele being sufficient to establish a diagnosis tion and immune response (HLA-DRB5-DRB1, INPP5D
of AD. and MEF2C), cell migration (PTK2B), lipid transport and
●● The clinical utility of ApoE genotyping may be as an endocytosis (SORL1). It has been postulated that newly
adjunct to confirm a diagnosis of AD in those present- identified pathways include hippocampal synaptic func-
ing with the clinical symptoms of AD. tion (MEF2C and PTK2B), regulation of gene expression
(INPP5D) and cytoskeletal function and axonal transport
(CELF1, NME8 and CASS4).
50.2.4 OTHER GENETIC CONTRIBUTIONS
UNDER INVESTIGATION
50.3 MOLECULAR GENETICS OF EOAD
●● Researchers have simultaneously implicated the
p.Arg47His genetic variant in TREM2. The frequency of
this genetic variant in the general population is 0.5%–1%. The three known genetic subtypes of familial EOAD APP,
●● A2M is located on chromosome 12 (Dodel et al., 2000; PSEN1 and PSEN2 can only be distinguished by molecular
Gibson et al., 2000; Depboylu et al., 2006). genetic testing (Tsuang et al., 1999). A diagnosis of famil-
●● CALHM1 is located on chromosome 10q24 and has ial EOAD is made in families with multiple cases of AD in
a role in calcium homeostasis and a single nucleotide which the mean age of onset is 60–65 years.
polymorphism (SNP) has been reported to be associated Mutations in three genes, APP, PSEN1 and PSEN2 genes,
with LOAD (Dreses-Werringloer et al., 2008). alter the production of the amyloid-β (Aβ) peptide, the
●● A SNP has been identified in CD33 (Bertram et al., principal component of senile plaques (Tanzi and Bertram,
2008). CD33 that inhibits microglia uptake of amyloid 2005). In the 20 years since the discovery of the APP gene,
beta has been implicated in AD (Griciuc et al., 2013). it has only been relatively recently that the interaction with
●● Clusterin has been reported to collate with AD sever- the PSEN1 and PSEN2 genes has been elucidated. Aβ is gen-
ity, pathology and clinical progression. Confirmatory erated from APP by sequential cleavage of two enzymes:
genetic association studies supported the finding as β-secretase and β-secretase. All of the pathogenic mutations
well as CR1 and a phosphatidylinositol-binding clath- that are currently known to cause autosomal dominant
rin assembly protein (PICALM) (Harold et al., 2009; EOAD are located either in the APP gene itself or in genes
Lambert et al., 2009). that encode the proteins that are located in the catalytic cen-
●● GAB2 is located on chromosome 11q14 and ApoE ε4 tre of the β-secretase complex.
(Reiman et al., 2007). Unique pathologies not seen in the ‘sporadic’ or LOAD
●● GST01 and GST02 are located on chromosome 10 (Li et have been reported when these genes are mutated. The
al., 2003). historical review of each gene gives a perspective into the
●● PLD3 is on chromosome 19q13.2 (Cruchaga et al., 2014). understanding of EOAD, the phenotypes and the early
●● TOMM40 is located in close proximity to the ApoE understanding of the biochemical pathways involved. To
locus on chromosome 19 and has been implicated in molgen website has a catalogue of all of the reported muta-
relation to LOAD first by linkage analysis (Potkin et tions in the APP, PSEN1 and PSEN2 genes (http://www.
al., 2009) and second by the presence of poly-t variable molgen.ua.ac.be/AD).
length repeat region (Roses et al., 2010).
50.3.1 MUTATIONS IN THE APP GENE

50.2.5 OTHER CANDIDATE GENES The APP gene was implicated as a potential locus for AD
mutations based on four main intersecting lines of evidence.
GWAS meta-analysis has implicated the following can- First, it is well known that individuals with Down’s syn-
didate genes, some of which have been previously indi- drome (Trisomy 21) develop the pathological lesions of AD
cated, as risk factors for AD (Lambert et al., 2013). Eleven by the fourth decade. This evidence implied a relationship
new susceptibility loci were identified in addition to the between chromosome 21 and AD pathology (Mann et al.,
already known apolipoprotein E (ApoE), ATP-binding cas- 1984, 1989). Second, a 40- to 42-amino acid proteolytic pep-
sette sub-family A member (ABCA7), bridging integrator tide (Aβ) of the full-length APP is a major constituent of
1 (BIN1), clusterin (CLU), complement component (3b/4b) senile plaques (Masters et al., 1985; Goldgaber et al., 1987
receptor (CRI), CD2-associated protein (CD2AP), EPHA1, Kang et al., 1987). Genetic linkage studies have shown an
membrane-spanning 4-domains, subfamily A, member 6A additional line of evidence for linkage to a locus associated
(MS4A6A-MS4A4E) and PICALM genes. The importance with EOAD in the region of chromosome 21 containing the
Genetics of Alzheimer’s disease 521

APP gene (St George-Hyslop et al., 1987; Goate et al., 1989). Aβ42 (Suzuki et al., 1994), which is known to form insoluble
Finally, a missense mutation (APP E693Q) was identified in amyloid fibrils (Selkoe, 1994). Synthetic peptides contain-
the Aβ region of the APP gene characterized by hereditary ing the HCHWA mutation show accelerated fibril formation
cerebral haemorrhage with amyloidosis of the Dutch type (Wisniewski et al., 1991). The majority of APP mutation’s
(HCHWA-D) (Levy et al., 1990; Kamino et al., 1992). This is molecular effect is to increase Aβ42 production (Hardy,
a rare disorder caused by severe βA4 amyloid deposition in 1997); however, overexpression of the Val715Met mutation
meningeal and cerebral microvessels (Levy et al., 1990; Van in human HEK293 cells and murine neurones reduces the
Broeckhoven et al., 1990). It was then shown that E693G total Aβ production and increases the recovery of the physi-
mutation causes AD by increasing amyloid β-protofibril ologically secreted product, APPβ. It was initially thought
formation and decreasing amyloid β levels in plasma and that some cases of FAD were associated with a reduction of
conditioned media (Nilsberth et al., 2000). Three different in the overall production of Aβ; however, it was shown to be
mutations were reported at codon 693: the ‘Dutch’ muta- caused by an increased production of truncated forms of Aβ
tion (Glu22Gln), the Arctic Glu22Gly mutation and ‘Italian’ ending at the 42 position (Ancolio et al., 1999).
mutation (Glu22Lys). Peptides of the wild-type ‘Dutch’ To date, there have been 32 APP mutations identified in
and ‘Italian’ variants were made and their cytotoxic effects 86 kindreds. These mutations exert their effect by increas-
were evaluated in human cerebral endothelial cells in cul- ing Aβ production or Aβ42/Aβ40 ration. The mutations
ture. The effect of each mutation was different under these are located in the Aβ coding region or close to the β- and
conditions, the E22Q peptide exhibiting the highest con- β-secretase sites of APP (Hardy, 1997). Recently, three
tent of beta-sheet formation and aggregation properties. groups have reported a chromosome 21 genomic dupli-
In contrast, the ‘Dutch’ variant peptide induced apoptosis cation of unknown size. The phenotype is variable with
of cerebral endothelial cells. These results indicated that a congophillic amyloid angiopathy and Lewy body demen-
change in amino acid at position 22 confers distinct struc- tia being described (Rovelet-Lecrux et al., 2006, 2007;
tural properties on the peptides that affect the onset of the Sleegers et al., 2006). The duplications are of unknown
inexorable disease process (Miravalle et al., 2000). After the size and include not only the APP gene but also NCAM2,
initial finding of the E693Q mutation, a sequence variation NMRPL39, JAM2, ATPJ, GABPA, CYYR1, ADAMTS1 and
affecting the valine at codon 717 (position 46 relative to the ADAMTS5 genes or APP, MRPL39, JAM2, ATP5J, GABPA,
βA4 sequence) was found to be associated with the EOAD CYYR1, ADAMTS1, ADAMTS5, ZNF294, USP16, CCT8
phenotype. At least 13 families worldwide were identi- and BACH1 genes (Rovelet-Lecrux et al., 2006). The age of
fied with a V717I mutation, conferring an age of onset for onset and duration of the early-onset disease are variable
AD in the mid-fifties (Goate et al., 1991; Hardy et al., 1991; across the eight kindreds reported.
Naruse et al., 1991; Fidani et al., 1992; Sorbi et al., 1993, 1995;
Brooks et al., 1995; Campion et al., 1996; Matsumura et al., 50.3.2 MUTATIONS IN THE PSEN1 GENE
1996; Campion et al., 1999; Finckh et al., 2000). A ‘mixed’
phenotype of AD and/or severe cerebral amyloid angiopa- After the initial finding of an EOAD locus on chromosome
thy is caused by a mutation (A692G) at the βA4 residue 21 21 it became apparent that the majority of EOAD kindreds
adjacent to the E693Q HCHWA-D (Hendriks et al., 1992). did not show linkage to chromosome 21 (Tanzi et al., 1987;
This finding provided supporting evidence for the hypoth- Van Broeckhoven et al., 1987; Schellenberg et al., 1988;
esis that HCHWA-D and AD are pathological variants of St George-Hyslop et al., 1990). The PSEN1 gene was iden-
the same disease, being associated with a phenotype of AD tified in 1995 through positional cloning strategies. Five
or other βA4 amyloidosis-related disorders. A double muta- mutations were identified in 8 of 14 pedigrees analysed
tion (K670N and M671L) in a large Swedish EOAD kindred (Sherrington et al., 1995). All the mutations occurred within
with a mean age of onset of 55 years was identified (Mullan highly conserved domains of the PS1 protein as shown by
et al., 1992). More recently, four other APP mutations comparison to the murine homologue that provides sup-
have been identified, the Austrian (Thr714Ile) (De Jonghe et porting evidence that these mutations are indeed patho-
al., 2000), Florida (Ile716Val) (Eckman et al., 1997), French genic. Not all mutations occur at residues that are conserved
(Val715Met) (Ancolio et al., 1999) and the Australian between PSEN1 and PSEN2. The PSEN1 gene was previ-
(Leu723Pro) (Kwok et al., 2000a). ously denoted as the S182 or AD3 locus; to date 177 muta-
The Swedish double mutation was the first APP muta- tions have been detected in 392 kindreds in the PSEN1 gene.
tion for which a direct effect on βA4 processing could be The age of onset of AD due to PS1 mutations is accel-
demonstrated (Mullan et al., 1992). This double point muta- erated; however, variability in age onset is observed.
tion (the ‘Swedish’ mutation) Lys670Met and Asp671Leu The M139V and M146V mutations have an age of onset
is upstream of the β-secretase cleavage site and results in around 40 years (Clark et al., 1995; Van Broeckhoven, 1995),
a fivefold to eightfold increase in the formation of both whereas for kindreds identified with the E280 and C410Y
Aβ40 and Aβ42 (Citron et al., 1992). Two single-point muta- mutations the age of onset is between 45 and 50 (Clark et
tions at amino acid 717 (the ‘London’ mutation Val717Ile; al., 1995; Sherrington et al., 1995); the Ala409Thr muta-
and the ‘Indiana’ mutation Val717Phe) are adjacent to the tion has an onset of 85 years (Aldudo et al., 1999). EOAD
β-secretase site and specifically increase the production of linked to chromosome 14 is estimated to account for 70%
522 Dementia

of all EOAD pedigrees (St George-Hyslop et al., 1992; Van The normal biological and pathological functions of
Broeckhoven et al., 1992). PSEN1 were at that time poorly understood; however, there
The phenotype of PSEN1 mutations is heterogeneous. was ample evidence for a role of PSEN1 in the proteolytic
It was initially thought that the only difference between processing of APP and in Notch processing (Wolfe et al.,
AD and EOAD was the earlier age of onset (Sherrington et 1999). PSEN1 is endoproteolytically cleaved between amino
al., 1995); however, there are five reports of AD associated acids 291 and 299 resulting in an N-terminal fragment of 30
with spastic paraparesis (SP) of the lower limbs. The brain kDa and a C-terminal fragment of 18 kDa (Thinakaran et
pathology in one kindred was described as unique with al., 1996). The proteolytic cleavage site is located in that part
large plaques lacking the classical core of amyloid fibrils. of PSEN1 encoded by exon 9; therefore, the delta 9 mutant
The term ‘cotton wool’ was coined by virtue of their size does not undergo cleavage and accumulates as full-length
and appearance (Crook et al., 1998). PSEN1 mutations asso- PS1 (Thinakaran et al., 1996). Conversely, the efficiency
ciated with SP include a small deletion in exon 4 (DeltaI83/ of processing was shown not to be affected by missense
M84) (Steiner et al., 2001), an insertion of six nucleotides in mutations in AD brains (Hendriks et al., 1997), in brains
the coding region of exon 5 (from TAC to TTTATATAC) of transgenic mice (Duff et al., 1996) and in transfected
between amino acids K155 and Y156, point mutations in the cells (Borchelt et al., 1996). Cells transfected with PSEN1
coding region of exon 8 (Val261Phe) (Farlow et al., 2000) mutants (M146V, A246G, A260V, G384A, C410T) have
and (Arg278Thr) (Kwok et al., 1997), a point mutation in the impaired proteolysis (Mercken et al., 1996) and in lympho-
splice acceptor consensus of intron 8 resulting in in-frame cytes of mutation carriers (Takahashi et al., 1999). There is
skipping of exon 9 and an amino acid change at the splice no correlation between different PS1 mutations, age onset
junction of exon 8 and exon 10 (g.58304G. A, c.869–955del) and Aβ42 (43) production in transfected embryonic kidney
(Sato et al., 1998; Kwok et al., 2000b), (g.58304G. T, c.869– cells (Mehta et al., 1998).
955del) (Perez-Tur et al., 1995; Hutton et al., 1996; Kwok Overall, PSEN1 mutations increase the Aβ42/Aβ40 ratio.
et al., 1997, 2000b) and large genomic deletions (g.56305– The mutations are located throughout the protein and it has
62162del) (Kwok et al., 1997; Smith et al., 2001) identified in been shown to have an enzymatic role in the β-secretase
an Australian kindred and (g.56681–61235) the Delta9Finn complex. To date, 177 PSEN1 mutations have been identi-
kindred (Crook et al., 1998). Interestingly, a point muta- fied in 392 kindreds, (http://www.molgen.ua.ac.be/AD).
tion in the splice donor consensus site of intron 4 resulting
in three different transcripts with a single-codon inser- 50.3.3 MUTATIONS IN THE PS2 GENE
tion, partial and complete deletion of exon 4, respectively,
the latter two resulting in a frameshift and premature stop Directly after the identification of the PSEN1 cDNA
codon (g.23024delG, c.338–339insTAC, c.170–338del, c.88– sequence, ‘expressed sequence tags’ (ESTs) were described
338del) (Tysoe et al., 1998) is not associated with SP. It was with a considerable homology to two different segments
hypothesized that this mutation leads to AD through haplo- within the PSEN1 open reading frame (T03796). Cloning
insufficiency of full-length PSEN1. This is in direct contrast of the full-length complementary DNA (cDNA) of this cor-
to other PSEN1 mutations, which are thought to confer a responding gene, mapped to chromosome 1 enabled the
‘toxic’ gain of function in which Aβ42 secretion is selectively identification of missense mutations in cDNAs in affected
increased (Tysoe et al., 1998). Other phenotypes associated Volga-German subjects (Levy-Lahad et al., 1995). Evidence
with EOAD are frontotemporal features (Raux et al., 2000), of the physical genome mapping of PSEN2 on chromosome
myoclonic seizures (Ezquerra et al., 1999) and a psychiatric 1q42.1 was performed using fluorescence in situ hybridiza-
disorder (Tedde et al., 2000). tion (Tokano et al., 1997). The majority of kindreds analysed
Mutations are located throughout the PSEN1 protein; exhibited the N141I missense mutation, thereby identifying
however, their distribution may not be random. Mutations a founder effect. EOAD onset in these kindred’s generally
tend to occur at residues conserved between PSEN1 and occurs later than in EOAD kindreds linked to chromosome
PSEN2 and as such the majority are in transmembrane 14 but with a broader range of age at onset from 54.8 ± 8.4
domains. Specifically, the cluster of mutations in trans- years. The mean duration of disease is 11.3 ± 4.6 years and
membrane 1, 2, 3, 4 and 6 line up on one side of the β-helix. the mean age at death approximately 66 years. Clinically
This helical face may be important for the interaction with kindreds with PSEN1 mutations have a more aggressive
other transmembrane proteins (Clark et al., 1995). It was disease than those with PSEN2 mutations (Lampe et al.,
suggested that disruption of the helical faces impaired 1994). The wide variation in age of onset indicates that other
presenilin function so that more Aβ42 is produced (Hardy genetic and or environmental factors influence the age of
and Crook, 2001). The majority of PS1 mutations cluster in onset. Non-penetrance over the age of 80 years has also been
exon 8, which is close to the cleavage site. If this is a func- observed (Sherrington et al., 1996). The penetrance is over
tional domain then disruption of its function may be critical 95% for PSEN2 kindreds; however, pre-symptomatic indi-
to pathogenicity. The clustering of APP mutations around viduals harbouring the mutation over the age of 80 may
the β-secretase, β-secretase and β-secretase sites lead to dif- escape disease.
ferent molecular mechanisms; however, the outcome is the The exon 5 T122P mutation was found to be highly pen-
same. etrant unlike the previously reported PS2 mutations N141I
Genetics of Alzheimer’s disease 523

and M239V (Levy-Lahad et al., 1995). Consistent with the


variable penetrance of the N141I and M239V mutations, the 50.5 CONCLUSIONS
M239I mutation has associated phenotypic variability with
some affected individuals having an earlier onset and more Genetic studies of EOAD have had a significant impact on
severe course than the index case (Finckh et al., 2000). our understanding of the pathogenesis of AD that in turn
Fourteen mutations have been identified in 23 kindreds has led to research into diagnostic and therapeutic strate-
(http://www.molgen.ua.ac.be/AD). The majority of these gies and the availability of predictive testing of EOAD fami-
mutations are localized to exons 3, 5, 6, 7, 10 and 12 of the lies. However, as people with EOAD represent only 5% of all
PSEN2 gene. those with AD, some of the focus of genetic research has now
shifted towards LOAD. Currently in LOAD, there appears to
be a complex interplay of many risk factors, much as is seen
50.4 DIAGNOSTIC GENETIC TESTING in other pathogenic processes such as atherosclerosis. The
current complexity of this area undermines the role for risk
FOR AD
factor testing in LOAD. Understanding this genetic inter-
play is pivotal and likely to be the focus of genetic research
The identification of three genes causative for EOAD, in the next decade.
APP and recently PS1 and PS2 raises the question of the
role for genetic testing in AD. A consensus statement dis- REFERENCES
cussing the ethical aspects of the clinical introduction of
genetic testing for AD concluded, ‘except for autosomal Aldudo, J., Bullido, M.J. and Valdivieso, F. (1999). DGGE
dominant early-onset families, genetic testing in asymp- method for the mutational analysis of the coding and
tomatic individuals is unwarranted’. Predictive testing proximal promoter regions of the Alzheimer’s dis-
for EOAD was offered in research settings following the ease presenilin-1 gene: Two novel mutations. Human
identification of APP mutations on chromosome 21 using Mutation, 14: 433–439.
established Huntington’s disease guidelines (Lennox et al., Ancolio, K., Dumanchin, C., Barelli, H. et al. (1999) Unusual
1994). The experience of three individuals belonging to a phenotypic alteration of beta amyloid precursor
Swedish kindred with early-onset autosomal dominant protein (betaAPP) maturation by a new Val-715>Met
AD was documented. Only three members of this large betaAPP-770 mutation responsible for probable early-
kindred proceeded with predictive testing, indicating a onset Alzheimer’s disease. Proceedings of the National
low demand for testing. One individual who was found Academy of Science of the United States of America,
to harbour the mutation reacted with depressive and sui- 96: 4119–4124.
cidal feelings, while the other two subjects expressed relief Bertram, L., Lange, C., Mullin, K. et al. (2008). Genome-
(Lannfelt et al., 1995). wide association analysis reveals putative Alzheimer’s
The ApoE gene, which confers some susceptibility to disease susceptibility loci in addition to ApoE.
LOAD, has high commercial potential but the sensitivity American Journal of Human Genetics, 83: 623–632.
and specificity of the test are low. A asymptomatic individual Borchelt, D.R., Thinakaran, G., Eckman, C.B. et al. (1996).
with an ε4 allele will not necessarily develop AD. However, Familial Alzheimer’s disease-linked presenilin 1 vari-
testing for ApoE ε4 may be useful as an aid for the diagno- ants elevate Ab1–42/1–40 ratio in vitro and in vivo.
sis of individuals presenting with AD. Use of ApoE genetic Neuron, 17: 1005–1013.
testing as a diagnostic adjunct in patients already present- Borth, W. (1992). Alpha 2-macroglobulin, a multifunctional
ing with dementia may prove useful, but it remains under binding protein with targeting characteristics. FASEB
investigation. The premature introduction of genetic testing Journal, 6: 3345–3353.
and possible adverse consequences ‘are to be avoided’ (Post Breitner, J.C., Wyse, B.W., Anthony, J.C. et al. (2009).
et al., 1997). Athena Neurosciences in 1996 introduced the ApoE-epsilon4 count predicts age when prevalence of
Admark ApoE Genetic Test ‘for greater certainty in differ- AD increases, then declines: The Cache County Study.
ential diagnosis of AD’. The requesting physician must sign Neurology, 53: 321–331.
a consent stating that the individual has dementia prior to Brooks, W.S., Martins, R.N., De Voecht, J. et al. (1995). A
ApoE genotyping being performed. mutation in codon 717 of the amyloid precursor pro-
Predictive testing is performed when a known family tein in an Australian family with Alzheimer’s disease.
mutation has been identified in at least one other affected Neuroscience Letters, 199: 183–186.
family member. Pretest assessment and counselling involves Campion, D., Brice, A., Hannequin, D. et al. (1996). No
obtaining an accurate pedigree, neurological examination founder effect in three novel Alzheimer’s disease
(not compulsory) and psychological appraisal. This is criti- families with APP 717 Val-->Ile mutation. Journal of
cal to understanding the individual’s ability to cope with Medical Genetics, 33: 661–664.
the results of the test. EOAD contributes less than 5% of all Campion, D., Dumanchin, C., Hannequin, D. et al. (1999).
AD cases; therefore, there is no logical reason to perform Early-onset autosomal dominant Alzheimer’s disease:
genetic testing beyond this small cohort. Prevalence, genetic heterogeneity, and mutation
524 Dementia

spectrum. American Journal of Human Genetics, 65: early-onset dementia detected by sequence analyses
664–670. of four different genes. American Journal of Human
Citron, M., Oltersdorf, T., Haass, C. et al. (1992). Mutation Genetics, 66: 110–117.
of the beta-amyloid precursor protein in familial Gibson, A.M., Singleton, A.B., Smith, G. et al. (2000). Lack
Alzheimer’s disease increases b-protein production. of association of the alpha2-macroglobulin locus on
Nature, 360: 672–674. chromosome 12 in AD. Neurology, 54:433–438.
Clark, R.F., Hutton, M., Fuldner, R.A. et al. (1995). The Goate, A., Chartier, H.M., Mullan, M. et al. (1991).
structure of the presenilin 1 gene and identification of Segregation of a missense mutation in the amyloid
six novel mutations in early onset AD families. Nature precursor protein gene with familial Alzheimer’s dis-
Genetics, 11: 219–222. ease. Nature, 349: 704–706.
Crook, R., Verkkoniemi, A., Perez-Tur, J. et al. (1998). A Goate, A., Haynes, A.R., Owen, M.J. et al. (1989).
variant of Alzheimer’s disease with spastic parapare- Predisposing locus for Alzheimer disease on chromo-
sis and unusual plaques due to deletion of exon 9 of some 21. Lancet, 1: 352–355.
presenilin. Nature Medicine, 4: 452–455. Goedert, M. and Spillantini, M.G. (2006). A century of
Cruchaga, C., Karch, C.M., Jin, S.C. et al. (2014). Rare cod- Alzheimer’s disease. Science, 314: 777–781.
ing variants in the phospholipase D3 gene confer risk Goldgaber, D., Lerman, M.I. and McBride, O.W. (1987).
for Alzheimer’s disease. Nature, 505: 550–554. Characterisation and chromosomal localization of a
De Jonghe, C., Kumar-Singh, S., Cruts, M. et al. (2000). cDNA encoding brain amyloid of Alzheimer disease on
Unusual Ab amyloid in Alzheimer’s disease due to chromosome 21. Science, 235: 877–880.
an APP T7141 mutation at the g42-secretase site. Griciuc, A., Serrano-Pozo, A. and Parrado, A.R. (2013).
Neurobiology of Aging, 21 (Suppl. 1): S200. Alzheimer’s disease risk gene CD33 inhibits microglial
Depboylu, C., Lohmuller, F., Du, Y. et al. (2006). Alpha2- uptake of amyloid beta. Neuron, 78: 631–43.
macroglobulin, lipoprotein receptor-related protein Hardy, J. (1997). Amyloid, the presenilins and Alzheimer’s
and lipoprotein receptor-associated protein and disease. Trends in Neuroscience, 20: 154–159.
the genetic risk for developing Alzheimer’s disease. Hardy, J. and Crook, R. (2001). Presenilin mutations line up
Neuroscience Letters, 400: 187–190. along transmembrane a-helices. Neuroscience Letters,
Dodel, R.C., Du, Y., Bales, K.R. et al. (2000). Alpha2 306: 203–205.
macroglobulin and the risk of Alzheimer’s disease. Hardy, J., Mullan, M., Chartier-Harlin, M.C. et al. (1991).
Neurology, 54: 438–442. Molecular classification of Alzheimer’s disease. Lancet,
Dreses-Werringloer, U., Lambert, J.C., Vingtdeux, V. et al. 337: 1342–1343.
(2008). A polymorphism in CALHM1 influences Ca2+ Harold, D., Abraham, R. and Hollingworth, P. (2009).
homeostasis, Abeta levels, and Alzheimer’s disease Genome-wide association study identifies variants at
risk. Cell, 133: 1149–1161. CLU and PICALM associated with Alzheimer’s disease.
Duff, K., Eckman, C., Zehr, C. et al. (1996). Increased Nature Genetics, 41: 1088–1093.
­amyloid-beta42(43) in brains of mice expressing Hendriks, L., Thinakaran, G., Harris, C.L. et al. (1997)
mutant presenilin 1. Nature, 383: 710–713. Processing of presenilin 1 in brains of patients with
Eckman, C.B., Mehta, N.D., Crook, R. et al. (1997). A new Alzheimer’s disease and controls. NeuroReport, 8:
pathogenic mutation in the APP gene (I716V) increases 1717–1721.
the relative proportion of A beta 42(43) Human Hendriks, L., van Duijn, C., Cras, P. et al. (1992). Presenilin
Molecular Genetics, 12: 2087–2089. dementia and cerebral haemorrhage linked to a muta-
Ezquerra, M., Carnero, C., Blesa, R. et al. (1999). A pre- tion at codon 692 of the B-amyloid precursor protein
senilin 1 mutation (Ser169Pro) associated with early- gene. Nature Genetics, 1: 218–221.
onset AD and myoclonic seizures. Neurology, 52: Hutton, M., Busfield, F., Wragg, M. et al. (1996). Complete
566–570. analysis of the presenilin 1 gene in early onset
Farlow, M.R., Murrell, J.R., Hulette, C.M. et al. (2000). Alzheimer’s disease. NeuroReport, 7: 801–805.
Hereditary lateral sclerosis and Alzheimer disease Jarvik, G., Larson, E.B., Goddard, K. et al. (1996). Influence
associated with mutation at codon 261 of the prese- of apolipoprotein E genotype on the transmission
nilin 1 (PS1) gene. Neurobiology of Aging, 21 (Suppl.): of Alzheimer disease in a community-based sample.
S62. American Journal of Human Genetics, 58: 191–200.
Fidani, L., Rooke, K., Chartier-Harlin, M. et al. (1992). Kamino, K., Orr, H.T., Payami, H. et al. (1992). Linkage
Screening for mutations in the open reading frame and mutational analysis of familial Alzheimer disease
and promoter of the b-amyloid precursor protein in kindreds for the APP gene region. American Journal of
familial Alzheimer’s disease: Identification of a further Human Genetics, 51: 998–1014.
family with APP Val-->Ile. Human Molecular Genetics, Kang, J., Lemaire, H., Unterbeck, A. et al. (1987). The
1: 165–168. precursor of Alzheimer’s disease amyloid A4 pro-
Finckh, U., Muller-Thomsen, T., Mann, U. et al. (2000). High tein resembles a cell-surface receptor. Nature, 325:
prevalence of pathogenic mutations in patients with 733–736.
Genetics of Alzheimer’s disease 525

Khachaturian, A.S., Corcoran, C.D., Mayer, L.S. et al. Mann, D.M.A., Yates, P.O. and Marcyniuk, B. (1984).
(2004). Apolipoprotein E epsilon4 count affects age Alzheimer’s presenile dementia, senile dementia,
at onset of Alzheimer disease, but not lifetime suscep- of the Alzheimer type, and Down’s syndrome in
tibility: The Cache County Study. Archives of General middle age form a continuum of pathologic changes.
Psychiatry, 61: 518–524. Neuropathology and Applied Neurobiology, 10:
Kwok, J.B.J., Li, Q.-X., Hallup, M. et al. (2000a). Novel 188–207.
Leu723Pro amyloid precursor protein mutation Martins, C.A., Oulhaj, A., de Jager, C.A. et al. (2005).
increases amyloid beta42(43) peptide levels and ApoE alleles predict the rate of cognitive decline in
induces apoptosis. Annals of Neurology, 47: 249–253. Alzheimer disease: A nonlinear model. Neurology, 65:
Kwok, J.B.J., Smith, M.J., Brooks, W.S. et al. (2000b). 1888–1893.
Variable presentation of Alzheimer’s disease and/or Masters, C.L., Simms, G., Weinman, N.A. et al. (1985).
spastic paraparesis phenotypes in pedigrees with a Amyloid plaque core protein in Alzheimer disease
novel PS-1 exon 9 deletion or exon 9 splice acceptor and Down syndrome. Proceedings of the National
mutations. Neurobiology of Aging, 21 (Suppl.): S25. Academy of Sciences of the United States of America,
Kwok, J.B., Taddei, K., Hallupp, M. et al. (1997). Two novel 82: 4245–4249.
(M233T and R278T) presenilin-1 mutations in early- Matsumura, Y., Kitamura, E., Miyoshi, K. et al. (1996).
onset Alzheimer’s disease pedigrees and preliminary Japanese siblings with missense mutation (717Val-
evidence for association of presenilin-1 mutations with ->Ile) in amyloid precursor protein of early-onset
a novel phenotype. NeuroReport, 8: 1537–1542. Alzheimer’s disease. Neurology, 46: 1721–1723.
Lambert, J.-C., Health, S., Even, G. et al. (2009). Genome- Mayeux, R., Saunders, A.M., Shea, S. et al. (1998). Utility
wide association study identifies variants at CLU and of the apolipoprotein E genotype in the diagnosis
CR1 associated with Alzheimer’s disease. Nature of Alzheimer’s disease. Alzheimer’s Disease Centers
Genetics, 14: 1094–1099. Consortium on Apolipoprotein E and Alzheimer’s
Lambert, J.-C., Ibrahim-Verbaas, C.A., Harold, D. et al. Disease. New England Journal of Medicine, 338:
(2013). Meta-analysis of 74,046 individuals identifies 11 506–511.
new susceptibility loci for Alzheimer’s disease. Nature Mehta, N.D., Refolo, L.M., Eckman, C. et al. (1998).
Genetics, 45: 1452–1458. Increased Abeta42(43) from cell lines expressing
Lampe, T.H., Bird, T.D., Nochlin, D. et al. (1994). ­presenilin 1 mutations. Annals of Neurology, 43:
Phenotype of chromosome 14-linked familial 256–258.
Alzheimer’s disease in a large kindred. Annals of Mercken, M., Takahashi, H., Honda, T. et al. (1996).
Neurology, 36: 368–378. Characterisation of human presenilin 1 using
Lannfelt, H., Axelman, K., Lilius, L. and Basun H. (1995). N-terminal specific monoclonal antibodies: Evidence
Genetic counseling of a Swedish Alzheimer family with that Alzheimer mutations affect proteolytic process-
amyloid precursor protein mutation. American Journal ing. FEBS Letters, 389: 297–303.
of Human Genetics, 56: 332–335. Miravalle, H., Tokuda, T., Chiarle, R. et al. (2000).
Lennox, A., Karlinsky, H., Meschino, W. et al. (1994). Substitutions at codon 22 of Alzheimer’s A-beta
Molecular genetic predictive testing for Alzheimer dis- peptide induce diverse conformational changes and
ease: Deliberations and preliminary recommendations. apoptotic effects in human cerebral endothelial cells.
Alzheimer Disease and Associated Disorders, 8: 126–147. Journal of Biological Chemistry, 275: 27110–27116.
Levy, E., Carman, M.D., Fernandez-Madrid, I.J. et al. Mullan, M., Crawford, F., Axelman, K. et al. (1992). A
(1990). Mutation of the Alzheimer’s disease amyloid pathogenic mutation for probable Alzheimer’s disease
gene in hereditary cerebral hemorrhage, Dutch Type. in the APP gene at the N-terminus of b-amyloid.
Science, 248: 1124–1126. Nature Genetics, 1: 345–347.
Levy-Lahad, E., Wijsman, E.M., Nemens, E. et al. (1995). A Naruse, S., Igarashi, S., Aoki, K. et al. (1991). Mis-sense
familial Alzheimer’s disease locus on chromosome 1. mutation Val to ILE in exon 17 of amyloid precur-
Science, 269: 970–973. sor protein in Japanese familial Alzheimer’s disease.
Li, Y.J., Oliveira, S.A., Xu, P. et al. (2003). Glutathione Lancet, 337: 978–979.
S-transferase omega-1 modifies age-at-onset of Nilsberth, C., Westlind-Danielsson, A., Eckman, C.B.
Alzheimer disease and Parkinson disease. Human et al. (2000). The Arctic mutation (E693G) causes
Molecular Genetics, 12: 3259–3267. Alzheimer’s disease through a novel mechanism:
Mann, D.M.A., Brown, A., Prinja, D. et al. (1989). An Increased amyloid b protofibril formation and
analysis of the morphology of senile plaques in Down’s decreased amyloid b levels in plasma and conditioned
syndrome patients of different ages using immuno- media. Neurobiology of Aging, 21 (Suppl. 1): S58.
cytochemical and lectin histochemical techniques. Nussbaum, R.L. and Ellis, C.E. (2003). Alzheimer’s disease
Neuropathology and Applied Neurobiology, 15: and Parkinson’s disease. New England Journal of
317–329. Medicine, 348: 1356–1364.
526 Dementia

Perez-Tur, J., Froelich, S., Prihar, G. et al. (1995). A muta- markers to familial Alzheimer’s disease. Science, 241:
tion in Alzheimer’s disease destroying a splice accep- 1507–1510.
tor site in the presenilin-1 gene. NeuroReport, 7: Selkoe, D. (1994). Cell biology of the amyloid beta-protein
297–301. precursor and the mechanism of Alzheimer’s disease.
Post, S.G., Whitehouse, P.J., Binstock, R.H. et al. (1997). Annual Review of Cell Biology, 10: 373–403.
The clinical introduction of genetic testing for Sherrington, R., Froelich, S., Sorbi, S. et al. (1996).
Alzheimer disease. JAMA: The Journal of the American Alzheimer’s disease associated with mutations in
Medical Association, 277: 832–836. presenilin 2 is rare and variably penetrant. Human
Potkin, S.G., Guffanti, G., Lakatos, A. et al. (2009). Molecular Genetics, 5: 985–988.
Alzheimer’s disease neuroimaging initiative; hippo- Sherrington, R., Rogaev, E.I., Liang, Y. et al. (1995). Cloning
campal atrophy as a quantitative trait in a genome- a gene bearing missense mutations in early-onset
wide association study identifying novel susceptibility familial Alzheimer’s disease. Nature, 375: 754–760.
genes for Alzheimer’s disease. PLoSONE, 4: e6501. Sleegers, K., Brouwers, N., Gijselinck, I. et al. (2006).
Raux, G., Gantier, R., Thomas-Anterion, C. et al. (2000). APP duplication is sufficient to cause early onset
Dementia with prominent frontotemporal fea- Alzheimer’s dementia with cerebral amyloid angiopa-
tures associated with L113P presenilin 1 mutation. thy. Brain, 129: 2977–2983.
Neurology, 55: 1577–1579. Sleegers, K. and Van Duijn, C.M. (2001). Alzheimer’s
Reiman, E.M., Webster, J.A., Myers, A.J. et al. (2007). disease: Genes, pathogenesis and risk prediction.
GAB2 alleles modify Alzheimer’s risk in ApoE varepsi- Community Genetics, 4: 197–203.
lon4 carriers. Neuron, 54: 713–720. Smith, M.J., Kwok, J.B.J., McLean, C.A. et al. (2001).
Rosenberg, R.N. (2000). The molecular and genetic Variable phenotype of Alzheimer’s disease with spastic
basis of AD: The end of the beginning: The 2000 paraparesis. Annals of Neurology, 49: 125–129.
Wartenberg lecture. Neurology, 54: 2045–2054. Sorbi, S., Nacmias, B., Forleo, P. et al. (1993). APP717 and
Roses, A., Lutz, M., Amrine-Madsen, H. et al. (2010). Alzheimer’s disease in Italy. Nature Genetics, 4: 10.
A TOMM 40 variable length polymorphism deter- Sorbi, S., Nacmias, B., Forleo, P. et al. (1995). Epistatic
mines the age of late-onset Alzheimer’s disease. effect of APP717 mutation and apolipoprotein E
Pharmacogenomics Journal, 10: 375–384. genotype in familial Alzheimer’s disease. Annals of
Roses, A.D. and Saunders, A.M. (2006). Perspective on a Neurology, 38: 124–127.
pathogenesis and treatment of Alzheimer’s disease. Steiner, H., Revesz, T., Neumann, M. et al. (2001). A
Alzheimers Dementia, 2: 59–70. pathogenic presenilin-1 deletion causes aberrant
Rovelet-Lecrux, A., Hannequin, D., Raux, G. et al. (2006). Ab42 production in the absence of congophilic
APP locus duplication causes autosomal dominant amyloid plaques. Journal of Biological Chemistry, 276:
early-onset Alzheimer disease with cerebral amyloid 7233–7239.
angiopathy. Nature Genetics, 38: 24–26. Suzuki, N., Cheung, T.T., Cai, X.D. et al. (1994). An
Rovelet-Lecrux, A., Frebourg, T., Tuominen, H. et al. increased percentage of long amyloid b protein
(2007). APP locus duplication in a Finnish family with secreted by familial amyloid b protein precursor
dementia an intracerebral haemorrhage. Journal (b APP717) mutants. Science, 264: 1336–1340.
of Neurology, Neurosurgery and Psychiatry, 78: Takahashi, H., Mercken, M., Honda, T. et al. (1999).
1158–1159. Impaired proteolytic processing of presenilin-1 in
Sato, S., Kamino, K., Miki, T. et al. (1998). Splicing muta- ­chromosome 14-linked familial Alzheimer’s disease
tion of presenilin-1 gene for early-onset familial patient lymphocytes. Neuroscience Letters, 260:
Alzheimer’s disease. Human Mutation, 11 (Suppl. 1): 121–124.
S91–S94. Tanzi, R.E. and Bertram, L. (2005). Twenty years of the
St George-Hyslop, P.H., Haines, J.L., Farrer, L.A. et Alzheimer’s disease amyloid hypothesis: A genetic
al. (1990). Genetic linkage studies suggest that perspective. Cell, 120: 545–555.
Alzheimer’s disease is not a single homogenous disor- Tanzi, R.E., St George-Hyslop, P.H., Haines, J.L. et al.
der. Nature, 347: 194–197. (1987). The genetic defect in familial Alzheimer’s
St George-Hyslop, P.H., Haines, J., Rogaev, E. et al. (1992). disease is not tightly linked to the amyloid b-protein
Genetic evidence for a novel familial Alzheimer’s gene. Nature, 329: 156–157.
disease locus on chromosome 14. Nature Genetics, 2: Tedde, A., Forleo, P., Nacmias, B. et al. (2000). A prese-
330–334. nilin 1 mutation (Leu392Pro) in a familial AD kindred
St George-Hyslop, P.H., Tanzi, R.E., Polinsky, R.J. et al. with psychiatric symptoms at onset. Neurology, 55:
(1987). The genetic defect causing familial Alzheimer 1590–1591.
disease maps on chromosome 21. Science, 235: Thinakaran, G., Borchelt, D.R., Lee, M.K. et al. (1996).
885–859. Endoproteolysis of presenilin 1 and accumula-
Schellenberg, G.D., Bird, T.D., Wijsman, E.M. et al. tion of processed derivatives in vivo. Neuron,
(1988). Absence of linkage of chromosome 21q21 17: 181–190.
Genetics of Alzheimer’s disease 527

Tokano, T., Sahara, N., Yamanouchi, Y. and Mori, H. Van Broeckhoven, C., Genthe, A.M., Vandenberghe, A. et
(1997). Assignment of Alzheimer’s presenilin-2 (PS-2) al. (1987). Failure of familial Alzheimer’s disease to seg-
gene to 1q42.1 by fluorescence in situ hybridisation. regate with the A4-amyloid gene in several European
Neuroscience Letters, 221: 205–207. families. Nature, 329: 153–155.
Tsuang, D., Larson, E.B., Bowen, J. et al. (1999). The utility Van Broeckhoven, C., Hann, J., Bakker, E. et al. (1990).
of apolipoprotein E genotyping in the diagnosis of Amyloid beta protein precursor gene and heredi-
Alzheimer disease in a community-based case series. tary cerebral hemorrhage with amyloidosis (Dutch).
Archives of Neurology, 56: 1489–1495. Science, 248: 1120–1122.
Tysoe, C., Whittaker, J., Xuereb, J. et al. (1998). A Wisniewski, T., Ghiso, J. and Frangione, B. (1991).
Presenilin-1 truncating mutation is present in two Peptides homologous to the amyloid protein of
cases with autopsy-confirmed early-onset Alzheimer Alzheimer’s disease containing a glutamine for
disease. American Journal of Human Genetics, 62: glutamic acid substitution have accelerated amyloid
70–76. fibril formation. Biochemical and Biophysical Research
Van Broeckhoven, C. (1995). Presenilins and Alzheimer’s Communications, 179: 1247–1254.
disease. Nature Genetics, 11: 230–232. Wolfe, M.S., Xia, W., Moore, C.L. et al. (1999).
Van Broeckhoven, C., Backhovens, H., Cruts, M. et al. Peptidomimetic probes and molecular modeling
(1992). Mapping of a gene predisposing to early-onset suggest Alzheimer’s g-secretases are intramembrane-­
Alzheimer’s disease to chromosome 14q24.3. Nature cleaving aspartyl proteases. Biochemistry, 38:
Genetics, 2: 335–339. 4720–4727.
51
Blood and cerebrospinal fluid biomarkers for
Alzheimer’s disease

SIMONE LISTA, HENRIK ZETTERBERG, SID E. O’BRYANT, KAJ BLENNOW AND HARALD
HAMPEL

(The Ronald and Nancy Reagan Research Institute of the


51.1 ROLE OF BIOMARKERS IN THE Alzheimer’s Association and National Institute on Aging
DIAGNOSIS AND TREATMENT OF Working Group, 1998). Although, several AD biomarker
ALZHEIMER’S DISEASE studies have been published, the clinical relevance and fea-
sibility of the investigated biomarkers are not completely
In general, a biomarker defines a biological process or dis- elucidated (Buerger and Hampel, 2009; O’Bryant, 2012;
ease characteristic that is objectively measured (Biomarkers Blennow et al., 2015a). A number of systematic development,
Definitions Working Group, 2001). Such measurements validation and standardization steps are required before a
may be used for diagnostic purposes, but also to study phys- biomarker becomes a reliable and precise asset to clinicians.
iological or pathophysiological mechanisms and to evaluate This process is as risky and may take as long as the devel-
desired pharmacodynamic effects or side effects of pharma- opment period of a pharmacological compound in the neu-
cological treatments. According to Biomarkers Definitions roscience sector moving from preclinical stages to Phases 1–3
Working Group: ‘Molecular and Biochemical Markers of and finally regulatory approval. For a long time, this develop-
Alzheimer’s Disease’, the ideal biomarker for Alzheimer’s ment has been primarily done in academic institutions and
disease (AD) should detect a fundamental feature of neu- consortia, mostly independent from industry-led drug devel-
ropathology and be validated in neuropathologically con- opments. In the advent of many failed trial programmes, the
firmed cases as well as have a diagnostic accuracy of at least significance of a parallel development of both compounds
80% (The Ronald and Nancy Reagan Research Institute and biomarkers through all development stages is begin-
of the Alzheimer’s Association and National Institute on ning to be appreciated. More than a decade of fragmented
Aging Working Group, 1998). With respect to clinically work has substantially slowed and weakened drug develop-
relevant questions, such as detection, diagnosis, prediction ment within the neuroscience sector, particularly, in the AD
and treatment of a given disease, biomarkers may serve cer- indication with the lowest number of successful regulatory
tain distinct functions (Hampel et al., 2010b; Hampel and approvals. Since the field is moving into earlier disease stages,
Lista, 2013), which are detailed in Table 51.1. prodromal forms and asymptomatic at risk stages, with no
clinical signs or symptoms, biomarker development is gain-
ing all the more importance in evolving trial designs.
First, the technical feasibility of the new marker has to
be established, including the availability of a validated assay
51.2 BIOMARKER DEVELOPMENT with high precision and reliability of measurement and well-
described reagents and standards. A relatively large number
Ideally, a biomarker should be reliable, reproducible, non- of potential markers have successfully passed this first step.
invasive, simple to perform and inexpensive. Recommended Second, the possible marker has to be evaluated in a rela-
steps to establish a biomarker include confirmation by at tively pure sample of diseased and comparison groups. This
least two independent studies conducted by qualified inves- is similar to the Phase 2 trial in therapeutics, but the goal
tigators with the results published in peer-reviewed journals here is to make an initial assessment of its sensitivity and

528
Blood and cerebrospinal fluid biomarkers for Alzheimer’s disease 529

Table 51.1 The same marker may subserve different listed functions, or it might be diagnostic and also be a useful
screening tool, or it may be both predictive and prognostic, or it can be used for monitoring purposes. It is also possible
that different biomarkers will be needed for different purposes

Function of biomarkers Description with Alzheimer’s disease (AD) as the example


Diagnostic function Biomarkers may assist in the clinical diagnosis. They may also facilitate the differentiation of
neurodegenerative from functional cognitive disorders (e.g. depression) in the elderly or
enable the differentiation of AD from non-AD dementias. A diagnostic biomarker should have
a sensitivity for detecting AD of at least 85% and its specificity in differentiating AD patients
from age-matched control subjects and from patients with other forms of dementia should
reach a sensitivity detection of at least 75% (The Ronald and Nancy Reagan Research Institute
of the Alzheimer’s Association and National Institute on Aging Working Group, 1998).
Screening function Biomarkers discriminate the ‘healthy’ state from early disease state, preferably in the
asymptomatic phase (pre-dementia stage of AD). They might assist early (preclinical)
diagnosis of AD and enable early intervention. Targets for the screening approach can be
entire population or subpopulations at risk (e.g. MCI).
Prognostic function Biomarkers can predict the likely course of the disease. They are associated with outcome
regardless of the treatment used on patients.
Predictive (stratification) Biomarkers may be associated with outcome from a particular therapy directed against a
function specific pathogenic mechanism identified by the biomarker. They predict the likely response
to a drug (e.g. an anti-amyloid substance) before starting treatment; thus, they classify
individuals as ‘responders’ as compared to likely ‘non-responders’.
Monitoring function Biomarkers can be used to monitor efficacy of a drug treatment once the responder status is
established. Within the setting of a clinical drug trial, a monitoring marker serves as a
‘surrogate marker’ if it is closely linked to a clinically meaningful treatment outcome (e.g.
cognition, functional abilities, quality of life); thus, it can be used as an outcome parameter
under a particular treatment.
Abbreviation: MCI, mild cognitive impairment.

specificity. Only very few AD biomarker candidates have high throughput-omics (i.e. proteomic/molecular) methods
passed this step. In the next step, the new marker has to be that try to identify biomarkers that can differentiate affected
studied in a more representative population-based sample, cases from healthy controls and other comparisons, such as
providing an assessment of its true diagnostic properties and various brain disorders, regardless of whether the biomarkers
thus demonstrating its clinical usefulness within the clini- may be directly known as linked to molecular or pathogenetic
cal setting and purpose for which the biomarker is intended. processes. Exploratory proteome-based methods include two-
Large-scale controlled multicentre biomarker trials are cur- dimensional gel electrophoresis (2-DE), protein chips, or liquid
rently being conducted in American, Asian, Australian and chromatography combined with liquid chromatography–mass
European Alzheimer networks in an attempt to systemati- spectrometry (LC-MS) (Brinkmalm et al., 2015). Recent
cally develop and validate core feasible candidate biomarkers approaches also involve the use of tandem mass tags for quan-
in research areas such as, neurobiochemistry and structural, titative proteomics and peptidomics, which allow for the detec-
functional and metabolic neuroimaging (Blennow et al., tion and quantification of hundreds of endogenous and tryptic
2014; Hampel et al., 2012, 2014; Lista et al., 2014). peptides in much larger sample numbers than what has been
Two different approaches exist for the identification of possible before (Brinkmalm et al., 2015). Till date, the -omics
cerebrospinal fluid (CSF) biomarkers: the hypothesis-driven (i.e. proteomics) approach has not provided any validated and
candidate biomarker approach and the hypothesis-indepen- standardized AD biomarkers.
dent exploratory -omics approach. The candidate biomarker
approach tries to identify a protein or molecule reflecting
known or hypothesized neurochemical changes underlying
mechanistic molecular and pathogenic processes, i.e. leading to 51.3 DIAGNOSTIC BIOMARKERS
neurodegeneration. In AD biomarker research, the candidate FOR AD
approach should reflect neuronal and synaptic degeneration,
‘mis-metabolism’ of the amyloid precursor protein (APP) and/ 51.3.1 ESTABLISHED CSF BIOMARKERS
or aggregation of Aβ with subsequent deposition in plaques FOR AD
and/or hyperphosphorylation of tau with subsequent formation
of paired helical filaments and tangles. The -omics based (i.e. A large number of studies have shown that three core fea-
proteome) exploratory approach uses unbiased hypothesis-free sible CSF biomarkers reflecting neuropathological changes
530 Dementia

associated with AD, namely the 42-amino acid form of biotech companies to develop upgraded and validated ver-
amyloid β (Aβ1-42), which correlates inversely with brain Aβ sions of assays and to set up CSF biomarker assays on fully
plaque pathology and total and phosphorylated forms of tau automated laboratory analyzers. These developments will
(T-tau and P-tau, respectively), which correlate with neu- pave the way for uniform cut-off levels for the CSF biomark-
rodegeneration and tau pathology in AD, distinguish AD ers and a more general use of these diagnostic tools.
patients from control individuals, as well as from patients
with other neurodegenerative diseases, such as Parkinson’s 51.3.2 PLASMA BIOMARKERS
disease and frontotemporal dementia (Blennow et al., 2010).
Further, these three biomarkers identify patients with mild Several efforts to discover and develop diagnostic biomark-
cognitive impairment (MCI) due to AD from patients with ers for AD in peripheral blood have been done, as summa-
MCI due to non-AD causes with positive and negative pre- rized above (Lista et al., 2013). Plasma or serum biomarkers
dictive values above 90% (Hansson et al., 2006; Shaw et al., are still experimental at Phases 1 and 2 of diagnostic vali-
2009; Visser et al., 2009) and are more than often dynami- dation; there are few candidates in pilot- and single-centre
cally altered already during preclinical disease stages (Skoog studies, first of their kind that seem to perform close to
et al., 2003; Fagan et al., 2014). The CSF Aβ and tau mark- the diagnostic accuracy range achieved by established core
ers were developed using a targeted approach; the molecu- feasible CSF biomarkers (see below). The best studied can-
lar composition of the plaque and tangle changes originally didate biomarker in plasma so far is Aβ, but the findings
described by Alois Alzheimer in 1906 was revealed during are contradictory. In plasma, current assays allow the reli-
the 1980s and quantitative enzyme-linked immunosorbent able measurement of Aβ1-40 and Aβ1-42 (although, additional
assays (ELISA) for the proteins were developed and refined species most likely exist) and several studies have exam-
during the 1990s (Blennow et al., 1995). ined their association with dementia, AD and/or cerebral
The clear diagnostic utility of CSF Aβ1-42, T-tau and P-tau β-amyloidosis. However, the results are less clear than those
have made it important to standardize the biomarker mea- derived from measuring CSF Aβ1-42 or from positron emis-
surements into clinical routine tests, where different meth- sion tomography (PET) studies and the significant associa-
ods can be calibrated to each other to give similar results tions, if any, are weak and go in either direction (Zetterberg
(low bias) with high analytical precision (low random and Blennow, 2006; Ritchie et al., 2014). As no correlation
variation). Standardization efforts are ongoing within the exists between CSF and plasma Aβ levels (Mehta et al., 2001;
Global Biomarkers Standardization Consortium (GBSC), Hansson et al., 2010), it is possible that most of the Aβ pep-
Materials and Methods Working Groups (available at http:// tides measured in plasma are derived from extra-cerebral
www.alz.org/research/funding/global_biomarker_CSF_­ sources such as platelets in which APP expression and pro-
materials.asp) within the Alzheimer’s Association (AA) cessing are high in the α granules (Van Nostrand et al.,
and the International Federation of Clinical Chemistry 1990). In the Alzheimer’s Disease Neuroimaging Initiative
and Laboratory Medicine Working Group for CSF Proteins (ADNI), plasma Aβ levels fail to differentiate AD patients
(IFCC-WG-CSF) (available at http://www.ifcc.org/ifcc from control individuals and amyloid-positive from amy-
-scientific-division/sd-­working-groups/csf-proteins-wg loid-negative individuals, although a weak positive rela-
-csf/), with the aim to standardize both pre-analytical and tionship between plasma Aβ1-40:Aβ1-42 ratio and Aβ ligand
laboratory procedures and to harmonize levels between assay retention on PET was observed in APOE ε4-negative subjects
formats (Carrillo et al., 2013). Protocols for standardization only (Swaminathan et al., 2014). Data from the Australian
of procedures for CSF collection and sample handling as well Imaging, Biomarker and Lifestyle Flagship Study of Ageing
as shipment have been proposed, together with laboratory (AIBL) have highlighted that plasma Aβ levels are influ-
procedures for run acceptance and batch bridging to main- enced by inflammatory and renal function covariates and
tain long-term stability of ELISA-based CSF biomarkers that absolute levels of either Aβ1-40 or Aβ1-42 do not associate
(Palmqvist et al., 2014). Within the IFCC-WG-CSF, selected with AD or neocortical Aβ burden (Rembach et al., 2014).
reaction monitoring (SRM) mass spectrometry-based refer- However, the Aβ1-42:Aβ1-40 ratio (note inverse ratio as com-
ence measurement procedures (RMP), i.e. ‘gold standard’ pared to the ADNI results above) was slightly reduced in
methods for CSF Aβ1-42, have been published (Leinenbach et patients with AD and correlated inversely with neocortical
al., 2014; Korecka et al., 2014) and projects to develop RMPs amyloid burden as determined by amyloid PET. In essence,
for CSF tau are ongoing. These methods for absolute quan- the ADNI and AIBL results corroborate each other and sug-
tification of CSF Aβ1-42 will be used to set the exact levels gest that plasma Aβ levels per se are not diagnostically use-
on a certified reference material (CRM), i.e. aliquoted CSF ful, but that the Aβ1-42:Aβ1-40 ratio may reflect mechanisms
pools with clinically relevant levels (high, medium and low) loosely associated with cerebral β-amyloidosis.
of Aβ1-42, that will be used to normalize levels between assay Interestingly, recent ‘spectacular’ data based on a
formats (Mattsson et al., 2012; Carrillo et al., 2013). Between- highly sensitive immunoprecipitation mass spectrometry
laboratory and longitudinal (batch-related) variations are approach have revealed a novel plasma Aβ fragment start-
monitored in the Alzheimer’s Association Quality Control ing at 3 amino acids N-terminally of Asp1 in the Aβ domain
(QC) Programme for CSF biomarkers (Mattsson et al., and ending at Val40 (Kaneko et al., 2014). The ratio of this
2011a). These standardization efforts have also stimulated fragment to Aβ1-42 discriminated 40 amyloid-positive (as
Blood and cerebrospinal fluid biomarkers for Alzheimer’s disease 531

determined by amyloid PET) from 22 amyloid-negative more successful. Results from earlier studies may have been
individuals with sensitivity and specificity of 92.5% and clouded by the presence of non-symptomatic AD patients
95.5%, respectively (Kaneko et al., 2014). These promising in the control groups. A recent study on the effect size of
pilot data await independent replication. the major susceptibility gene for AD, APOE ε4, supports
With regard to other blood biomarker candidates, this reasoning; when disregarding clinical information on
researchers from AIBL, ADNI and AddNeuroMed have patients with cognitive impairment and simply grouping
identified multi-protein plasma biomarker panels that them on the basis of CSF tau and Aβ markers, the associa-
together seem to identify individuals with pathological tion of APOE ε4 with AD was twice as strong as compared to
brain Aβ accumulation with reasonable accuracy (Kiddle et classifying patients according to clinical status (Andreasson
al., 2012; Burnham et al., 2014). As a consequence, a con- et al., 2014).
siderable recent attention has been devoted towards multi- The potential functions of AD biomarkers are many.
marker blood-based biomarker panels that have utility in Besides diagnostics, CSF biomarkers may be utilized for
detecting AD with recent work also examining the capacity prognosis, assessing disease progression, developing treat-
of the multi-marker approach in distinguishing AD from ments, monitoring treatment effects and studying disease
non-AD dementias (O’Bryant et al., 2010; Doecke et al., mechanisms. Several studies have investigated the AD bio-
2012; Hu et al., 2012; Burnham et al., 2014; Sattlecker et al., marker potential of various analytes related to Aβ.
2014).
In a seminal study, Ray et al. (2007) identified a panel of 18 51.3.4 BACE1
markers that were highly accurate in detecting AD as well as
predicting conversion from MCI to AD. Unfortunately, this For Aβ to be produced, the APP is cut by two different
work was not replicated on an independent assay platform enzymes, β-secretase and γ-secretase. The major β-secretase
(Soares et al., 2009). On the other hand, recent analyses have in the brain is called β-site APP cleaving enzyme-1 (BACE1).
been validated across cohorts (O’Bryant et al., 2011; Doecke Several studies have investigated the levels of CSF BACE1
et al., 2012; Hu et al., 2012), assay platforms (Kiddle et al., activity in patients with MCI and AD but the results are not
2014; O’Bryant et al., 2014a) and even species (O’Bryant et univocal. Three small studies found that patients with AD
al., 2014b). Recent investigation also demonstrates the pos- had increased BACE1 activity compared with non-demented
sibility that the blood-based biomarker profile approach can controls (Holsinger et al., 2004; Verheijen et al., 2006) or
be accurate in discriminating AD from Parkinson’s disease patients with other dementia disorders (Holsinger et al.,
(O’Bryant et al., 2014b). Altogether, these data suggest that 2006). One study found higher activity in patients with MCI
blood-based biomarker profiles are expected to be of great and dementia due to AD compared with controls (Zetterberg
utility in detecting AD and need to be validated in larger et al., 2008). Another study found significantly elevated
population-based and/or primary care clinical settings. BACE1 activity in subjects with MCI but not in patients
However, it should also be taken into account that the with AD when compared with controls (Zhong et al., 2007).
different biomarker panels are non-overlapping, some pro- One study failed to show any differences in BACE1 activ-
tein identities are surprising, with no clear link to known ity between MCI and AD patients compared with controls
disease mechanisms and therefore, additional validation (Mulder et al., 2010). However, when patients with a patho-
work is needed. Especially, multi-marker blood-based logic profile of the core AD biomarkers were compared with
biomarker studies are based on analysing a large set of controls with a normal biomarker pattern, a significant eleva-
unselected proteins followed by generation of a multivari- tion of BACE1 activity was found in the patient group. The
ate models showing diagnostic separation, while individ- MCI subjects contributed the most to this elevation. Finally, a
ual proteins show huge overlaps. Such statistical models large study conducted did not find any differences in activity
introduce a large risk of statistical overfitting of data (with between AD patients and controls (Rosén et al., 2012). When
many more proteins than patients and controls introduced the AD patients were stratified into mild and moderate-to-
into the model). Thus, models based on specific cohort severe AD, an increased BACE1 activity could be seen in
often fail to replicate in independent clinical cohorts. One the group with mild AD compared to the more affected AD
can conclude that additional efforts are required in order patients and controls. These studies indicate that the activity
to determine the potential of these approaches in discrim- of BACE1 may be mildly elevated in the early stages of AD
inating AD from non-AD dementia diseases. but the diagnostic usability of the biomarker seems limited.
BACE1 may, however, prove to be valuable for target engage-
51.3.3 NOVEL CANDIDATE BIOMARKERS ment or outcome in clinical trials of BACE1 inhibitors.

The search for new AD biomarker candidates may be 51.3.5 SAPPα/SAPPβ


facilitated by the enrichment of core CSF AD biomarkers.
By including patients with characteristic biological signs The levels of sAPPα and sAPPβ correlate very well in
of an AD pathological process and ensuring that control AD patients as well as controls (Zetterberg et al., 2008).
subjects lack this profile, future biomarker studies may be Several studies have failed to show any differences in the
532 Dementia

levels of these biomarkers between AD patients and con- little characterization of what the various assays are even
trols (Olsson et al., 2003; Zetterberg et al., 2008; Hertze measuring. Two recent studies have employed the ultra-
et al., 2010; Johansson et al., 2011; Rosén et al., 2012). sensitive Erenna® platform (Singulex, Alameda, CA) to
One study found higher levels of sAPPβ in MCI subjects measure CSF Aβ oligomers using oligomer-selective anti-
compared with controls (Olsson et al., 2003) and another bodies, one of which reported elevated (Savage et al., 2014)
showed MCI subjects that upon follow-up developed AD and the other unaltered (Yang et al., 2015) CSF levels of Aβ
and had higher levels of sAPPβ than patients who did oligomers.
not (Perneczky et al., 2011). However, Hertze et al. (2010)
found no differences in sAPP levels in MCI subjects with 51.3.7 OTHER CANDIDATE MARKERS
incipient AD compared to stable MCI or patients that
developed other dementias. Studies that compared sub- In addition to Αβ, tau and related markers, many other
jects with MCI or dementia that had a pathologic core biomarkers have been investigated to detect pathophysi-
CSF AD biomarker profile with controls with a normal ological processes that may be involved in or reflect AD
profile found that the former group had increased lev- pathogenesis, e.g. microglial activation and synaptic
els of sAPPα and sAPPβ, but there were large overlaps degeneration, or may aggravate the effects of AD neuro-
between these groups (Gabelle et al., 2010; Lewczuk et al., pathology more or less independently of the primary dis-
2010, 2012;). The diagnostic value of sAPPα and sAPPβ ease process, e.g. cerebrovascular disease or concomitant
thus appears limited. Yet, the proteins may be used for Lewy body disease (LBD) or TAR-DNA-binding protein 43
studying effects on the APP metabolism in clinical trials (TDP-43) pathology.
(Blennow et al., 2010). The best established CSF biomarkers for microglial
activation so far are chitotriosidase, YKL-40 and che-
51.3.6 Aβ OLIGOMERS mokine CC motif ligand 2 (CCL2) (Craig-Schapiro et al.,
2010; Mattsson et al., 2011b). These markers are elevated
Soluble oligomers of Aβ may be more toxic than fibrillar in CSF AD, but till date we do not know if they also reflect
Aβ aggregates (Sakono and Zako, 2010). They can inhibit disease intensity and clinical disease progression. To
hippocampal long-term potentiation in vivo (Walsh et al., that end, longitudinal studies are needed. With regard to
2002) and cause abnormal tau phosphorylation and neu- synaptic degeneration, several research groups have suc-
ritic dystrophy (Jin et al., 2011). A relatively large number cessfully detected synaptic proteins in CSF (Davidsson
of assay formats to measure Aβ oligomers in CSF have been and Blennow, 1998; Davidsson et al., 1999; Thompson
published. These papers differ in size and molecular weight et al., 2003; Zougman et al., 2008; Thorsell et al., 2010).
of investigated oligomers and have shown that the CSF level However, these studies have been performed on relatively
of Aβ oligomers is generally very low, probably, less than 1% large quantities of pooled CSF. Moreover, it has also been
of total Aβ levels, and thereby very difficult to quantify in a necessary to selectively purify and concentrate the tar-
reliable manner. get proteins in several steps and the quantitative aspects
In AD patients, elevated Aβ oligomer levels have been of the techniques have been suboptimal. Recent break-
found in brains (Shankar et al., 2008; Bruggink et al., 2013) throughs in ultra-sensitive measurement techniques may
and CSF (Pitschke et al., 1998; Gao et al., 2010; Fukumoto help resolving these issues (Rissin et al., 2010). There are
et al., 2010; Hölttä et al., 2013). However, not all studies a number of other tau-independent markers of neurode-
have found altered levels in AD patients versus controls generation also, e.g. heart-type fatty acid-binding protein
(Santos et al., 2012; Bruggink et al., 2013), but the latter of and visinin-like protein 1 (VILIP-1) (Steinacker et al.,
these found a negative correlation between oligomer levels 2004; Lee et al., 2008), which may be useful in clinical
and cognitive status, indicating a potential use for assess- trials to detect effects of tau-targeting therapies on neu-
ing disease stage even as late as the AD dementia stage rodegeneration in which the tau markers per se may be
(not correlation for the core CSF markers). This suggests a affected by the treatment.
potential value for Abeta oligomers as surrogate outcomes. Cerebrovascular disease is at present best diagnosed
One study found elevated levels of oligomers in human and using neuroimaging, but there are CSF biomarker pat-
mouse brains but not in CSF (Yang et al., 2013). Handoko terns that may indicate changes typical of cerebral small
et al. (2013) found that CSF oligomer levels increased vessel disease, including elevated levels of neurofilament
with age and correlated with levels of T-tau in cognitively light and matrix metalloproteinase proteins (Bjerke et al.,
normal older adults. Also, increased levels were found in 2011). Alpha-synuclein (α-syn) is a potential biomarker
cognitively normal subjects with a biomarker profile indi- for LBD, but this association is less well established
cating impending AD (Handoko et al., 2013). Except for than the relationship between CSF Aβ1-42 and cerebral
technical difficulties to measure minute amounts of Aβ β-amyloidosis (Zetterberg et al., 2014). Assays of CSF
oligomers in CSF samples, it is also possible that different TDP-43 have been published (Geser et al., 2011), but the
assays measure different variants of Aβ oligomers, which association of this marker with TDP-43 pathology is so
might explain the divergent results. The studies provide far unknown.
Blood and cerebrospinal fluid biomarkers for Alzheimer’s disease 533

of AD patients be identified and targeted for precision-


51.4 THE ROLE OF BLOOD AND based medicine approaches rather than the ‘one-size-fits-
CSF BIOMARKERS IN DRUG all’ approach? Proof of concept data was recently presented
DEVELOPMENT demonstrating that a blood-based profile of baseline
inflammatory status was highly accurate in identifying
51.4.1 BIOMARKERS AS END POINTS IN treatment responders as well as adverse responders in a
previously conducted nonsteroidal anti-inflammatory
AD CLINICAL DRUG TRIALS
drug (NSAID) trial in AD (O’Bryant et al., 2014a). It seems
51.4.1.1 Biomarkers for inclusion possible that CSF and blood-based biomarkers have poten-
tial for advancing a precision-medicine approach to treat-
The diagnostic accuracy of clinical MCI criteria for pre- ing and preventing AD.
dementia or prodromal AD is low to moderate (Visser et
al., 2005). Their use may lead to inclusion of many patients 51.4.1.2 Pharmacodynamic biomarkers
who do not have pre-dementia AD or exclusion of many
who suffer from it. This might partly explain the failure of Depending on the mechanism of action of the drug, dif-
many of the previous drug trials in MCI target populations ferent biomarkers may be needed to detect target engage-
(Jelic et al., 2006). In future trials, CSF-derived biomarkers ment. Most disease-modifying drug candidates so far are
(e.g. different species of Aβ and tau protein) can serve as directed against Aβ pathology and different CSF Aβ mark-
early diagnostic indicators in order to enrich the number ers (reviewed above) or amyloid PET may be useful to
of high-risk ‘true’ cases among subjects selected primar- detect pharmacodynamic effects of such drug candidates.
ily on clinical criteria (Hampel and Broich, 2009; Hampel A ­disease-modifying drug against AD is not expected to
et al., 2015). There are currently two biomarkers for cere- enhance cognition but should stop or at least slow down the
bral β-amyloidosis: (1) CSF levels of Aβ1-42 and (2) amyloid neurodegenerative process, which eventually should trans-
PET. Both have been validated against neuropathology and late into reduced cognitive decline over time (Citron, 2010).
they show high diagnostic performance to identify AD as In order to help detecting positive drug effects on neu-
well as are highly concordant with approximately 90% of rodegeneration, a number of CSF biomarkers are available.
cases being either positive or negative for both biomarkers These include biomarkers for (1) neuroaxonal degeneration,
(Blennow et al., 2015b). CSF Aβ1-42 and amyloid PET have a (2) tangle pathology and (3) synaptic loss.
central position in the newly revised criteria for prodromal CSF biomarkers for neuroaxonal degeneration include
AD outlined by the International Working Group (IWG) for T-tau (measured using assays that detect all tau isoforms
the diagnosis of AD (Dubois et al., 2014) and in the National irrespective of phosphorylation state), neurofilament light
Institute on Aging and Alzheimer’s Association (NIA-AA) and VILIP-1. Although tau is highly expressed in thin
criteria for AD dementia (McKhann et al., 2011) and MCI unmyelinated axons of the cerebral cortex (Trojanowski et
due to AD (Albert et al., 2011). Although, these two Aβ bio- al., 1989), neurofilament light is predominantly a marker
marker modalities provide independent information about of large calibre myelinated axons that extend subcortically
some aspects of the disease process related to AD (Mattsson connecting brain regions (Blennow et al., 2010). VILIP-1 is
et al., 2014), the high concordance between CSF and PET highly expressed in neurones (Laterza et al., 2006) and CSF
Aβ for classification of AD suggests that they may be used levels of VILIP-1 correlate tightly with T-tau levels (Lee et
interchangeably for diagnosis (Blennow et al., 2015b). This al., 2008). Both CSF T-tau and VILIP-1 levels are robustly
approach could considerably increase the statistical power elevated in AD but not specific to the disease process as
and, in turn, decrease sample size and trial costs. Core such (Lee et al., 2008; Hampel et al., 2010a). CSF neurofila-
feasible CSF biomarker candidates, in addition to APOE ment light levels are elevated in frontotemporal dementia,
genotyping are currently recommended to be introduced amyotrophic lateral sclerosis and other disorders with pre-
for meaningful enrichment in MCI populations to decrease dominant long white matter fibre tract involvement such
heterogeneity of study in target population, data variance as vascular dementia, but is also significantly increased
and enhance rate of cognitive decline and progression as in AD, especially, in late onset of the disease (Sjögren et
well as time taken to conversion to AD, particularly, in al., 2000; Zetterberg et al., 2007; Skillbäck et al., 2014a).
proof of concept studies (Siemers et al., 2007, 2009; Hampel All three markers correlate positively with measures of
and Broich, 2009). As an example, the Phase 3 solanezumab disease intensity and predict both clinical disease progres-
and gantenerumab trials employ CSF Aβ1-42 or amyloid PET sion and survival, not only in AD but also in other dis-
(depending on availability) of enrichment of prodromal AD eases (Wallin et al., 2010; Fagan et al., 2014; Skillbäck et
cases in the screening of MCI cases. al., 2014a, 2014b).
It has also been proposed that the cardiovascular dis- Elevated CSF P-tau levels have been regarded as a rather
ease approach of subgrouping patients based on molecular AD-characteristic phenomenon, which indicates tau phos-
biomarkers has potential for improving therapeutic inter- phorylation in a manner that may be downstream of Aβ
ventions in AD. That is to say, should specific subgroups pathology (Hampel et al., 2010a; Buchhave et al., 2012). CSF
534 Dementia

P-tau levels correlate with presence of cortical tangle pathol- subcortical and cortical biomarkers in vascular demen-
ogy (Buerger et al., 2006; Seppälä et al., 2012) and elevated tia and Alzheimer’s disease. The Journal of Alzheimer’s
CSF P-tau levels support a diagnosis of AD (Hampel et al., Disease, 27: 665–676.
2004). There are, in fact, differences in the classification Blennow, K., Dubois, B., Fagan, A.M. et al. (2015a).
performance of different P-tau epitopes developed as ELISA Clinical utility of cerebrospinal fluid biomarkers in the
assays, favouring P-tau phosphorylated at threonine 231 diagnosis of early Alzheimer’s disease. Alzheimer’s &
as an earlier marker and significantly better differentiator Dementia: The Journal of the Alzheimer’s Association,
between AD and frontotemporal dementia (FTD) (Hampel 11: 58–69.
et al., 2004). Research into developing and comparing dif- Blennow, K., Hampel, H., Weiner, M. et al. (2010).
ferent assays looking at a variety of tau epitopes, which may Cerebrospinal fluid and plasma biomarkers in
reflect different molecular signatures of distinct neurode- Alzheimer’s disease. Nature Reviews Neurology, 6:
generative diseases is promising and part of currently ongo- 131–144.
ing studies. Blennow, K., Hampel, H. and Zetterberg, H. (2014).
Finally, synaptic loss and degeneration in AD and most Biomarkers in amyloid-β immunotherapy trials in
likely in other neurodegenerative diseases also may be Alzheimer’s disease. Neuropsychopharmacology, 39:
detected and monitored using CSF levels of the dendritic 189–201.
protein neurogranin (Thorsell et al., 2010; Kvartsberg et al., Blennow, K., Mattsson, N., Schöll, M. et al. (2015b).
2015) and the presynaptic SNARE complex component syn- Amyloid biomarkers in Alzheimer’s disease. Trends in
aptosomal-associated protein 25 (SNAP-25) (Brinkmalm Pharmacological Science, 36: 297–309.
et al., 2014). Blennow, K., Wallin, A., Ågren, H., Spenger, C. et al.
These three CSF biomarker categories all reflect inter- (1995). Tau protein in cerebrospinal fluid: A bio-
related aspects of molecular mechanisms and subsequent chemical diagnostic marker for axonal degenera-
neuropathology, such as neurodegeneration in AD. Drug tion in Alzheimer’s disease? Molecular and Chemical
candidates with a disease-modifying effect should nor- Neuropathology, 26: 231–245.
malize or lower CSF levels of these proteins over a 6- to Brinkmalm, A., Brinkmalm, G., Honer, W.G. et al.
12-month period of treatment. This time window is at pres- (2014). SNAP-25 is a promising novel cere-
ent hypothetical and extrapolated from studies on acute brospinal fluid biomarker for synapse degen-
conditions such as stroke and traumatic brain injury (Hesse eration in Alzheimer’s disease. Molecular
et al., 2001; Neselius et al., 2014) and the time window may Neurodegeneration, 9: 53.
also depend on the mode of action of the drug and how Brinkmalm, A., Portelius, E., Öhrfelt, A. et al. (2015).
strong the disease-modifying effect is. Absence of such Explorative and targeted neuroproteomics in
changes speaks against the drug, irrespective of its precise Alzheimer’s disease. Biochimica et Biophysica Acta,
mode of action, being effective against the generic patho- 1854: 769–778.
genic process in AD. Bruggink, K.A., Jongbloed, W., Biemans, E.A. et al. (2013).
Amyloid-β oligomer detection by ELISA in cerebrospi-
nal fluid and brain tissue. Analytical Biochemistry, 433:
REFERENCES 112–120.
Buchhave, P., Minthon, L., Zetterberg, H. et al. (2012).
Albert, M.S., DeKosky, S.T., Dickson, D. et al. (2011). Cerebrospinal fluid levels of β-amyloid 1–42, but not
The diagnosis of mild cognitive impairment due to of tau, are fully changed already 5 to 10 years before
Alzheimer’s disease: Recommendations from the the onset of Alzheimer dementia. Archives of General
National Institute on Aging-Alzheimer’s Association Psychiatry, 69 (1): 98–106.
workgroups on diagnostic guidelines for Alzheimer’s Buerger, K., Ewers, M., Pirttilä, T. et al. (2006). CSF phos-
disease. Alzheimer’s & Dementia: The Journal of the phorylated tau protein correlates with neocortical
Alzheimer’s Association, 7: 270–279. neurofibrillary pathology in Alzheimer’s disease. Brain,
Andreasson, U., Lautner, R., Schott, J.M. et al. (2014). 129: 3035–3041.
CSF biomarkers for Alzheimer’s pathology and the Buerger, K. and Hampel, H. (2009). Chapter 23:
effect size of APOE ɛ4. Molecular Psychiatry, 19: Biomarkers for the dementias. In Weiner, M.F.
148–149. and Lipton, A.M. (eds.), The American Psychiatric
Biomarkers Definitions Working Group (2001). Biomarkers Publishing Textbook of Alzheimer Disease and Other
and surrogate endpoints: Preferred definitions and Dementias. Arlington, VA: American Psychiatric
conceptual framework. Clinical Pharmacology and Publishing, pp. 407–421.
Therapeutics, 69: 89–95. Burnham, S.C., Faux, N.G., Wilson, W. et al. (2014).
Bjerke, M., Zetterberg, H., Edman, Å. et al. (2011). A blood-based predictor for neocortical Aβ burden
Cerebrospinal fluid matrix metalloproteinases and tis- in Alzheimer’s disease: Results from the AIBL study.
sue inhibitor of metalloproteinases in combination with Molecular Psychiatry, 19: 519–526.
Blood and cerebrospinal fluid biomarkers for Alzheimer’s disease 535

Carrillo, M.C., Blennow, K., Soares, H. et al. (2013). Hampel, H., Buerger, K., Zinkowski, R. et al. (2004).
Global standardization measurement of cerebral Measurement of phosphorylated tau epitopes in the
spinal fluid for Alzheimer’s disease: An update from differential diagnosis of Alzheimer disease: A com-
the Alzheimer’s Association Global Biomarkers parative cerebrospinal fluid study. Archives of General
Consortium. Alzheimer’s & Dementia: The Journal of Psychiatry, 61: 95–102.
the Alzheimer’s Association, 9: 137–140. Hampel, H., Frank, R., Broich, K. et al. (2010b). Biomarkers
Citron, M. (2010). Alzheimer’s disease: Strategies for for Alzheimer’s disease: Academic, industry and regu-
disease modification. Nature Reviews Drug Discovery, latory perspectives. Nature Reviews Drug Discovery, 9:
9: 387–398. 560–574.
Craig-Schapiro, R., Perrin, R.J., Roe, C.M. et al. (2010). Hampel, H. and Lista, S. (2013). Use of biomarkers and
YKL-40: A novel prognostic fluid biomarker for pre- imaging to assess pathophysiology, mechanisms
clinical Alzheimer’s disease. Biological Psychiatry, 68: of action and target engagement. The Journal of
903–912. Nutrition, Health and Aging, 17: 54–63.
Davidsson, P. and Blennow, K. (1998). Neurochemical dis- Hampel, H., Lista, S. and Khachaturian, Z.S. (2012).
section of synaptic pathology in Alzheimer’s disease. Development of biomarkers to chart all Alzheimer’s
International Psychogeriatrics, 10: 11–23. disease stages: The royal road to cutting the therapeu-
Davidsson, P., Puchades, M. and Blennow, K. (1999). tic Gordian Knot. Alzheimer’s & Dementia: The Journal
Identification of synaptic vesicle, pre- and postsyn- of the Alzheimer’s Association, 8: 312–336.
aptic proteins in human cerebrospinal fluid using Hampel, H., Lista, S., Teipel, S.J. et al. (2014). Perspective
liquid-phase isoelectric focusing. Electrophoresis, 20: on future role of biological markers in clinical therapy
431–437. trials of Alzheimer’s disease: A long-range point of
Doecke, J., Laws, S.M., Faux, N.G. et al. (2012). Blood- view beyond 2020. Biochemical Pharmacology, 88:
based protein biomarkers for the diagnosis of 426–449.
Alzheimer’s disease. Archives of Neurology, 69: Hampel, H., Schneider, L.S., Giacobini, E. et al. (2015).
1318–1325. Advances in the therapy of Alzheimer’s disease:
Dubois, B., Feldman, H.H., Jacova, C. et al. (2014). Targeting amyloid beta and tau and perspectives for the
Advancing research diagnostic criteria for Alzheimer’s future. Expert Review of Neurotherapeutics, 15: 83–105.
disease: The IWG-2 criteria. The Lancet Neurology, 13: Handoko, M., Grant, M., Kuskowski, M. et al. (2013).
614–629. Correlation of specific amyloid-β oligomers with tau
Fagan, A.M., Xiong, C., Jasielec, M.S. et al. (2014). in cerebrospinal fluid from cognitively normal older
Longitudinal change in CSF biomarkers in autosomal- adults. JAMA Neurology, 70 (5): 594–599.
dominant Alzheimer’s disease. Science Translational Hansson, O., Zetterberg, H., Buchhave, P. et al. (2006).
Medicine, 6 (226): 226ra30. Association between CSF biomarkers and incipient
Fukumoto, H., Tokuda, T., Kasai, T. et al. (2010). High- Alzheimer’s disease in patients with mild cognitive
molecular-weight beta-amyloid oligomers are elevated impairment: A follow-up study. The Lancet Neurology,
in cerebrospinal fluid of Alzheimer patients. The FASEB 5: 228–234.
Journal, 24 (8): 2716–2726. Hansson, O., Zetterberg, H., Vanmechelen, E. et al.
Gabelle, A., Roche, S., Gény, C. et al. (2010). Correlations (2010). Evaluation of plasma Abeta(40) and Abeta(42)
between soluble α/β forms of amyloid precursor as predictors of conversion to Alzheimer’s disease in
protein and Aβ38, 40, and 42 in human cerebrospinal patients with mild cognitive impairment. Neurobiology
fluid. Brain Research, 1357: 175–183. of Aging, 31: 357–367.
Gao, C.M., Yam, A.Y., Wang, X. et al. (2010). Aβ40 oligo- Hertze, J., Minthon, L., Zetterberg, H. et al. (2010).
mers identified as a potential biomarker for the diag- Evaluation of CSF biomarkers as predictors of
nosis of Alzheimer’s disease. PLoS ONE, 5: e15725. Alzheimer’s disease: A clinical follow-up study of
Geser, F., Prvulovic, D., O’Dwyer, L. et al. (2011). On the 4.7 years. Journal of Alzheimer’s Disease, 21 (4):
development of markers for pathological TDP-43 in 1119–1128.
amyotrophic lateral sclerosis with and without demen- Hesse, C., Rosengren, L., Andreasen, N. et al. (2001).
tia. Progress in Neurobiology, 95: 649–662. Transient increase in total tau but not phospho-
Hampel, H., Blennow, K., Shaw, L.M. et al. (2010a). Total tau in human cerebrospinal fluid after acute stroke.
and phosphorylated tau protein as biological markers Neuroscience Letters, 297: 187–190.
of Alzheimer’s disease. Experimental Gerontology, 45: Holsinger, R.M., Lee, J.S., Boyd, A. et al. (2006). CSF
30–40. BACE1 activity is increased in CJD and Alzheimer dis-
Hampel, H. and Broich, K. (2009). Enrichment of MCI ease versus other dementias. Neurology, 67: 710–712.
and early Alzheimer’s disease treatment trials using Holsinger, R.M., McLean, C.A., Collins, S.J. et al. (2004).
neurochemical and imaging candidate biomark- Increased beta-secretase activity in cerebrospinal fluid
ers. The Journal of Nutrition, Health and Aging, 13: of Alzheimer’s disease subjects. Annals of Neurology,
373–375. 55: 898–899.
536 Dementia

Hölttä, M., Hansson, O., Andreasson, U. et al. (2013). Lewczuk, P., Kamrowski-Kruck, H., Peters, O. et al. (2010).
Evaluating amyloid-β oligomers in cerebrospinal fluid Soluble amyloid precursor proteins in the cerebrospi-
as a biomarker for Alzheimer’s disease. PLoS ONE, 8: nal fluid as novel potential biomarkers of Alzheimer’s
e66381. disease: A multicenter study. Molecular Psychiatry, 15:
Hu, W.T., Holtzman, D.M., Fagan, A.M. et al. (2012). 138–145.
Plasma multianalyte profiling in mild cognitive Lewczuk, P., Popp, J., Lelental, N. et al. (2012).
impairment and Alzheimer’s disease. Neurology, 79: Cerebrospinal fluid soluble amyloid-β protein precur-
897–905. sor as a potential novel biomarkers of Alzheimer’s
Jelic, V., Kivipelto, M. and Winblad, B. (2006). Clinical trials disease. Journal of Alzheimer’s Disease, 28: 119–125.
in mild cognitive impairment: Lessons for the future. Lista, S., Faltraco, F., Prvulovic, D. et al. (2013). Blood
Journal of Neurology, Neurosurgery, and Psychiatry, and plasma-based proteomic biomarker research in
77: 429–438. Alzheimer’s disease. Progress in Neurobiology, 101: 1–17.
Jin, M., Shepardson, N., Yang, T., et al. (2011). Soluble Lista, S., Garaci, F.G., Ewers, M. et al. (2014). CSF Aβ1-42
amyloid beta-protein dimers isolated from Alzheimer combined with neuroimaging biomarkers in the early
cortex directly induce Tau hyperphosphorylation and detection, diagnosis and prediction of Alzheimer’s
neuritic degeneration. Proceedings of the National disease. Alzheimer’s & Dementia: The Journal of the
Academy of Science USA, 108: 5819–5824. Alzheimer’s Association, 10: 381–392.
Johansson, P., Mattsson, N., Hansson, O. et al. (2011). Mattsson, N., Andreasson, U., Persson, S. et al. (2011a).
Cerebrospinal fluid biomarkers for Alzheimer’s disease: The Alzheimer’s Association external quality
Diagnostic performance in a homogeneous mono- control program for cerebrospinal fluid biomark-
center population. Journal of Alzheimer’s Disease, 24: ers. Alzheimer’s & Dementia: The Journal of the
537–546. Alzheimer’s Association, 7: 386–395.
Kaneko, N., Nakamura, A., Washimi, Y. et al. (2014). Novel Mattsson, N., Tabatabaei, S., Johansson, P. et al. (2011b).
plasma biomarker surrogating cerebral amyloid depo- Cerebrospinal fluid microglial markers in Alzheimer’s
sition. Proceedings of the Japan Academy Series B, disease: Elevated chitotriosidase activity but lack
Physical and Biological Sciences, 90: 353–364. of diagnostic utility. Neuromolecular Medicine, 13:
Kiddle, S.J., Sattlecker, M., Proitsi, P. et al. (2014). 151–159.
Candidate blood proteome markers of Alzheimer’s Mattsson, N., Tosun, D., Insel, P.S. et al. (2014).
disease onset and progression: A systematic review Association of brain amyloid-β with cerebral perfusion
and replication study. Journal of Alzheimer’s Disease, and structure in Alzheimer’s disease and mild cogni-
38: 515–531. tive impairment. Brain, 137: 1550–1561.
Kiddle, S.J., Thambisetty, M., Simmons, A. et al. (2012). Mattsson, N., Zegers, I., Andreasson, U. et al. (2012).
Plasma based markers of [11C] PiB-PET brain amyloid Reference measurement procedures for Alzheimer’s
burden. PLoS ONE, 7: e44260. disease cerebrospinal fluid biomarkers: Definitions and
Korecka, M., Waligorska, T., Figurski, M. et al. (2014). approaches with focus on amyloid β42. Biomarkers in
Qualification of a surrogate matrix-based absolute Medicine, 6: 409–417.
quantification method for amyloid-β42 in human McKhann, G.M., Knopman, D.S., Chertkow, H. et al. (2011).
cerebrospinal fluid using 2D UPLC-tandem mass The diagnosis of dementia due to Alzheimer’s disease:
spectrometry. Journal of Alzheimer’s Disease, 41: Recommendations from the National Institute on
441–451. Aging-Alzheimer’s Association workgroups on diag-
Kvartsberg, H., Duits, F.H., Ingelsson, M. et al. (2015). nostic guidelines for Alzheimer’s disease. Alzheimer’s
Cerebrospinal fluid levels of the synaptic protein & Dementia: The Journal of the Alzheimer’s
neurogranin correlates with cognitive decline in pro- Association, 7 (3): 263–269.
dromal Alzheimer’s disease. Alzheimer’s & Dementia: Mehta, P.D., Pirttila, T., Patrick, B.A. et al. (2001). Amyloid
The Journal of the Alzheimer’s Association. 11 (10): beta protein 1–40 and 1–42 levels in matched cerebro-
1180–1190. spinal fluid and plasma from patients with Alzheimer
Laterza, O.F., Modur, V.R., Crimmins, D.L. et al. (2006). disease. Neuroscience Letters, 304: 102–106.
Identification of novel brain biomarkers. Clinical Mulder, S.D., van der Flier, W.M., Verheijen, J.H. et al.
Chemistry, 52: 1713–1721. (2010). BACE1 activity in cerebrospinal fluid and
Lee, J.M., Blennow, K., Andreasen, N. et al. (2008). The its relation to markers of AD pathology. Journal of
brain injury biomarker VLP-1 is increased in the cere- Alzheimer’s Disease, 20: 253–260.
brospinal fluid of Alzheimer disease patients. Clinical Neselius, S., Brisby, H., Granholm, F. et al. (2014).
Chemistry, 54: 1617–1623. Monitoring concussion in a knocked-out boxer by CSF
Leinenbach, A, Pannee, J., Dülffer, T. et al. (2014). Mass biomarker analysis. Knee Surgery Sports Traumatology
spectrometry-based candidate reference measure- Arthroscopy, 23 (9): 2536–2539.
ment procedure for quantification of amyloid-β in O’Bryant, S.E. (2012). Using blood markers for Alzheimer
cerebrospinal fluid. Clinical Chemistry, 60: 987–994. disease in clinical practice? Neurology, 79: 846–847.
Blood and cerebrospinal fluid biomarkers for Alzheimer’s disease 537

O’Bryant, S.E., Rissman, R.A. and Lyketsos, C. (2014a). Working Group on: “Molecular and biochemical mark-
A proinflammatory endophentoype predicts treat- ers of Alzheimer’s disease”. Neurobiology of Aging, 19
ment response in a multicenter trial of NSAIDs in (2): 109–116.
AD. Alzheimer’s & Dementia: The Journal of the Rosén, C., Andreasson, U., Mattsson, N. et al. (2012).
Alzheimer’s Association, 10: 273–274. Cerebrospinal fluid profiles of amyloid β-related
O’Bryant, S.E., Xiao, G., Barber, R. et al. (2011). A blood- biomarkers in Alzheimer’s disease. Neuromolecular
based screening tool for Alzheimer’s disease that Medicine, 14: 65–73.
spans serum and plasma: Findings from TARC and Sakono, M. and Zako, T. (2010). Amyloid oligomers:
ADNI. PLoS ONE, 6: e28092. formation and toxicity of Abeta oligomers. The FEBS
O’Bryant, S.E., Xiao, G., Barber, R. et al. (2010). A Journal, 277: 1348–1358.
serum protein-based algorithm for the detection Santos, A.N., Ewers, M., Minthon, L. et al. (2012).
of Alzheimer disease. Archives of Neurology, 67: Amyloid-β oligomers in cerebrospinal fluid are
1077–1081. associated with cognitive decline in patients with
O’Bryant, S.E., Xiao, G., Zhang, F. et al. (2014b). Validation Alzheimer’s disease. Journal of Alzheimer’s Disease,
of a serum screen for Alzheimer’s disease across assay 29: 171–176.
platforms, species, and tissues. Journal of Alzheimer’s Sattlecker, M., Kiddle, S.J., Newhouse, S. et al.
Disease, 42: 1325–1335. (2014). Alzheimer’s disease biomarker discovery
Olsson, A., Höglund, K., Sjögren, M. et al. (2003). using SOMAscan multiplexed protein technol-
Measurement of alpha- and beta-secretase cleaved ogy. Alzheimer’s & Dementia: The Journal of the
amyloid precursor protein in cerebrospinal fluid from Alzheimer’s Association, 10: 724–734.
Alzheimer patients. Experimental Neurology, 183: Savage, M.J., Kalinina, J., Wolfe, A. et al. (2014). A sensi-
74–80. tive aβ oligomer assay discriminates Alzheimer’s
Palmqvist, S., Zetterberg, H., Blennow, K. et al. (2014). and aged control cerebrospinal fluid. Journal of
Accuracy of brain amyloid detection in clinical practice Neuroscience, 34: 2884–2897.
using cerebrospinal fluid β-amyloid 42: A cross-valida- Seppälä, T.T., Nerg, O., Koivisto, A.M. et al. (2012).
tion study against amyloid positron emission tomogra- CSF biomarkers for Alzheimer disease correlate
phy. JAMA Neurology, 71: 1282–1289. with cortical brain biopsy findings. Neurology, 78:
Perneczky, R., Tsolakidou, A., Arnold, A. et al. (2011). 1568–1575.
CSF soluble amyloid precursor proteins in the diag- Shankar, G.M., Li, S., Mehta, T.H. et al. (2008). Amyloid-
nosis of incipient Alzheimer disease. Neurology, 77: beta protein dimers isolated directly from Alzheimer’s
35–38. brains impair synaptic plasticity and memory. Nature
Pitschke, M., Prior, R., Haupt, M. et al. (1998). Detection Medicine, 14: 837–842.
of single amyloid beta-protein aggregates in the Shaw, L.M., Vanderstichele, H., Knapik-Czajka, M. et al.
cerebrospinal fluid of Alzheimer’s patients by fluores- (2009). Cerebrospinal fluid biomarker signature in
cence correlation spectroscopy. Nature Medicine, 4: Alzheimer’s disease neuroimaging initiative subjects.
832–834. Annals of Neurology, 65: 403–413.
Ray, S., Britschgi, M., Herbert, C. et al. (2007). Siemers, E. (2009). Biochemical markers as endpoint in
Classification and prediction of clinical Alzheimer’s clinical trials: Applications in phase 1, 2 and 3 stud-
diagnosis based on plasma signaling proteins. Nature ies. Alzheimer’s & Dementia: The Journal of the
Medicine, 13: 1359–1362. Alzheimer’s Association, 5 (Suppl. 1): P95.
Rembach, A., Faux, N.G., Watt, A.D. et al. (2014). Changes Siemers, E.R., Dean, R.A., Friedrich, S. et al. (2007).
in plasma amyloid beta in a longitudinal study of aging Safety, tolerability, and effects on plasma and
and Alzheimer’s disease. Alzheimer’s & Dementia: The cerebrospinal fluid amyloid-beta after inhibition of
Journal of the Alzheimer’s Association, 10: 53–61. gamma-secretase. Clinical Neuropharmacology, 30:
Rissin, D.M., Kan, C.W., Campbell, T.G. et al. (2010). 317–325.
Single-molecule enzyme-linked immunosorbent assay Sjögren, M., Rosengren, L., Minthon, L. et al. (2000).
detects serum proteins at subfemtomolar concentra- Cytoskeleton proteins in CSF distinguish frontotempo-
tions. Nature Biotechnology, 28: 595–599. ral dementia from AD. Neurology, 54: 1960–1964.
Ritchie, C., Smailagic, N., Noel-Storr, A.H. et al. (2014). Skillbäck, T., Farahmand, B., Bartlett, J.W. et al. (2014a).
Plasma and cerebrospinal fluid amyloid beta for the CSF neurofilament light differs in neurodegenerative
diagnosis of Alzheimer’s disease dementia and other diseases and predicts severity and survival. Neurology,
dementias in people with mild cognitive impairment 83: 1945–1953.
(MCI). Cochrane Database of Systematic Review, (6): Skillbäck, T., Rosén, C., Asztely, F. et al. (2014b).
CD008782. Diagnostic performance of cerebrospinal fluid total tau
The Ronald and Nancy Reagan Research Institute of the and phosphorylated tau in Creutzfeldt-Jakob disease:
Alzheimer’s Association and National Institute on Results from the Swedish Mortality Registry. JAMA
Aging Working Group (1998). Consensus report of the Neurology, 71: 476–483.
538 Dementia

Skoog, I., Davidsson, P., Aevarsson, O. et al. (2003). Visser, P.J., Verhey, F., Knol, D.L. et al. (2009). Prevalence
Cerebrospinal fluid beta-amyloid 42 is reduced and prognostic value of cerebrospinal fluid markers of
before the onset of sporadic dementia: A population- Alzheimer pathology in subjects with subjective cogni-
based study in 85-year-olds. Dementia and Geriatric tive impairment and mild cognitive impairment. The
Cognitive Disorders, 15: 169–176. DESCRIPA study. The Lancet Neurology, 8: 619–627.
Soares, H.D., Chen, Y., Sabbagh, M. et al. (2009). Wallin, A.K., Blennow, K., Zetterberg, H. et al. (2010).
Identifying early markers of Alzheimer’s disease using CSF biomarkers predict a more malignant outcome in
quantitative multiplex proteomic immunoassay panels. Alzheimer disease. Neurology, 74: 1531–1537.
Annals of the New York Academy of Sciences, 1180: Walsh, D.M., Klyubin, I., Fadeeva, J.V. et al. (2002).
56–67. Naturally secreted oligomers of amyloid beta protein
Steinacker, P., Mollenhauer, B., Bibl, M. et al. (2004). Heart potently inhibit hippocampal long-term potentiation in
fatty acid binding protein as a potential diagnostic vivo. Nature, 416: 535–539.
marker for neurodegenerative diseases. Neuroscience Yang, T., Hong, S., O’Malley, T. et al. (2013). New ELISAs
Letters, 370: 36–39. with high specificity for soluble oligomers of amyloid
Swaminathan, S., Risacher, S.L., Yoder, K.K. et al. (2014). β-protein detect natural Aβ oligomers in human brain
Association of plasma and cortical amyloid beta but not CSF. Alzheimer’s & Dementia: The Journal of
is modulated by APOE ε4 status. Alzheimer’s & the Alzheimer’s Association, 9: 99–112.
Dementia: The Journal of the Alzheimer’s Association, Yang, T, O’Malley, T.T., Kanmert, D. et al. (2015). A highly
10: e9–e18. sensitive novel immunoassay specifically detects low
Thompson, P.M., Kelley, M., Yao, J. et al. (2003). Elevated levels of soluble Aβ oligomers in human cerebrospinal
cerebrospinal fluid SNAP-25 in schizophrenia. fluid. Alzheimer’s Research and Therapy, 7: 14.
Biological Psychiatry, 53: 1132–1137. Zetterberg, H., Andreasson, U., Hansson, O. et al. (2008).
Thorsell, A., Bjerke, M., Gobom, J. et al. (2010). Elevated cerebrospinal fluid BACE1 activity in incipient
Neurogranin in cerebrospinal fluid as a marker of Alzheimer disease. Archives of Neurology, 65: 1102–1107.
synaptic degeneration in Alzheimer’s disease. Brain Zetterberg, H. and Blennow, K. (2006). Plasma Abeta
Research, 1362: 13–22. in Alzheimer’s disease – Up or down? The Lancet
Trojanowski, J.Q., Schuck, T., Schmidt, M.L. et al. (1989). Neurology, 5: 638–639.
Distribution of tau proteins in the normal human Zetterberg, H., Jacobsson, J., Rosengren, L. et al. (2007).
central and peripheral nervous system. Journal of Cerebrospinal fluid neurofilament light levels in amyo-
Histochemistry and Cytochemistry, 37: 209–215. trophic lateral sclerosis: Impact of SOD1 genotype.
Van Nostrand, W.E., Schmaier, A.H., Farrow, J.S. et al. European Journal of Neurology, 14: 1329–1333.
(1990). Protease nexin-II (amyloid beta-protein precur- Zetterberg, H., Petzold, M. and Magdalinou, N. (2014).
sor): A platelet alpha-granule protein. Science, 248: Cerebrospinal fluid α-synuclein levels in Parkinson’s
745–748. disease – Changed or unchanged? European Journal
Verheijen, J.H., Huisman, L.G., van Lent. N. et al. (2006). of Neurology, 21: 365–367.
Detection of a soluble form of BACE-1 in human Zhong, Z., Ewers, M., Teipel, S. et al. (2007). Levels of
cerebrospinal fluid by a sensitive activity assay. Clinical beta-secretase (BACE1) in cerebrospinal fluid as a
Chemistry, 52: 1168–1174. predictor of risk in mild cognitive impairment. Archives
Visser, P.J., Scheltens, P. and Verhey, F.R. (2005). Do of General Psychiatry, 64: 718–726.
MCI criteria in drug trials accurately identify sub- Zougman, A., Pilch, B., Podtelejnikov, A. et al. (2008).
jects with predementia Alzheimer’s disease? Journal Integrated analysis of the cerebrospinal fluid pepti-
of Neurology, Neurosurgery and Psychiatry, 76: dome and proteome. Journal of Proteome Research, 7:
1348–1354. 386–399.
52
Established treatments for Alzheimer’s disease

ALEXANDER KURZ AND NICOLA T. LAUTENSCHLAGER

treatment and patients receiving placebo on two or more rele-


52.1 INTRODUCTION vant domains. The U.S. Food and Drug Administration (FDA)
applies a dual outcome, which includes cognition to document
Approved treatments for Alzheimer’s disease (AD) target the specificity of the effect and a global assessment of overall
biochemical abnormalities, which are a consequence of functioning to ensure that the effect is clinically meaningful
nerve cell loss in forebrain nuclei and in the cerebral cor- (Ad-hoc-FDA-Dementia-Assessment-Task-Force, 1991). The
tex. Their mode of action thus addresses the final stretch of European regulatory authority (The European Medicines
a complex pathological cascade (Hardy, 2009) but does not Agency [EMEA]) favours a triple outcome including global
interfere with the mechanisms that induce neuronal degen- impression, cognition and functional ability (Broich, 2007;
eration. Cholinesterase inhibitors (ChEIs) partially restore Katona et al., 2007). Treatment differences on these domains
the deficit in acetylcholine, which arises from a significant are assessed using standardized and validated psychometric
deficit of neurones in the nucleus basalis of Meynert and instruments. Some frequently used clinical scales are listed in
in the central septal area, both of which project to corti- Table 52.1. Since AD is a progressive disorder, deterioration
cal regions (Bartus et al., 1982). Memantine attenuates the will occur in the placebo group over time. Hence, a difference
toxic effects of glutamate, which is released in excess from between active treatment and placebo does not necessarily
degenerating cortical neurones, but preserves physiological imply improvement from baseline, but often indicates less
glutamate-mediated signalling (Greenamyre et al., 1988). worsening in actively treated patients. Participants in clinical
Whether these therapies confer additional neuroprotective drug trials, particularly, in studies commissioned by manufac-
potential in addition to purely symptomatic effects, has been turers, are usually selected in terms of high treatment expecta-
widely debated, but has not been demonstrated in human tions, low co-morbidity, restricted concomitant medications
studies (Lleó et al., 2006). ChEIs and memantine may only and few neuropsychiatric symptoms. Therefore, these popula-
be used for the treatment of patients with AD diagnosed tions are often not representative of patients seen in primary
with dementia. In subjects with pre-dementia AD, ChEIs or secondary care settings. Industry-sponsored trials typically
did not achieve a clinically meaningful slowing of progres- apply strict inclusion and exclusion criteria, which exclude the
sion in the disease (Raschetti et al., 2007). The potential of majority of outpatients with AD (Gill et al., 2004). Treatment
memantine for the secondary prevention of dementia has duration is usually short, not exceeding 12–24 weeks. Only
not been explored. few studies of anti-dementia drugs provide ­placebo-controlled
data beyond 6 months. For these reasons, clinical trials pur-
vey an incomplete and biased perspective on the efficacy and
tolerability of anti-dementia treatments. Many important
52.2 CLINICAL EFFICACY questions regarding generalizability of effects on less selected
patient populations, optimal duration of treatment, impact
The clinical efficacy of anti-dementia treatments has been on neuropsychiatric symptoms of dementia and relevance to
evaluated in double-blind, randomized controlled clini- patient or caregiver quality of life remain largely unanswered.
cal trials (RCTs). These studies are usually designed to meet This chapter draws primarily on meta-analysis. Individual
the requirements for approval by regulatory authorities. studies are referred to, with regard to patient subgroups who
Current criteria for efficacy require demonstration of statisti- are not covered by these analyses and in consideration of
cally significant differences between patients receiving active less frequently used outcomes, treatment combinations and
539
540 Dementia

Table 52.1 Frequently used assessment instruments in clinical antidementia trials

Domain Instrument Score range Polarity References


Global assessment CIBIC, CIBIC-Plus 1–7 ≤3 better (Schneider, 1997)
4 no change
≥5 worse
GBS 0–162 Lower better (Gottfries et al., 1982)
CDR-SB 0–18 Lower better (Morris, 1993)
Cognitive ability MMSE 0–30 Higher better (Folstein et al., 1975)
ADAS-cog 0–70 Lower better (Rosen et al., 1984)
SIB 0–100 Higher better (Schmitt et al., 1997)
Activities of daily ADCS-ADL 0–110 Higher better (Galasko et al., 1997)
living
ADCS-ADL sev 0–54 Higher better (Galasko et al., 2005)
PDS 0–100 Higher better (DeJong et al., 1989)
DAD 0–40 Higher better (Gelinas, 1999)
Neuropsychiatric NPI 0–14 Lower better (Cummings et al.,
symptoms 1994)
Abbreviations: CIBIC, Clinician’s Interview-Based Impression of Change; CIBIC-Plus, Clinician-Based Impression of Change-Plus Caregiver
Input); GBS, The Gottfries–Brane–Steen Scale; CDR-SB, Clinical Dementia Rating Sum of Boxes; MMSE, The Mini-Mental
State Examination; ADAS-cog, Alzheimer’s Disease Assessment Scale-cognitive subscale; SIB, Severe Impairment Battery;
ADCS-ADL, Alzheimer’s Disease Cooperative Study-Activities of Daily Living scale; ADCS-ADL sev, Alzheimer’s Disease
Cooperative Study-Activities of Daily Living Scale for severe impairment; PDS, Progressive Deterioration Scale; DAD,
The Disability Assessment for Dementia; NPI, The Neuropsychiatric Inventory.

comparisons between drugs prescribed. Data are presented design of which three employed an enrichment strategy
on intent-to-treat ­populations if available. (Qizilbash et al., 1998). Outcome measures varied greatly
across these studies. In these studies, which had durations
52.2.1 TACRINE of 12–30 weeks, significant a­ dvantages were found favour-
ing tacrine on global assessment and cognition, but not on
Tacrine (tetrahydroaminoacridine, THA) is a non-­ activities of daily living (ADL). The difference from placebo
competitive, reversible ChEI with a higher affinity for
­ was 2 points on the Alzheimer’s Disease Assessment Scale-
butyryl cholinesterase (BChE) than acetylcholinesterase cognitive subscale (ADAS-cog) at 12 weeks and 1.4 units on
(AChE). In addition to its effects on cholinesterases, tacrine the Mini-Mental State Examination (MMSE) after 24 weeks
inhibits monoamine oxidase activity, affects noradrena- (Table 52.2). The major side effect of tacrine is liver toxicity,
line, dopamine and serotonin uptake and release, increases which manifests as serum transaminase elevation (Watkins
histamine levels, stimulates insulin secretion and increases et al., 1994). An increase above the upper limit of the norm
glucose metabolism (Wagstaff and McTavish, 1994). Tacrine occurs in 36%–66% of patients, an increase more than three
is metabolized in the liver. The plasma elimination half-life times the upper limit of the norm is observed in 13%–50%
is 3–4 hours with repeated oral doses. Tacrine was the first of treated individuals (Wagstaff and McTavish, 1994). Most
ChEI approved for use in mild-to-moderate AD in the year instances of liver toxicity are asymptomatic and reverse
1993. Dosage is titrated according to tolerability, starting within 4 weeks. Changes in liver function are rare. Tacrine
with 10 mg four times daily to a patient, increasing up to treatment is also associated with dose-dependent cholinergic
40 mg four times daily and a further increase at 4-to 6-week adverse events including nausea, vomiting, diarrhoea, dys-
intervals. Weekly monitoring of serum transaminase levels pepsia and anorexia. Reported incidence rates are 14%–35%
is recommended for the first 18 weeks of therapy and for for nausea and 4%–18% for diarrhoea (Raina et al., 2008).
the 6 weeks following each increase in dosage. Monitoring In non-enriched studies, the likelihood of withdrawal was
should be carried out every 3 months after this course is significantly greater for tacrine as compared with ­placebo.
over. The clinical efficacy of tacrine was evaluated initially The most frequent reason for premature termination was
in crossover studies taking into consideration small patient transaminase elevation.
samples with mixed results. These studies were followed by
larger trials, which shaped the current methodology of anti-
dementia drug evaluation (Wagstaff and McTavish, 1994). 52.2.2 DONEPEZIL
In a meta-analysis, data were summarized from 12 studies
on a total of 1984 patients. Six studies had a crossover design Donepezil is a piperidine-based, non-competitive revers-
of which four involved exposure to tacrine prior to ran- ible ChEI, which shows much greater selectivity for AChE
domization (enrichment), another six had a parallel-group than for BChE. It has a half-life of approximately 70 hours,
Established treatments for Alzheimer’s disease 541

allowing once daily administration. Absorption of donepezil patients rated as improved on the Clinician’s Interview-
is complete and uninfluenced by food or time of adminis- Based Impression of Change Plus Caregiver Input (CIBIC-
tration. The drug is available in 5 and 10 mg preparations, Plus) was 10%–15% larger in actively treated patients than
the lower dose often being prescribed in the initial 4 weeks in patients receiving placebo. On cognitive scales, there
of treatment. Donepezil is metabolized via the liver and were significant treatment differences on the ADAS-cog of
eliminated primarily through the kidneys (Wilkinson, 2.0 and 2.8 points for the 5 and 10 mg doses, respectively,
1999). It received FDA approval for the treatment of mild- on the MMSE of 1.4 points at 24 weeks independently of the
to-­moderate dementia in AD in the year 1996, which was dose and 1.8 points for the high dose at 52 weeks and on the
expanded to the treatment of severe dementia in AD in Severe Impairment Battery (SIB) of 5.5 points for the high
the year 2006. The evidence that accrued from 15 clinical dose at 24 weeks. Donepezil significantly improved ADL
trials including 4406 participants was summarized in a and neuropsychiatric symptoms relative to placebo levels.
meta-analysis (Birks and Harvey, 2006) (Table 52.3). Global Withdrawals due to adverse events and patients suffering at
assessment scales revealed significant advantages favour- least one adverse event were significantly more frequent in
ing donepezil at both the 5 and 10 mg daily doses after 24 the donepezil than in the placebo groups. Adverse events
and 52 weeks of treatment, respectively. The proportion of seen more frequently in the donepezil groups than in the

Table 52.2 Summary of clinical trials with tacrine

Statistical
Domain Instrument Duration (weeks) Treatment difference significance
Global assessment CIBIC 12–30 OR 1.58 0.002
Cognitive ability ADAS-cog 12 2.07 <0.001
MMSE 24 1.41 0.001
Activities of daily PDS 6 0.75 n.s.
living
Tolerability Withdrawals 12–30 OR 3.68 <0.001
Source: Qizilbash, N. et al., JAMA, 281, 2287–2288, 1998.
Abbreviations: OR, odds ratio; n.s., not significant.

Table 52.3 Summary of clinical trials with donepezil

Dosage (mg/ Duration Treatment Statistical


Domain Instrument day) (weeks) difference significance
Global assessment CIBIC-Plus 5 24 15 <0.0001
% improved 10 11 <0.0001
GBS 10 52 6.01 0.05
CDR-SB 5 24 0.51 <0.0001
Cognitive ability ADAS-cog 5 24 2.02 <0.0001
10 2.81 <0.0001
MMSE 5 24 1.44 0.0004
10 52 1.34 <0.0001
10 1.84 0.006
SIB 10 24 5.55 <0.0001
Activities of daily DAD 10 24 8.00 0.0004
living ADCS-ADL 10 52 1.60 0.02
PDS 10 3.80 0.0004
Neuropsychiatric NPI 10 24 2.62 0.02
symptoms
Tolerability Withdrawals due 10 24 5 0.003
to adverse
events %
Patients with at 5 24 7 0.03
least one 10 5 0.008
adverse
event %
Source: Birks, J., Cochrane Database Systematic Reviews, (1), CD005593, 2006.
542 Dementia

placebo groups were cholinergic in nature and included on nursing home admissions and progression of disability,
nausea, vomiting and diarrhoea. No significant group dif- which had been defined as the primary outcomes in this
ference was found in this trial regarding global assessment. study (Courtney et al., 2004). Due to methodological prob-
In patients with moderate-to-severe dementia (MMSE base- lems including lack of statistical power, high dropout rate
line score 5–17) statistically significant advantages of done- and unusual treatment schemes, this trial probably underes-
pezil in relation to placebo were observed at 24 weeks on timated the true drug effects (Black and Szalai, 2004).
global assessment, cognitive ability, ADL and behavioural The symptomatic benefits of ChEI treatment may be
disturbances of similar magnitude as seen in the mild-to- lost with disease progression despite continued treatment.
moderate subgroup (Feldman et al., 2001). In one study con- It was, therefore, hypothesized that higher than standard
ducted on patients with severe dementia (baseline MMSE doses might sustain treatment-related benefits for longer. In
score ≤ 10), a statistically significant improvement of cog- a study on 1371 patients with moderate-to-severe demen-
nitive ability over baseline was observed in conjunction tia in AD who had received donepezil at 10 mg/day for ≥12
with less decline on ADL (Winblad et al., 2006). In a second weeks were randomized to receive 23 mg/day or conven-
independent study on patients with severe dementia (base- tional 10 mg/day for 24 weeks. Small but statistically signifi-
line MMSE score ≤ 12), subjects receiving donepezil showed cant benefits favouring the higher-than-standard dose were
a significant advantage of cognitive ability in relation to observed for cognitive ability but not for global assessment.
placebo-treated patients at 24 weeks, which was mainly The rate of treatment-associated adverse events was higher
due to deterioration in the placebo group. There was also in the higher-dosage group (Farlow et al., 2010).
a significant advantage on global assessment but no treat-
ment difference on ADL in this study (Black et al., 2007).
No effect of donepezil on neuropsychiatric symptoms was
seen in studies involving patients with severe dementia. 52.2.3 RIVASTIGMINE
At the other end of the severity spectrum, in subjects with
amnestic mild cognitive impairment (aMCI), donepezil had Rivastigmine is a slowly reversible, non-competitive
a small effect on cognitive ability but not on global func- inhibitor of AChE and BChE and has a phenylcarbamate
tion after 48 weeks of treatment (Doody et al., 2009). There structure. Since the activity of AChE declines, the activ-
was no significant effect of donepezil on the progression to ity of BChE remains unchanged or even increases as AD
dementia within 3 years in a patient (Petersen et al., 2005). progresses (Perry et al., 1978), the dual-enzyme inhibition
Donepezil is the only anti-dementia medication for holds particular promise for patients with moderate and
which placebo-controlled data are available beyond treat- severe dementia. The metabolism of rivastigmine is largely
ment durations of 6 months. Information on 12 months was independent of the cytochrome P450 (CYP) liver enzyme
obtained from a trial on 286 patients with mild-to-­moderate system and the risk of interactions with other drugs is low.
dementia (MMSE scores 10–26). On a global assessment After binding to its target enzyme, the carbamate por-
scale (Gottfries–Bråne–Steen [GBS] scale) patients receiving tion of rivastigmine is slowly hydrolyzed and eliminated.
donepezil had declined by week 52, as had placebo-treated Rivastigmine has a half-life of 2 hours and requires at least
subjects, but there was a statistically significant advantage twice daily oral dosage, starting with 3 mg/day, increased
for active treatment. Regarding cognitive ability, patients to between 6 and 12 mg/day, according to individual tol-
treated with donepezil showed little deterioration on the erability (Polinsky, 1998). Rivastigmine was approved for
MMSE at study end point relative to baseline, whereas the treatment of mild-to-moderate dementia in AD in the
worsening by 2.2 points had occurred in patients receiv- year 2000. A meta-analysis summarized the results of 13
ing placebo. This indicates that donepezil offset cognitive RCTs, which included 5121 participants with mild-to-
decline in this patient group by almost 1 year. No statisti- severe dementia in AD (baseline MMSE 10–26) who were
cally significant effects on neuropsychiatric symptoms were treated for up to 12 months (Birks et al., 2009) (Table 52.4).
observed (Winblad et al., 2001). Another 12-month study on After 24 to 26 weeks of treatment, rivastigmine at a dose
531 patients with moderate dementia (MMSE score 10–20) of 6–12 mg/day was associated with significant advantages
used predefined clinically evident decline in function as the relative to placebo on global assessment, cognitive abil-
primary outcome. Significantly, more patients in the pla- ity and ADL. The treatment differences were 2 ADAS-cog
cebo group than in the donepezil group reached this end points, 0.8 MMSE points and 4.5 points on the SIB. No sig-
point. Survival analysis revealed that the mean time to clini- nificant effect on neuropsychiatric symptoms was found.
cally evident functional decline was 5 months longer in the The lower dosage of 1–4 mg/day was associated with sig-
actively treated patients (Mohs et al., 2001). The only study nificant treatment differences on global assessment and
which provided randomized and placebo-controlled evi- cognition, but not on ADL. The incidence of withdrawals
dence on the efficacy of donepezil beyond 1 year was carried due to adverse events and the proportion of patients suf-
out in a primary care setting, independent of the manufac- fering at least one adverse event were significantly higher
turer. Small but statistically significant treatment differences in patients treated with 6–12 mg/day rivastigmine than in
on cognitive ability and ADL were maintained for 2 years. patients receiving placebo. The most frequent adverse event
However, no significant effect of donepezil was detected seen in these patients were vomiting, diarrhoea, anorexia,
Established treatments for Alzheimer’s disease 543

Table 52.4 Summary of clinical trials with rivastigmine

Dosage (mg/ Duration Treatment Statistical


Domain Instrument day) (weeks) difference significance
Global assessment CIBIC-Plus 1–4 26 6 0.01
% improved 6–12 7 <0.0001
Cognitive ability ADAS-cog 1–4 26 0.84 0.01
6–12 1.99 <0.0001
ADAS-cog 1–4 26 2 n.s.
% ≥ 4 units 6–12 6 <0.01
MMSE 1–4 26 0.43 0.02
6–12 0.83 <0.0001
SIB 6–12 26 4.53 0.03
Activities of daily PDS 1–4 26 0.4 n.s.
living ADCS-ADL 6–12 2.15 <0.0001
6–12 1.8 0.03
Neuropsychiatric NPI 3–12 24 2.2 n.s.
symptoms
Tolerability Withdrawals due 1–4 26 0 n.s.
to adverse 6–12 15 <0.01
events %
Patients with at 1–4 26 1 n.s.
least one 6–12 12 <0.01
adverse event
%
Source: Birks, J. et al., Cochrane Database Systematic Reviews, (1), CD001191, 2009.

headache, syncope, abdominal pain and dizziness. In one including nausea, vomiting and weight loss was particu-
study on patients with severe dementia in AD (MMSE larly reduced. Mild skin irritation appears to be common
score 5–12), there were statistically significant advantages with 7%–8% of patients experiencing moderate-to-severe
of rivastigmine relative to placebo on global assessment erythema or pruritus (Cummings et al., 2010). As the effi-
and cognitive ability but not on ADL or behavioural dis- cacy of rivastigmine has been shown to be dose dependent
turbance at 26 weeks (López-Pousa et al., 2004). The treat- a 13.3 mg/24 hours patch was developed and approved for
ment difference was 4 points on the SIB. There is only one mild-to-severe dementia in the United States. A study on
small placebo-controlled RCT on rivastigmine over 12 567 patients with mild-to-moderate dementia in AD who
months (Karaman et al., 2005). Patients were included showed a prespecified amount of cognitive and functional
with ‘advanced moderate’ dementia in AD (MMSE score decline following 24 to 48 weeks of open-label treatment
≤ 14). At study end point, statistically significant treat- with 9.5 mg/24 hours rivastigmine were randomized to
ment differences favouring rivastigmine were observed receive either 9.5 or 813.3 mg/24 hours patch. At all time
in 0.4 units on the Alzheimer’s Disease Cooperative Study points, cognitive scores and ratings of ADLs favoured
(ADCS) – Clinical Global Impression of Change (CGIC), the higher dosage formulation. On the other hand, gas-
of 5.3 points on the ADAS-cog and of 4.3 units on the trointestinal side effects were more frequent in the higher
Progressive Deterioration Scale (PDS). Patients treated dosage group (Cummings et al., 2012). Specifically, nau-
with 6–12 mg/day showed greater benefit than patients sea, vomiting, lack of appetite and weight loss were about
treated with a lower dose. In subjects with aMCI, no dif- twice as common with the 13.3 mg/24 hours patch as
ferences between rivastigmine and placebo were observed compared with the 9.5 mg/24 hours patch. However, this
on cognitive ability or on progression to dementia after 48 is still a lower rate than that associated with the capsule
months of treatment (Feldman et al., 2007). formulation.
In 2007, a transdermal formulation of rivastigmine
became available, which delivers 9.5 mg/24 hours. The effi- 52.2.4 GALANTAMINE
cacy of the rivastigmine patch is equivalent to the capsule
at a dose of 12 mg/day, but tolerability is better (Winblad Galantamine is a tertiary alkaloid, first isolated from
et al., 2007). The number of patients experiencing at least the bulbs of snowdrop and narcissus, now produced
one adverse event was 51% in patients using the patch ­synthetically. It is a competitive, reversible inhibitor of
as compared to 63% in patients treated with capsules AChE with very little BChE activity (Harvey, 1995). In
(p = 0.02). The incidence of gastrointestinal side effects addition, galantamine is an allosteric potentiatior of the
544 Dementia

action of AChE on nicotinic receptors (Maelicke, 2000). It efficacy and tolerability to immediate release galantamine
has a half-life of approximately 6 hours. The recommended at comparable doses (Brodaty et al., 2005).
maintenance dose is 16–24 mg/day taken in two daily
doses. Galantamine was approved by the FDA for the 52.2.5 MEMANTINE
treatment of mild-to-moderate dementia in AD in the year
2001. A meta-analysis summarized the data from 10 clini- Memantine is a non-competitive modulator of the N-methyl
cal trials on a total of 6805 patients with mild-to-moder- d-aspartate (NMDA) receptor and works by normalizing
ate dementia (Loy and Schneider, 2004) (Table 52.5). In glutamatergic neurotransmission. It prevents excitatory
6-month studies, galantamine treatment was associated amino acid neurotoxicity, but does not interfere with the
with significant advantages relative to placebo on global physiological function of glutamate for learning and mem-
assessment, cognitive ability, ADL and neuropsychiatric ory (Butterfield and Pocernich, 2003). Memantine reaches
symptoms at the 16 mg/day dose. For the 8 mg/day dose, maximal plasma concentrations 3–8 hours after oral
the only significant treatment difference was observed on administration, is eliminated renally and has no inhibi-
measures of cognition. The 24 mg/day dose was not supe- tory effect on the cytochrome P450 (CYP) system, as well
rior to the 16 mg/day dose. The treatment difference on as does not inhibit ChEIs (Jarvis and Figgitt, 2003). It is
the ADAS-cog was 3 points at 6 months. At a dose of 24 administered twice daily up to a maximum dose of 20 mg/
mg/day, but not at a dose of 16 mg/day, the incidence of day. Memantine was approved by the FDA in 2003 for use in
withdrawals due to adverse events was higher in patients moderate-to-severe dementia in AD. A meta-analysis sum-
treated with galantamine than in patients receiving pla- marized data from three clinical trials on 1291 patients with
cebo. Adverse events that were frequently observed in moderate-to-severe dementia (baseline MMSE 3–14) and
patients on an active treatment with 16 mg/day galan- from three unpublished studies in 997 patients with mild-
tamine included nausea, vomiting and diarrhoea. The to-moderate dementia in AD, all of which were conducted
higher dose galantamine treatment was associated with over 6-month periods (McShane et al., 2009) (Table 52.6).
an increased incidence of nausea, vomiting, dizziness, In the moderate-to-severe groups data pooled indicated a
weight loss, anorexia, tremor and headache. In nursing beneficial effect after 6 months on cognition, activities of
home residents with severe dementia (n = 407), galan- daily living, neuropsychiatric symptoms and global assess-
tamine titrated up to a maximum dose of 24 mg/day over ment. In the mild-to-moderate groups, there was a marginal
6 months was associated with a significant advantage over beneficial on cognition that was barely detectable clinically
placebo on cognitive ability but not on ADL (Burns et al., and was not accompanied by effects on behaviour or every-
2009). Two studies on individuals with aMCI galantamine day functioning. The tolerability of memantine does not
did not show an effect on progression to dementia within differ from placebo (Farlow et al., 2008). There are no con-
24 months (Winblad et al., 2008). Since 2005, galantamine trolled data on the efficacy of memantine beyond 6 months.
has been available as a prolonged release once daily dos- In individuals with age-associated memory impairment,
age formulation (8, 16 and 24 mg), which shows similar memantine improves attention and information processing

Table 52.5 Summary of clinical trials with galantamine

Dosage (mg/ Duration Treatment Statistical


Domain Instrument day) (months) difference significance
Global assessment CIBIC-Plus 8 6 6 n.s.
% no change or 16 17 <0.0001
improved 24 16 <0.0001
Cognitive ability ADAS-cog 8 6 1.30 <0.05
16 3.10 <0.0001
24 3.13 <0.0001
Activities of daily ADCS-ADL 8 6 0.60 n.s.
living 16 3.10 <0.0001
24 2.30 <0.01
DAD 24 6 3.66 <0.01
Neuropsychiatric NPI 8 6 0.30 n.s.
symptoms 16 2.10 0.034
24 1.49 n.s.
Tolerability Withdrawals due 8 6 1 n.s.
to adverse 16 0 n.s.
events % 24 7 <0.001
Source: Loy, C. and Schneider, L., Cochrane Database Systematic Reviews, (1), CD001747, 2004.
Established treatments for Alzheimer’s disease 545

Table 52.6 Summary of clinical trials with memantine

Duration Treatment Statistical


Domain Instrument Severity (months) difference significance
Global assessment CIBIC-Plus Mild to 6 0.13 0.03
moderate 0.28 <0.0001
Moderate to
severe
Cognitive ability ADAS-cog Mild to 6 0.99 0.001
moderate
SIB Moderate to 6 2.97 <0.0001
severe
Activities of daily ADCS-ADL Mild to 6 0.20 n.s.
living moderate
ADCS-ADL sev Moderate to 6 1.27 0.003
severe
Neuropsychiatric NPI Mild to 6 0.25 n.s.
symptoms moderate 2.76 0.004
Moderate to
severe
Tolerability Withdrawals Mild to 6 1.4 n.s.
before end % moderate 7.6a 0.005
Moderate to
severe
Patients with at Mild to 6 3 n.s.
least one moderate 2 n.s.
adverse event Moderate to
% severe
Source: McShane, R. et al., Cochrane Database Systematic Reviews., (1), CD003154, 2009.
a In favour of memantine.

speed (Ferris et al., 2007). No studies have been conducted the donepezil group (89%) than in the rivastigmine group
to determine whether memantine has a potential of delay- (60%), whereas the frequency of adverse events resulting in
ing the progression from aMCI to dementia. In 2010 an premature discontinuation was higher in the rivastigmine
extended-release formulation of memantine was approved group (22%) than in the donepezil group (11%) (Wilkinson
for the treatment of moderate-to-severe dementia in AD in et al., 2002). The 2-year study was sponsored by the manu-
the United States and other countries, but not in Europe. facturer of rivastigmine. It included 998 individuals with
The advantages of this once-daily capsule are more conve- moderate dementia and preserved blinding by employing
nient dose regimen and lower pill burden (Plosker, 2015). a double-dummy design and identical visit schedules in
Efficacy and tolerability of the extended-release formulation both groups. There was an almost identical decline of cog-
was examined in a RCT on an already ongoing ChEI treat- nitive ability from baseline in both groups at end point.
ment (see Section 52.2.7). On ADL, however, patients treated with donepezil showed
significantly greater deterioration than patients receiving
52.2.6 COMPARISON BETWEEN rivastigmine. The treatment difference was 2 units on the
TREATMENTS Alzheimer’s Disease Cooperative Study-Activities of Daily
Living (ADCS-ADL) scale. No statistically significant dif-
Donepezil was directly compared with rivastigmine in a ference was detected between the two groups on neuro-
12-week trial and a 2-year study. The short-term trial was psychiatric symptoms. During the 16-week dose, titration
sponsored by the makers of donepezil and included 111 sub- period of the study, more gastrointestinal adverse events
jects with mild-to-moderate dementia. The study was not and more premature discontinuations were recorded in the
blinded since dosage scheme and number of study visits rivastigmine group than in the donepezil group. During
was different. Mean changes from baseline were similar in the maintenance period, however, the incidence of gastro-
both groups, with treatment differences of 0.16 points on intestinal adverse events and the number of withdrawals
the ADAS-cog and 0.49 points on the MMSE. ADL and before the end of study was similar in both groups (Bullock
behavioural disturbance were not assessed in this study. The et al., 2005). There are also two studies comparing done-
proportion of study completers was significantly greater in pezil with galantamine. A 12-week study sponsored by
546 Dementia

the makers of donepezil which included 120 subjects with in sufficiently large patient samples, whether they are
mild-to-­moderate dementia showed significantly greater clinically meaningful or not is questionable (Molnar et al.,
improvements from baseline on cognitive ability and ADL 2009). In the absence of a general consensus, several criteria
in the donepezil group as compared with the galantamine have been proposed for the evaluation of treatment effects
group. Treatment differences were 2.5 points on the ADAS- in the real-world context. These include improvement on
cog, 1 point on the MMSE and 2.1 units on the Disability global assessment, slowing of cognitive deterioration, delay
Assessment for Dementia (DAD) (Jones et al., 2004). In con- in reaching significant clinical end points, reduction of
trast, a 52-week head-to-head trial including 182 patients behavioural disturbance, attainment of individual patient
with moderate dementia sponsored by the manufacturer and caregiver goals and lowering the incidence of nursing
of galantamine did not demonstrate statistically signifi- home admissions (Chin, 2008; Qaseem et al., 2008; Molnar
cant differences between the two treatments on cognition et al., 2009).
(ADAS-cog, MMSE), Bristol Activities of Daily Living Scale
and neuropsychiatric symptoms (the Neuropsychiatric 52.3.1 IMPROVEMENT ON GLOBAL
Inventory [NPI]). Rates of gastrointestinal adverse events
RATING
were similar in both groups (Wilcock et al., 2003). The only
study that directly compared donepezil with memantine A 1-unit change on a seven-stage global assessment instru-
was conducted in China, where 100 patients with mild- ment such as the CIBIC, would probably qualify as clini-
to-moderate dementia were randomized to receive either cally meaningful. However, the average advantage of
donepezil at 5mg/day or memantine titrated up to 20 mg/ anti-dementia treatments relative to placebo is less than 0.5
day throughout the trial. At the 16-month end point there units. In a pooled analysis of clinical trials with ChEIs, the
were no statistically significant differences between the two proportion of patients on active treatment who were rated as
groups regarding cognitive ability (MMSE) or ADL (Blessed improved on a global scale varied between 18% and 35% as
scale) (Hu et al., 2006). Results are questionable because compared with 11%–28% in patients receiving placebo. This
donepezil was administered at a low dose and the instru- means that less than 10% of patients improve with treatment
ment used for assessing everyday activities probably lacked than without the same (Lanctôt et al., 2003).
sensitivity to change.

52.2.7 TREATMENT COMBINATIONS 52.3.2 SLOWING OF COGNITIVE


DETERIORATION
Pooled data from four trials including 1549 patients with
moderate-to-severe dementia in AD demonstrated small According to the FDA, compensating the natural history
but significant beneficial effects of the combination of ChEI of cognitive decline by at least 6 months is clinically rel-
and memantine over ChEI alone for global assessment, cog- evant. This equates to a 4 points or greater difference on
nitive ability and neuropsychiatric symptoms. There were the ADAS-cog (Kramer-Ginsberg et al., 1988). None of the
no major differences between the combination therapy and approved AD treatments has achieved an average effect of
ChEI monotherapy regarding the rate of adverse events this magnitude. However, it should be noted that in a 1-year
(Schmidt et al., 2015). This systematic review and meta- study on patients with mild-to-moderate dementia in AD,
analysis includes a large RCT on 677 patients, which showed donepezil delayed the decline of cognitive ability by almost
that the extended release formulation of memantine (28 mg/ 1 year (Winblad et al., 2001) and memantine treatment
day) added to ongoing ChEI treatment also provided sig- reduced the proportion of patients with moderate-to-severe
nificant gains on these three outcomes and was generally dementia showing marked clinical worsening (including a
well tolerated (Grossberg et al., 2013). deterioration on the ADAS-cog by ≥4 points) from about
21% to 11% (Wilkinson and Andersen, 2007).

52.3 CLINICAL IMPORTANCE 52.3.3 DELAY IN REACHING SIGNIFICANT


CLINICAL END POINTS
The advantages provided by the active treatments relative to Progression to disability is a robust criterion for the clini-
P0125 placebo are small in comparison to the range of the cal relevance of treatment effects. This outcome was
assessment instruments used for measurement. The average addressed in two double-blind, placebo-controlled studies
treatment differences of ChEIs are 3%–4% on the ADAS- with donepezil. A 1-year study showed that donepezil treat-
cog (range 0–70), 3%–7% on the MMSE (range 0–30), ment extended the time to loss of critical ADL by 5 months
1%–2% on the ADCS-ADL (range 0–110) and 1%–2% on the (Mohs et al., 2001). On the other hand, a long-term study
NPI (range 0–144). For memantine, the average treatment conducted independently of the pharmaceutical industry
differences are less than 2% on the ADAS-cog, less than 1% in a primary care setting failed to demonstrate a delay in
on the ADCS-ADL and 1%–2% on the NPI. Even if treat- the progression of disability and in reaching the threshold
ment gains of this magnitude attain statistical significance of severe cognitive impairment. However, methodological
Established treatments for Alzheimer’s disease 547

problems (idiosyncratic patient selection procedures and factors which are intangible in anti-dementia drugs includ-
criteria, lower than expected recruitment, high attrition ing patient and caregiver physical health and availability
and repeated washout periods) may have affected the results of community resources (Gaugel et al., 2009). Moreover,
detrimentally (Courtney et al., 2004). depending on individual circumstances, nursing home
care may be a better alternative for patients with dementia
than remaining in the community. For ChEIs, uncontrolled
52.3.4 REDUCTION OF studies have demonstrated that higher dosage and longer
NEUROPSYCHIATRIC SYMPTOMS duration of the treatment were associated with reduced
Clinical trials of anti-dementia drugs usually were not incidence (Knopman et al., 1996; Beusterien et al., 2004) or
designed to evaluate their effects on neuropsychiatric delay (Lopez et al., 2002; Geldmacher et al., 2003; Feldman
symptoms associated with dementia, but measures of et al., 2009) of nursing home admissions. The risk of insti-
behavioural problems were often secondary outcomes. Two tutionalization was further reduced by combining ChEI
independent meta-analysis studies evaluating the behav- treatment with memantine (Lopez et al., 2009). These inves-
ioural effects of ChEIs concluded that active treatment was tigations are flawed, however, because patients were not ran-
associated with an advantage relative to placebo of less than domly allocated to dosage and duration of drug exposure
2 units on the NPI. Even smaller effects were seen in patients (Karlawish, 2004; Schneider and Quizilbash, 2004). The
with moderate-to-severe dementia where neuropsychiatric only long-term placebo-controlled RCT which incorpo-
symptoms are particularly prevalent and effective treatment rated nursing home admission as a primary outcome cri-
is most needed (Trinh et al., 2003; Campbell et al., 2008). A terion failed to show an impact of donepezil on the rate of
systematic review concluded that ChEIs have a weak impact institutionalization (Courtney et al., 2004).
on behavioural disturbance of doubtful clinical impor-
tance (Grimmer and Kurz, 2006). The only RCT examining
the effects of a ChEI on acute neuropsychiatric symptoms 52.4 PRACTICAL CONSIDERATIONS
in patients with severe dementia was negative (Holmes et
al., 2007). According to a review of three double-blind,
placebo-controlled trials with memantine in patients with The pharmacological effects of current AD treatments are
moderate-to-severe dementia in AD, two studies found no not large. Among the various outcomes examined, the delay
statistically significant difference between memantine and of cognitive and functional decline can be considered as
placebo. One study detected a significant advantage favour- clinically meaningful, whereas the impact on neuropsychi-
ing memantine of 3.84 units on the NPI which was due to atric symptoms and caregiver burden is of questionable sig-
worsening in the placebo group (Grossberg et al., 2009). nificance. Even modest clinical benefits, however, are highly
desirable in a disease which deprives affected individuals of
their intellectual capacity, undermines personal autonomy
52.3.5 ATTAINMENT OF INDIVIDUAL and heavily burdens family carers (Burns and O’Brien,
PATIENT AND CAREGIVER GOALS 2006). Against this background, opportunities for better
quality of life, more activity and increased participation
In a 24-week study with galantamine, 12 weeks of which must not be missed, while avoiding treatment which lacks
were double-blind and placebo-controlled, patients on practical utility. Achievement of these goals require timely
active treatment showed statistically significant advantages initiation of treatment, careful monitoring of individual
relative to placebo of 2 points on the ADAS-cog and 0.4 units response, attention to safety issues, skilful use of switching
on the CIBIC-Plus. These treatment gains were entirely con- and combination strategies and long-term continuation.
sistent with the results of the meta-analyses reviewed above.
The attainment of individual goals, however, as defined by
patients and caregivers, was not different between the two 52.4.1 TREATMENT INITIATION
groups (Rockwood et al., 2006). Few trials included care- To take full advantage of the symptom-delaying effect of
giver burden or time spent on caregiving as an outcome and current medications, treatment should be initiated early, as
showed small overall effects (Lingler et al., 2005). soon as the clinical diagnosis can be established with confi-
dence. Due to ceiling effects of the most common assessment
52.3.6 LOWERING THE INCIDENCE OF instruments (MMSE, ADAS-cog) this rational strategy col-
NURSING HOME ADMISSIONS lides with treatment guidelines requiring improvement
rather than maintenance of cognition as a criterion of effi-
As a criterion for the clinical relevance of anti-dementia cacy. The role of early treatment is to preserve a maximum
drug effects, the incidence of nursing home admissions is of cognitive ability and functional independence for as long
problematic. Institutionalization is not exclusively deter- as possible. In patients with mild dementia, a ChEI should
mined by factors that are modifiable through medications be used; in patients with moderate dementia either a ChEI
such as cognitive impairment, limitation of everyday activi- or memantine may be prescribed. Treatment expectations
ties or neuropsychiatric symptoms. It is also triggered by of patients and carers should be focused on delaying decline
548 Dementia

and extending patient independence, rather than on overall a second agent should only be performed after full resolu-
and durable improvement of symptoms (Farlow et al., 2008). tion of side effects associated with the initial agent. In the
case of lack of efficacy, switching can be done overnight,
52.4.2 SAFETY ISSUES with a quicker titration scheme thereafter (Massoud et al.,
2011). In patients with moderate-to-severe dementia switch-
The only contraindication to the use of anti-dementia med- ing to memantine is another possibility. Open-label studies
ication is known hypersensitivity to a specific drug or its have demonstrated that many patients experience improve-
derivatives. ChEIs should be used with caution in patients ment or stabilization after previously unsatisfactory treat-
with a previous history of severe liver disease, pre-­existing ment response and that tolerability is good (Waldemar
bradycardia, peptic ulcer disease, current alcoholism, et al., 2008). Conversely, improvement has been reported
asthma or chronic obstructive pulmonary disease (Bonner in patients with moderate-to-severe dementia who were
and Peskind, 2002). Memantine should be used at a lower switched to donepezil and who experienced lack of efficacy
dose than usual in patients with severe renal impairment. from previous treatment with memantine (Sakka et al.,
Possible interactions with drugs which inhibit the CYP 2007).
pathways can be observed in the case of tacrine, donepezil
and galantamine, but are uncommon in the case of rivastig- 52.4.5 TREATMENT COMBINATIONS
mine (Standridge, 2004). No metabolic interactions with
other drugs have been observed with memantine (Farlow Patients with moderate-to-severe AD who have received
et al., 2008). donepezil for several months may derive significant ben-
efits from the addition of memantine in terms of cognition,
52.4.3 MONITORING TREATMENT activities of daily living, global outome and behavioural
RESPONSE symptoms. Combination treatment may also be beneficial
for patients who experience tolerability problems or unusual
Evaluating whether and to what extent an individual deterioration upon treatment with a ChEI (Cummings et al.,
patient has responded to the treatment provided, is a dif- 2015). Another important way to optimize the management
ficult task in a gradually progressive disease such as AD. of dementia is to combine pharmacological treatment with
The rate of clinical worsening varies widely across patients, non-pharmacological interventions, since the effects of the
is unforeseeable and previous deterioration is not help- two strategies are partly synergistic (Rodakowski et al.,
ful for predicting further decline (Swanwick et al., 1998). 2015). An enhancement of clinical efficacy over standard
Response to treatment is also highly variable with only a pharmacological treatment has been particularly demon-
minority of patients showing true drug-related improve- strated for cognitive stimulation (Ballard et al., 2011).
ment. On the other hand, placebo effects are large. For
these reasons, the paradigm of treatment decisions guided 52.4.6 DISCONTINUATION
by the patient’s feeling or doing better is inappropriate with
regard to AD (Swanwick and Lawlor, 1999). In particular, There is no evidence from RCTs on the efficacy of cur-
lack of improvement does not provide an indication for sus- rent treatments beyond 12 months. After this duration of
pending treatment. No change in the patient’s condition is treatment, patients receiving donepezil do significantly
the most likely outcome after 3 to 6 months’ treatment with better in terms of cognitive performance and functional
an anti-dementia agent. Moreover, a true pharmacological ability than patients on placebo. Several open-label exten-
gain may be masked by a dip in the natural progression. sion studies have suggested that treatment gains are main-
Therefore, the only rational grounds for discontinuation tained for significantly longer periods, although, both the
are poor tolerance, low compliance, dramatic decline fol- actively treated group and the placebo group will continue
lowing the initiation of treatment and absence of deterio- to decline. These results must be interpreted with caution
ration after a drug-free period of 6 weeks (Swanwick and because patients remaining in treatment were positively
Lawlor, 1999). selected in terms of treatment response and drug tolerabil-
ity and because placebo data were generated by statistical
52.4.4 SWITCHING DRUGS extrapolation (Rodda and Walker, 2009). It is likely that
the trajectories of treated and placebo groups will meet
The ChEIs share one basic mode of action but differ with but it is not clear when this will occur. In a well-conducted
regard to other pharmacological properties. In case of poor 12-month double-blind drug discontinuation study of
tolerance or rapid deterioration despite treatment, switch- 295 patients with ­moderate-to-severe dementia due to AD,
ing to another drug within the same class is a viable thera- Howard et al. (2012) found that patients who discontinued
peutic option (Gauthier et al., 2003). Several open-label donepezil, scored 1.9 points worse on the MMSE than those
studies have demonstrated that up to 50% of patients who who continued it for 12 months. Discontinued patients also
experienced lack of efficacy or poor tolerability with the did worse on an ADL measure. Donepezil-treated patients
initial drug responded favourably to subsequent treatment who switched to memantine did nearly as well as those who
with another ChEI. In the case of intolerance, switching to stayed on donepezil.
Established treatments for Alzheimer’s disease 549

Gradual deterioration despite treatment is not a criterion for treatment with cholinesterase inhibitors up to 1 year. At
for withdrawing treatment because the natural course of that point, patients on active treatment do better in terms of
symptoms in an individual patient is unknown. A drug-free global assessment, cognitive ability and ADL than patients
interval of several weeks is one way to determine whether receiving placebo. It is likely that treatment differences are
a patient still benefits from treatment. On the other hand, maintained beyond 12 months, although gradual decline
drug withdrawal carries a risk of rapid deterioration, which will invariably occur. How long ChEI treatment can main-
may not be fully compensated by reinitiating treatment. tain patients above placebo levels is unclear. To provide
At present, it appears to be a rational strategy to continue meaningful benefits to patients with AD and their carers,
treatment as long as neither tolerability problems nor rapid the modest effects of current treatments must be applied
worsening occurs and as long as the patient’s condition can within a skilful management strategy. It includes early
be maintained at a stage where the pharmacological prop- treatment initiation, observation of safety issues and moni-
erties of the present treatment still have a chance of being toring of individual treatment response using appropriate
beneficial. criteria. Physicians must be aware of existing alternatives
in case of tolerability problems or marked deterioration
and they should not give up treatment too early. Wherever
possible, pharmacological treatments should be combined
52.5 CONCLUSIONS with non-pharmacological interventions such as cognitive
stimulation, reminiscence therapy, occupational therapy or
We concur with other comparative reviews in concluding physical exercise (Hogan et al., 2008). The current approved
that the clinical effects of the currently approved AD treat- symptomatic AD treatments will remain the standard of
ments are modest (Harry and Zakzanis, 2005; Birks, 2006; care until novel disease-modifying therapies are deter-
Hansen et al., 2007; Shah et al., 2008; Rodda and Walker, mined to be safe and efficacious. They will continue to be
2009). The most salient outcome is a temporary delay of an important component of dementia managment even
decline in cognitive performance and everyday activi- after new pharmacological strategies become available.
ties by up to 12 months. In some patients, improvement
of cognition and functional ability occurs, which is clini-
cally detectable. The effect size is dose related for donepezil REFERENCES
and rivastigmine (Ritchie et al., 2004). Impacts on neu-
ropsychiatric symptoms have been demonstrated but are Ad hoc FDA Dementia Assessment Task Force (1991).
probably not clinically important (Campbell et al., 2008; Antidementia drug assessment symposium.
Cummings et al., 2008). None of the current pharmaco- Neurobiology of Aging, 12 (4): 379–382.
logical treatments for AD has a potential of delaying the Ballard, C., Khan, Z., Clack, H. and Corbett, A. (2011).
progression from the pre-dementia stage to the dementia Nonpharmacological treatment of Alzheimer disease.
stage (Tricco et al., 2013). The ChEIs do not show clinically Canadian Journal of Psychiatry, 56: 589–595.
relevant differences regarding efficacy and are therefore Bartus, R.T., Dean, R.L., Beer, B. and Lippa, A.S. (1982).
interchangeable. Advantages for the dual-enzyme inhibi- The cholinergic hypothesis of geriatric meory dysfunc-
tion of rivastigmine or for the nicotinic receptor enhance- tion. Science, 217: 408–417.
ment of galantamine are not clinically apparent. Most Beusterien, K.M., Thomas, S.K., Gause, D. et al. (2004).
likely, ChEIs achieve similar clinical benefits in patients Impact of rivastigmine use on the risk of nursing
with mild dementia and in individuals with severe demen- home palcement in a US sample. CNS Drugs, 18:
tia as in the mild-to-moderate dementia subgroup (Birks, 1143–1148.
2006). Memantine is probably less efficacious than ChEI Birks, J. (2006). Cholinesterase inhibitors for Alzheimer’s
in patients with mild dementia and has not been approved disease. Cochrane Database Systematic Reviews, (1):
for this population. In patients with moderate-to-severe CD005593.
dementia, data on clinical efficacy are less consistent for Birks, J., Grimley-Evans, J., Lakovidou, V. et al. (2009).
memantine than for ChEIs. Premature withdrawals are Rivastigmine for Alzheimer’s disease. Cochrane
more frequent in patients treated with ChEIs compared Database Systematic Reviews, (1): CD001191.
to patients receiving placebo. The most common adverse Birks, J. and Harvey, R.J. (2006). Donepezil for demen-
events are nausea, vomiting, diarrhoea, dizziness and tia due to Alzheimer’s disease (review). Cochrane
weight loss. Differences between drugs regarding the fre- Database of Systematic Reviews, (1): CD001190.
quency of these adverse events (Lanctôt et al., 2003) have Black, S.E., Doody, R., Li, H. et al. (2007). Donepezil
become negligible with the introduction of a rivastigmine preserves cognition and global function in patients
patch. A high incidence of liver enzyme elevations in con- with severe Alzheimer disease. Neurology, 69 (5):
junction with a four times daily dosage scheme significantly 459–469.
limits the usefulness of tacrine. Memantine has an excellent Black, S.E. and Szalai, J.P. (2004). Are there long-term
tolerability with adverse event rates being no different from benefits of donepezil in Alzheimer’s disease? Canadian
placebo. Reliable placebo-controlled data are only available Medical Association Journal, 171: 1174–1175.
550 Dementia

Bonner, L.T. and Peskind, E.R. (2002). Pharmacologic treat- Cummings, J.L., Mackell, J. and Kaufer, D. (2008).
ments of dementia. Medical Clinics of North America, Behavioral effects of current Alzheimer’s disease treat-
86: 657–674. ments: A descriptive review. Alzheimer’s & Dementia:
Brodaty, H., Corey-Bloom, J., Potocnik, F.C.V. et al. (2005). The Journal of the Alzheimer’s Association, 4 (1):
Galantamine prolonged-release formulation in the 49–60.
treatment of mild to moderate Alzheimer’s disease. Cummings, J.L., Mega, M., Gray, K. et al. (1994). The
Dementia and Geriatric Cognitive Disorders, 20: neuropsychiatric inventory: Comprehensive assess-
120–132. ment of psychopathology in dementia. Neurology, 44:
Broich, K. (2007). Outcome measures in clinical trials on 2308–2314.
medicinal products for the tretment of dementia: DeJong, R., Ostersund, O.W. and Roy, G.W. (1989).
A European regulatory perspective. International Measurement of quality-of-life changes in patients
Psychogeriatrics, 19: 509–524. with Alzheimer’s disease. Clinical Therapeutics, 11:
Bullock, R. et al. (2005). Rivastigmine and donepezil treat- 545–554.
ment in moderate to moderately-severe Alzheimer’s Doody, R.S., Ferris, S.H., Salloway, S. et al. (2009).
disease over a 2-year period. Current Medical Research Donepezil treatment of patients with MCI: A 48-week
and Opinion, 21: 1317–1327. randomized, placebo-controlled trial. Neurology, 72
Burns, A., Bernabei, R., Bullock, R. et al. (2009). Safety and (18): 1555–1561.
efficacy of galantamine (Reminyl) in severe Alzheimer’s Farlow, M.R., Miller, M.L. and Pejovic, V. (2008). Treatment
disease (the SERAD study): A randomised, placebo- options in Alzheimer’s disease: Maximizing benefit,
controlled, double blind trial. The Lancet Neurology, 8: managing expectations. Dementia and Geriatric
39–47. Cognitive Disorders, 25: 408–422.
Burns, A. and O’Brien, J. (2006). Clinical practice with Farlow, M.R., Salloway, S., Tariot, P.N. et al. (2010).
anti-dementia drugs: A consensus statement of the Effectiveness and tolerability of high-does (23 mg/d)
British Association for Psychopharmacology. Journal of versus standard-dose (10 mg/d) donepezil in moderate
Psychopharmacology, 20: 732–755. to severe Alzheimer’s disease: A 24-week, random-
Butterfield, D.A. and Pocernich, C.B. (2003). The gluta- ized, double-blind study. Clinical Therapeutics, 32:
matergic system and Alzheimer’s disease. Therapeutic 1234–1251.
implications. CNS Drugs, 17: 641–652. Feldman, H.H., Ferris, S., Winblad, B. et al. (2007). Effect
Campbell, N., Ayub, A., Boustani, M.A. et al. (2008). of rivastigmine on delay to diagnosis of Alzheimer’s
Impact of cholinesterase inhibitors on behavioral disease from mild cognitive impairment: The InDDEx
and psychological symptoms of Alzheimer’s disease: study. The Lancet Neurology, 6: 501–512.
A meta-analysis. Clinical Interventions in Aging, 3 (4): Feldman, H., Gauthier, S., Hecker, J. (2001). A 24-week,
719–728. randomized, double-blind study of donepezil in mod-
Chin, J.J. (2008). Alzheimer’s disease – Towards more erate to severe Alzheimer’s disease. Neurology, 57:
patient-centred and meaningful clinical outcomes. 613–620.
Annals of the Academy of Medicine Singapore, 37: Feldman, H., Pirttila, T., Dartigues, J.F. et al. (2009).
535–537. Treatment with galantamine and time to nursing home
Courtney, C., Farrell, D., Gray, R. et al. (2004). Long-term placement in Alzheimer’s disease patients with and
donepezil treatment in 656 patients with Alzheimer’s without cerebrovascular disease. International Journal
disease (AD2000): Randomised double-blind trial. The of Geriatric Psychiatry, 24: 479–488.
Lancet, 363: 2105–2115. Ferris, S., Schneider, L., Farmer, M. et al. (2007).
Cummings, J.L., Farlow, M.R., Meng, X. et al. (2010). A ­double-blind, placebo-controlled trial of memantine
Rivastigmine transdermal patch skin tolerability: in age-associated memory impairment (memantine in
Results of a 1-year clinical trial in patient with mild- AAMI). International Journal of Geriatric Psychiatry, 22:
to-moderate Alzheimer’s disease. Clinical Drug 448–455.
Investigation, 30: 41–49. Folstein, M.F., Folstein, S.E. and McHugh, P.R. (1975).
Cummings, J., Froelich, L., Black, S.E. et al. (2012). Mini mental state: A practical method for grading the
Randomized, double-blind, parallel-group, 48-week cognitive state of patients for the clinician. Journal of
study for efficacy and safety of a higher-dose rivastig- Psychiatric Research, 12: 189–198.
mine patch (15 vs. 10 cm2) in Alzheimer’s disease. Galasko, D., Bennett, D., Sano, M. et al. (1997). An inven-
Dementia and Geriatric Cognitive Disorders, 33 (5): tory to assess activities of daily living for clinical trials
341–353. in patients with Alzheimer’s disase. Alzheimer Disease
Cummings, J.L., Isaacson, R.S., Schmitt, F.A. and and Associated Disorders, 11: S33–S39.
Velting, D.M. (2015). As practical algorithm for man- Galasko, D., Schmitt, F., Thomas, R. et al. (2005). Detailed
aging Alzheimer’s disease: What, when, and why? assessment of activities of daily living in moderate to
Annals of Clinical and Translational Neurology, 2: severe Alzheimer’s disease. Journal of the International
307–323. Neuropsychological Society, 11: 446–453.
Established treatments for Alzheimer’s disease 551

Gaugel, J.E., Yu, F., Krichbaum, K. and Wyman, J.F. (2009). Hogan, D.B., Bailey, P., Black, S. et al. (2008). Diagnosis
Predictors of nursing home admission for persons with and treatment of dementia: 5. Nonpharmacologic
dementia. Medical Care, 47: 191–198. and pharmacologic therapy for mild to moderate
Gauthier, S., Emre, M., Farlow, M.R. et al. (2003). Strategies dementai. Canadian Medical Association Journal, 179:
for continued successful treatment of Alzheimer’s 1019–1026.
disease: Switching cholinesterase inhibitors. Current Holmes, C., Wilkinson, D., Dean, C. et al. (2007).
Medical Research and Opinion, 19: 707–714. Risperidone and rivastigmine and agitated behaviour
Geldmacher, D.S., Provenzano, G., McRae, T. et al. in severe Alzheimer’s disease: A randomised double
(2003). Donepezil is aasociated with delayed nursing blind placebo controlled study. International Journal of
home placement in patients with Alzheimer’s dis- Geriatric Psychiatry, 22: 380–381.
ease. Journal of the American Geriatrics Society, 51: Howard, R., McShane, R., Lindesay J. et al. (2012).
937–944. Donepezil and meantine for moderate-to-severe
Gelinas, I. (1999). Development of a functional measure Alzheimer’s disease. New England Journal of
for persons with Alzheimer’s disease: The disabil- Medicine, 366: 893–903.
ity assessment for dementia. American Journal of Hu, H.T., Zhang, Z.X., Yao, J.L. et al. (2006). Clinical effi-
Occupational Therapy, 53: 471–481. cacy and safety of akatinol memantine in treatment
Gill, S.S., Bronskill, S.E., Mamdani, M. et al. (2004). of mild to moderate Alzheimer disease: A donepezil-
Representation of patients with dementia in clini- controlled, randomized trial. Zhonghua Nei Ke Za Zhi,
cal trials of donepezil. Canadian Journal of Clinical 45: 277–280.
Pharmacology, 11: e274–e285. Jarvis, B. and Figgitt, D.P. (2003). Memantine. Drugs and
Gottfries, C.G., Brane, G., Gullberg, B. and Steen, G. Aging, 20: 465–476.
(1982). A new rating scale for dementia syndromes. Jones, R.W., Soininen, H., Hager, K. et al. (2004). A multi-
Archives of Gerontology and Geriatrics, 1: 311–330. national, randomised, 12-week study comparing the
Greenamyre, J.T., Maragos, E.F., Albin, R.L. et al. (1988). effects of donepezil and galantamine in patients with
Glutamate transmission and toicity in Alzheimer’s mild to moderate Alzheimer’s disease. International
disease. Progress in Neuro-Psychopharmacology and Journal of Geriatric Psychiatry, 19: 58–67.
Biological Psychiatry, 12: 421–430. Karaman, V., Edrogan, F., Köseoglu, E. et al. (2005).
Grimmer, T. and Kurz, A. (2006). Effects of cholinesterase A 12-month study of the efficacy of rivastigmine in
inhibitors on behavioural disturbances in Alzheimer’s patients with advanced moderate Alzheimer’s disease.
disease: A systematic review. Drugs and Aging, 23: Dementia and Geriatric Cognitive Disorders, 19: 51–56.
957–967. Karlawish, J.H.T. (2004). Donepezil delay to nursing home
Grossberg, G.T., Manes, F., Allegri, R.F. et al. (2013). placement study is flawed. Journal of the American
The safety, tolerability, and efficacy of once-daily Geriatrics Society, 52: 845.
memantine (28 mg): A multinational, randomized, Katona, C., Livingston, G., Cooper, C. et al. (2007).
double-blind, placebo-controlled trial in patients with International Psychogeriatric Association consensus
moderate-to-severe Alzheimer’s disease taking cholin- statement on defining and measuring treatment
esterase inhibitors. CNS Drugs, 27 (6): 469–478. benefits in dementia. Internatonal Psychogeriatrics, 19:
Grossberg, G.T., Pejovic, V., Miller, M.L. and Graham, S.M. 345–354.
(2009). Mementine therapy of behavioral symptoms in Knopman, D., Schneider, L., Davis, K. et al. (1996). Long-
community-dwelling patients with moderate to severe term tacrine (Cognex) treatment: Effects on nurs-
Alzheimer’s disease. Dementia and Geriatric Cognitive ing home placement and mortality. Neurology, 47:
Disorders, 27: 164–172. 166–177.
Hansen, R.A., Garlehner, G., Lohr, K.N. and Kaufer, D.I. Kramer-Ginsberg, E., Mohs, R.C., Aryan, M. et al.
(2007). Functional outcomes of drug treatment in (1988). Clinical predictors of course for Alzheimer
Alzheimer’s disease: A systematic review and meta- patients in a longitudinal study: A preliminary report.
analysis. Drugs and Aging, 24: 155–167. Psychopharmacology Bulletin, 24: 458–462.
Hardy, J. (2009). The amyloid hypothesis for Lanctôt, K.L., Herrmann, N., Yau, K.K. et al. (2003).
Alzheimer’s disease: A critical reappraisal. Journal of Efficacy and safety of cholinesterase inhibitors in
Neurochemistry, 110: 1129–1134. Alzheimer’s disease: A meta-analysis. Canadian
Harry, R.D.J. and Zakzanis, K.K. (2005). A comparison Medical Association Journal, 169: 557–563.
of donepezil and galantamine in the treatment of Lingler, J.H., Martire, L.M. and Schulz, R. (2005). Caregiver-
cognitive symptoms of Alzheimer’s disease: A meta- specific outcomes in antidementia clinical drug trials:
analysis. Human Psychopharmacology: Clinical and A systematic review and meta-analysis. Journal of the
Experimental, 20: 183–187. American Geriatrics Society, 53: 983–990.
Harvey, A.L. (1995). The pharmacology of galanthamine Lleó, A., Greenberg, S.M. and Growdon, J.H. (2006).
and its analogues. Pharmacology and Therapeutics, 68: Current pharmacotherapy for Alzheimer’s disease.
113–128. Annual Review of Medicine, 57: 513–533.
552 Dementia

Lopez, O.L., Becker, J.T., Wahed, A.S. et al. (2009). Long- Qizilbash, N., Whitehead, A., Higgins, J. et al. (1998).
term effects of the concomitant use of memantine with Cholinesterase inhibition for Alzheimer disease:
cholinesterase inhibition in Alzheimer disease. Journal of A meta-analysis of the tacrine trials. JAMA: The Journal
Neurology, Neurosurgery and Psychiatry, 80: 600–607. of the American Medical Association, 281: 2287–2288.
Lopez, O.L., Becker, J.T., Wisniewski, S. et al. (2002). Raina, P. et al. (2008). Effectiveness of cholinesterase
Cholinesterase inhibitor treatment alters the natural inhibitors and memantine or treating dementia:
history of Alzheimer’s disease. Journal of Neurology, Evidence review for a clinical practice guideline.
Neurosurgery and Psychiatry, 72: 310–314. Annals of Internal Medicine, 148: 379–397.
López-Pousa, S., Vilalta-Franch, J., Hernandez, B. and Raschetti, R., Albanese, E., Vanacore, N. and Maggini,
Rapatz, G. (2004). Efficacy of rivastigmine in patients M. (2007). Cholinesterase inhibitors in mild cognitive
with severe Alzheimer’s disease: A double-blind, ran- impairment: A systgematic review of randomised trials.
domized pilot study. Brain Aging, 4: 26–34. PLoS Medicine, 4: 1818–1828.
Loy, C. and Schneider, L. (2004). Galantamine for Ritchie, C.W., Ames, D., Clayton, T. and Lai, R. (2004).
Alzheimer’s disease. Cochrane Database Systematic Meta-analysis of randomized trials of the efficacy and
Review, (1): CD001747. safety of donepezil, galantamine, and rivastigmine for
Maelicke, A. (2000). Allosteric modulation of nicotinic the treatment of Alzheimer disease. Amerian Journal
receptors as a treatment strategy for Alzheimer’s of Geriatric Psychiatry, 12: 358–369.
disease. Dementia and Geriatric Cognitive Disorders, Rockwood, K., Fay, S., Song, X. et al. (2006). Attainment
11: 11–18. of goals by people with Alzheimer’s disease receiving
Massoud, F., Desmarais, J.E. and Gauthier, S. (2011). galantamine: A randomized controlled trial. Canadian
Switching cholinesterase inhibitors in older adults with Medical Association Journal, 174: 1099–1105.
dementia. International Psychogeriatrics, 23: 372–378. Rodakowski, J., Saghafi, E., Butters, M.A. and Skidmore,
McShane, R., Areosa-Satre, A. and Minakaran, N. (2009). E.R. (2015). Non-pharmacological interventions for
Memantine for dementia. Cochrane Database adults with mild cognitive impairment and early stage
Systematic Reviews, (1): CD003154. dementia: An updated scoping review. Molecular
Mohs, R.C., Doody, R.S., Morris, J.C. et al. (2001). A 1-year, Aspects of Medicine, 43: 38–53.
placebo-controlled preservation of function survival Rodda, J. and Walker, Z. (2009). Ten years of cholines-
study of donepezil in AD patients. Neurology, 57: terase inhibitors. International Journal of Geriatric
481–488. Psychiatry, 24: 437–442.
Molnar, F.J., Man-Son-Hing, M. and Fergusson, D. (2009). Rosen, W.G., Mohs, R. and Davis, K.L. (1984). A new rating
Systematic review of measures of clinical significance scale for Alzheimer’s disease. American Journal of
employed in randomized controlled trials of drugs for Psychiatry, 11: 1356–1364.
dementia. Journal of the American Geriatrics Society, Sakka, P., Tsolaki, M., Hort, J. et al. (2007). Effectiveness
57: 536–546. of open-label donepezil treatment in patients with
Morris, J.C. (1993). The Clinical Dementia Rating (CDR): Alzheimer’s disease discontinuing memantine mono-
Current version and scoring rules. Neurology, 43: therapy. Current Medical Research and Opinion, 23:
2412–2414. 3153–3165.
Perry, E.K., Perry, R.H., Blessed, G. and Tomlinson, B.E. Schmidt, R., Hofer, E., Bouwman, F.H. et al. (2015). EFNS-
(1978). Changes in brain cholinesterases in senile ENS/EAN Guideline on concomitant use of cholines-
dementia of Alzheimer type. Neuropathology and terase inhbitors in moderate to severe Alzheimer’s
Applied Neurobiology, 4: 273–277. disease. European Journal of Neurology, 22: 889–898.
Petersen, R.C., Thomas, R.G., Grundman, M. et al. (2005). Schmitt, F.A., Ashford, W., Ernesto, C. et al. (1997). The
Vitamin E and donepezil for the treatment of mild cog- Severe Impairment Battery: Concurrent validity and
nitive impairment. New England Journal of Medicine, the assessment of longitudinal change in Alzheimer’s
352: 2379–2388. disease. The Alzheimer’s Disease Cooperative Society.
Plosker, G.L. (2015). Memantine extended release (28 mg Alzheimer Disease and Associated Disorders, 11
once daily): A review of its use in Alzheimer’s disease. (Suppl. 2): S51–S56.
Drugs, 75: 887–897. Schneider, L.S. (1997). Validity and reliability of the
Polinsky, R.N. (1998). Clinical pharmacology of rivastig- Alzheimer’s Disease Study – Clinical Global Impression
mine: A new-generation acetylcholinesterase inhibi- of Change (ADCS-CGIC). Alzheimer Disease and
tor for the treatment of Alzheimer’s disease. Clinical Associated Disorders, 11: 13–23.
Therapeutics, 20: 634–647. Schneider, L. and Quizilbash, N. (2004). Delay in nursing
Qaseem, A. et al. (2008). Current pharmacologic treat- home placement with donepezil. Journal of American
ment of dementia: A clinicl practice guideline from Geriatrics Society, 52: 1024–1026.
the American College of Physicians and the American Shah, R.S. (2008). Current approaches in the treat-
Academy of Family Physicians. Annals of Internal ment of Alzheimer’s disease. Biomedicine and
Medicine, 148: 370–378. Pharmacotherapy, 62: 199-207.
Established treatments for Alzheimer’s disease 553

Standridge, J.B. (2004). Pharmacotherapeutic approaches Wilcock, G., Howe, I., Coles, H. et al. (2003). A long-term
to the treatment of Alzheimer’s disease. Clinical comparison of galantamine and donepezil in the treat-
Therapeutics, 26: 615–630. ment of Alzheimer’s disease. Drugs and Aging, 20 (10):
Swanwick, G.R.J., Coen, R.F., Coakley, D. and Lawlor, 777–789.
B.A. (1998). Assessment of progression and progno- Wilkinson, D.G. (1999). The pharmacology of donepe-
sis in ‘possible’ and ‘probable’ Alzheimer’s disease. zil: A new treatment for Alzheimer’s disease. Expert
International Journal of Geriatric Psychiatry, 13: 331–335. Opinion on Pharmacotherapy, 1: 121–135.
Swanwick, G.R.J. and Lawlor, B.A. (1999). Initiating and Wilkinson, D. and Andersen, H.F. (2007). Analysis of the
monitoring cholinesterase inhibitor treatment for effect of memantine in reducing the worsening of
Alzheimer’s disease. International Journal of Geriatric clinical symptoms in patients with moderate to severe
Psychiatry, 14: 244–248. Alzheimer’s disease. Dementia and Geriatric Cognitive
Tricco, A.C., Soobiah, C., Berliner, S. et al. (2013). Efficacy Disorders, 24: 138–145.
and safety of cognitive enhancers for patients with Wilkinson, D.G., Passmore, A.P., Bullock, R. et al.
mild cognitive impairment: A systematic review and (2002). A multinational, randomised, 12-week,
meta-analysis. Canadian Medical Association Journal, comparative study of donepezil and rivastigmine in
185: 1393–1401. patients with mild to moderate Alzheimer’s dis-
Trinh, N.H., Hoblyn, J., Mohanty, S. and Yaffe, K. (2003). ease. International Journal of Clinical Practice, 56:
Efficacy of cholinesterase inhibitors in the treatment of 441–446.
neuropsychiatric symptoms and functional impairment in Winblad, B., Engedal, K., Soininen, H. et al. (2001).
Alzheimer disease: A meta-analysis. JAMA: The Journal A 1-year, randomized, placebo-conrolled study of
of the American Medical Association, 289: 210–216. donepezil in patients with mild to moderate AD.
Wagstaff, A.J. and McTavish, D. (1994). Tacrine: A review Neurology, 57: 489–495.
of its pharmacodynamic and pharmacokinetic proper- Winblad, B., Gauthier, S., Scinto, L. et al. (2008).
ties, and therapeutic efficacy in Alzheimer’s disease. Safety and efficacy of galantamine in subjects
Drugs and Aging, 4: 510–540. with mild cognitive impairment. Neurology, 70:
Waldemar, G., Hyvärinen, M., Krog-Josiassen, M.A.K. 2024–2035.
et al. (2008). Tolerability of switching from donepezil Winblad, B., Grossberg, G., Frölich, L. et al. (2007). IDEAL:
to memantine treatment in patients with moderate to A 6-month, double-blind, placebo-controlled study of
severe Alzheimer’s disease. International Journal of the first skin patch for Alzheimer disease. Neurology,
Geriatric Psychiatry, 23: 979–981. 69 (4 Suppl. 1): S14–S22.
Watkins, P.B., Zimmerman, H.J., Knapp, M.G. et al. Winblad, B., Kilander, L., Eriksson, S. et al. (2006).
(1994). Hepatotoxic effecs of tacrine administration in Donepezil in patients with severe Alzheimer’s disease:
patients with Alzheimer’s disease. JAMA: The Journal Double-blind, parallel-group, placebo-controlled
of the American Medical Association, 271: 992–998. study. The Lancet, 367: 1057–1065.
53
Drug treatments in development for
Alzheimer’s disease

PAUL YATES AND MICHAEL WOODWARD

would need to be very safe, as it will be given to many peo-


53.1 INTRODUCTION ple who would never develop dementia. These therapeutic
agents may also be effective in treating established demen-
Approaching 30 years after initial descriptions of the ther- tia too. Conversely, a drug that fails in dementia therapeu-
apeutic benefits of tacrine (Summers et al., 1986), we still tic trials may be effective as a preventative agent.
only have symptomatic therapies for Alzheimer’s disease The non-steroidal anti-inflammatory drugs (NSAIDs)
(AD) and no therapies for the other dementias. Only four could fall into this class – all therapeutic trials for AD were
drugs are regularly used for AD (donepezil, galantamine, negative but they still may have a preventative role. Indeed,
rivastigmine and memantine) and no new drug has been this hypothesis was to be tested in the ADAPT trial of the
marketed for almost 20 years. Disease-modifying therapies AD Cooperative Studies group (ADAPT Research Group,
for dementia are desperately needed. 2008), but the trial was terminated early due to concerns
In many respects the course of developing therapies for about toxicity of the agent (celecoxib) in a large healthy pop-
AD has resembled that of developing cancer drugs. There ulation. The ASPREE trial of aspirin in primary prevention
were initially only symptomatic therapies, then, as the may yet shed more light on this area (ASPREE Investigator
molecular basis of cancer was better understood, an enthu- Group, 2013).
siasm emerged that a single magic bullet would be devel- There is some recent evidence from several epidemiolog-
oped that would cure and indeed prevent all cancer. It is ical cohort studies suggesting that public health interven-
likely that the future course of AD drug development will tions such as cardiovascular risk modification may already
also track that of cancer drugs, with therapies that have be playing a role in reducing dementia incidence – although
modest effects on the disease process often used sequen- with a growing aged population, the prevalence is still rising
tially or in combination, along with symptomatic therapies, (Qiu et al., 2013). Chapter 55 addresses prevention of AD.
without a sudden major development that cures all. As with Over recent years, observational cohort studies in early-
some cancer therapies, there are likely to be some drugs onset/genetic (Dominantly Inherited Alzheimer Network
that dramatically alter the course of some types of demen- [DIAN]) and late-onset/sporadic AD (e.g. Alzheimer’s
tia (e.g. ­progranulin-mutation-related frontotemporal lobar Disease Neuroimaging Initiative [ADNI] and the Australian
degeneration [FTLD]) rather than being effective for all Imaging, Biomarkers and Lifestyle [AIBL] studies) have led
dementias. Over decades, we may achieve a situation, where to an appreciation of an evolution of preclinical changes in
most people with dementia treated with the new therapies brain, cerebrospinal fluid (CSF) and blood-based biomark-
can look forward to another 15–20 years of productive life, ers occurring more than 20 years prior to the emergence of
rather than the current average of 5–10 years. Unlike cancer, cognitive sequelae and dementia diagnosis (Bateman et al.,
most people with dementia are already of advanced age and 2012; Villemagne et al., 2013). This suggests that there may
even without dementia their life expectancy is limited. be a substantial window period during which therapeutics
The holy grail of dementia therapies is primary could be implemented to slow or even ward off the progres-
­prevention – identifying those at increased risk of develop- sion of dementia due to AD.
ing a form of dementia, then treating them with a therapy The ability to identify and monitor prodromal and
that prevents dementia from developing. Such a therapy preclinical AD using disease-specific biomarkers (e.g.

554
Drug treatments in development for Alzheimer’s disease 555

β-amyloid positron emission tomography [PET] and CSF Aβ is produced by sequential cleavage of the transmem-
β-amyloid and tau assays) has revolutionized clinical trial brane amyloid precursor protein (APP) by enzymes called
design and given fresh hope to research in AD therapeutics secretases. APP processing may either follow a protec-
(Sperling et al., 2011). tive/neutral or an injurious/harmful pathway. In the for-
However, the recent run of disappointing results from mer, initial cleavage of APP by α-secretase promotes the
trials of high-profile agents targeting β-amyloid (e.g. bap- ‘non-amyloidogenic’ pathway, resulting in the harmless
ineuzumab, semagacestat, solanezumab and gantenerumab) fragments sAPP-α and C83. In the latter, APP is cleaved
has led to many questioning the very basis for the ‘amyloid by β-secretase (BACE1) in the extracellular region and
hypothesis’ of AD pathogenesis – although several of these γ-secretase within the transmembrane domain to release
trials had an unacceptably high proportion of ‘biomarker- the soluble Aβ monomer, which may be 38–42 amino
negative’ participants, who undermined their potential to acid residues long. The Aβ42 fragments then aggregate as
demonstrate efficacy (Broadstock et al., 2014). oligomers and insoluble extracellular β-amyloid plaques.
At the time of writing there are close to 200 therapeutic Aβ42, in particular its oligomeric species, is has neuro-
agents that have been tested for AD treatment in humans. toxic properties and is usually considered to be implicated
Only five have been approved for clinical use: the cholines- in neurodegeneration in AD. By contrast, Aβ40 tends to
terase inhibitors (ChEIs), such as donepezil, galantamine, accumulate within cerebral vessel walls as cerebral amy-
rivastigmine and tacrine, and the N-methyl-d-aspartate loid angiopathy (CAA) and is associated with intracere-
(NMDA) antagonist memantine. Of the remainder, 73 agents bral haemorrhage (Chiang and Koo, 2014).
are no longer in active trials, and at least 87 are active in cur- Aβ within the circulation can also be taken up into the
rent trials. Nineteen therapeutics are in Phase III (including brain via the receptor for advanced glycation end products
Phase II/III) trials, 45 are in Phase II trials (including Phase (RAGE), a multiligand cell-surface receptor. When circu-
I/II) and 17 are in Phase I trials. There are also six Phase IV lating lipids and proteins are exposed to sugars (as occurs
trials of agents already approved for use in other conditions in diabetes and with ageing), they become glycated to form
(including prazocin, carvedilol and simvastatin). advanced glycosylation end products (AGEs), which bind
Among the trials in Phase II/III stages are a number cur- to RAGE resulting in oxidative damage and inflamma-
rently in recruitment that involves participants with pre- tion. RAGE, which is upregulated in AD, also transports
clinical or prodromal AD. circulating Aβ across the blood–brain barrier (BBB) into
In addition to approaches directed at β-amyloid produc- the brain and interacts with Aβ to mediate inflammatory
tion and clearance, there are agents targeting alternative responses, oxidative stress and reduced blood flow (Deane,
disease pathways such as tau-related neurodegeneration 2012). Aβ–apolipoprotein J (Aβ–ApoJ) complexes are also
(with trials for both AD and FTLD participants) and novel taken up by the low-density lipoprotein receptor-related
cholinergic and other neurotransmitter targets, with results protein 2 (LRP2/gp330/megalin) (Ghiso et al., 2004).
to become available over coming years. Aβ is cleared from the brain by several mechanisms.
These include enzymatic degradation (e.g. insulin-degrad-
ing enzyme [IDE] or neprilysin) or transport from brain
interstitial space to the circulation via low-density lipopro-
53.2 TARGETS OF DEMENTIA tein receptor-related protein (LRP, also influenced by apo-
THERAPIES lipoprotein E [ApoE]). In addition, some solutes and waste
products, including Aβ, can exit the brain interstitium via
An increase in understanding of the genetic, cellular and convective bulk flow through aquaporin-4 water channels
molecular basis of dementia has led to a wide range of on astrocyte end-feet (termed the ‘glymphatic’ system) (Iliff
potential therapeutic targets. Only those targets with cur- and Nedergaard, 2013).
rent therapeutic trials are described in this chapter, but it Once in the peripheral circulation, the majority of Aβ is
is highly likely that many more molecules and disease pro- taken up and cleared by the liver, with a smaller proportion
cesses will be targeted in future. excreted by the kidneys or taken up into circulating mono-
The majority of dementia therapeutics currently in clini- cytes and erythrocytes (Liu et al. 2014).
cal trials target AD; however, a growing number of thera- Thus, potential anti-amyloid approaches include those
pies in other dementias are also in development. that reduce production (including inhibitors of γ- and
β-secretase and enhancers of α-secretase), that increase
53.2.1 AMYLOID clearance from the brain or from the peripheral circulation
and that mitigate its deleterious effects.
The ‘amyloid cascade’ hypothesis of AD proposes that
either overproduction or reduced clearance of the protein 53.2.2 TAU
amyloid-β (Aβ) from the brain (or both) lead to both direct
neuronal toxicity and downstream damage via hyperphos- Tau is a protein that stabilizes microtubules, allowing
phorylation of intracellular tau (see Section 53.2.2) (Hardy transport of vesicles and other products of neuronal cell
and Selkoe, 2002; Chiang and Koo, 2014). bodies down the axon to the synapse. In AD, tau is noted
556 Dementia

to become hyperphosphorylated, with associated disrup- 53.2.8 INSULIN-SENSITIZING


tion to the microtubules and eventual aggregation into TREATMENTS AND PEROXISOME
neurofibrillary tangles. Several mechanisms can be tar- PROLIFERATOR–ACTIVATED
geted therapeutically, including hyperphosphorylation RECEPTOR-γ (PPAR-γ)AGONISTS
and tau aggregation. Such approaches may also work in
some of the non-AD dementias, including types of FTLD Diabetes increases risk for AD, and impaired glucose toler-
(Warren et al., 2013). ance and insulin resistance have been associated with hippo-
campal volume loss and cognitive decline. Patients with AD
53.2.3 NEURONES AND SYNAPSES have reduced expression of insulin and insulin-like growth
factor (IGF) within the brain and reduced insulin- and IGF-
Neural regeneration and replacement is being attempted receptor sensitivity. In the brain, insulin and β amyloid also
with stem cell transplants/infusions and the delivery of share a common enzymatic degradation pathway (IDE),
nerve growth factors (NGFs) and other neurotrophic meaning that peripheral and central nervous system (CNS)
agents. It is reassuring that in regions of most intense neu- insulin resistance could contribute to the neuropathology
ral response to dementia pathology (e.g. around amyloid and clinical features of AD (Craft, 2005).
plaques), there are often neurones that can be revived and
any cell death is more from apoptosis than necrosis, mean-
ing damage to other cells is limited. However, the brain is 53.2.9 OTHER MECHANISMS
the most complex organ with respect to cellular structure,
There are innumerable other approaches to remediate
so neural regeneration or replacement may never be fully
pathology and symptoms in dementia, as well as therapies
possible.
to target non-AD forms of dementia (see Section 53.3.9).

53.2.4 INFLAMMATION
There is an inflammatory response around amyloid plaques 53.3 THERAPEUTIC TRIALS
in AD, proximal to ischaemic or hypoxic tissue with cere-
brovascular disorders, and inflammation may also occur
with other dementias. This presents potential targets, using The numerous potential targets described above have led to
anti-inflammatory agents to limit cell damage and modify a plethora of well-constructed trials of new dementia thera-
the clinical course of dementia. pies. This chapter concentrates on therapeutic areas where
trials have been conducted and results presented. However,
it is likely that many new targets and agents are being
53.2.5 MITOCHONDRIA developed and may soon have sufficient data to review. As
These organelles generate the energy required for cellular the field is constantly changing, some data are still as yet
processes and damage to mitochondria has been implicated unpublished and available only from conference proceed-
as one of the primary causes of cell damage in dementia. ings or media statements. We have preferentially included
Mitochondria have their own genes and are the site of references to large pivotal publications; however, other
numerous metabolic pathways, many of which present information contained within this chapter has been sourced
potential targets for dementia therapies. from the public domain including alzforum.org and clini-
caltrials.org.
53.2.6 OXIDATIVE PROCESSES
53.3.1 AMYLOID
Closely linked with mitochondrial function, all cells gen-
erate damaging oxidative species and have regulatory Anti-amyloid approaches include those that reduce produc-
antioxidant mechanisms. AD therapies that enhance this tion of Aβ, that increase clearance and that mitigate its del-
antioxidant response may protect cells from damage and eterious effects. Some may also exert their effects through
alter dementia’s course. multiple mechanisms (e.g. CHF5074, a γ-secretase modu-
lator, also has effects on neurogenesis, reduces tau and has
53.2.7 NEUROTRANSMITTERS AND anti-inflammatory properties).
RECEPTORS
β -Secretase
53.3.1.1 
Symptomatic AD therapies to date have targeted a small
number of neurotransmitters and their receptors, and other The BACE1 enzyme releases the major portion of APP and
neurotransmitter symptoms are potential targets for new subsequently the γ-secretase complex liberates Aβ – either
symptomatic therapies. These include the various histamin- Aβ40 or the more toxic and aggregation-prone Aβ42.
ergic, 5-hydroxytryptamine (5-HT) and glutamatergic neu- Interestingly, ‘proof of mechanism’ for BACE1-inhibition
rotransmitters and receptors. has been demonstrated in individuals expressing A673T
Drug treatments in development for Alzheimer’s disease 557

mutations in the APP gene. This mutation, proximal to the is change CDR-SB relative to placebo. Secondary outcome
BACE1 cleavage site, prevents the action of BACE1, reduc- measures include time to clinical diagnosis of probable AD
ing Aβ production in vitro by 40% and protect against the dementia, as well as change in cognitive (three-domain
development of cognitive decline (Jonsson et al., 2012). composite cognition score), functional (ADCS-ADL score)
BACE1 is very large, which presents difficulties in pro- and biomarker (hippocampal volume) measures, with
ducing an inhibitor capable of crossing the BBB. In addi- optional PET and CSF substudies as per EPOCH. The esti-
tion, many early BACE1 inhibitors were also substrates for mated completion date is March 2018.
p-glycoprotein-mediated efflux from the CNS, impeding
attempts to achieve therapeutic concentrations at its site of 53.3.1.1.2 E2609
action (Vassar et al., 2009). Several Phase I trials have been completed for the Eisai
The enzyme has been crystallized and knowledge of its BACE1-inhibitor E2609, comprising 336 healthy controls
structure has assisted rational drug development, with sev- and people with early AD. In the first, single ascending-dose
eral small molecule inhibitors that bind to the active enzy- study of 73 healthy adult participants, E2609 reduced plasma
matic site now in trials. Aβ by between 52% (5 mg dose) and 92% (800 mg dose) rela-
Recent Phase I data presented from Eisai/Biogen (E2609), tive to baseline levels. In a second, multiple ascending dose
Merck (Verubecestat/MK-8931), Astra Zeneca (AZD3293) study of 50 healthy adults administered 25–400 mg/day for
and Boeringer Ingelheim/Vitae (BI1181181) are very promis- 14 days; there were both reductions in plasma Aβ and a dose-
ing, with each demonstrating significant reductions in plasma dependent reduction in CSF Aβ of up to 80% compared with
and CSF Aβ. However, as BACE1 also has other substrates, placebo. No clinically significant adverse effects were noted;
there are potential for side effects. BACE knockout mice however, headache and dizziness were the most common
exhibit behavioural change, seizures, myelination abnor- adverse events (AEs) reported. A third Phase I study tested
malities and retinal vascular pathologies. In mice treated a single dose of E2609 in 65 participants with MCI with bio-
with high doses of BACE1-inhibitors, dendritic spine loss and marker evidence of AD, with the primary outcome measure
altered synaptic function have also been observed (although change in CSF Aβ levels 36 hours post-dose. Although this
these findings did not occur at lower doses). Careful safety trial was completed in October 2013, no data have been dis-
monitoring of in-human studies is essential (Hu et al., 2010)! closed to date. Despite this, a Phase II trial has commenced,
In 2013, Eli Lilly and Company terminated trials of enrolling participants with MCI due to AD or mild AD
LY2886721 due to hepatotoxicity, which emerged in almost dementia. Screening includes amyloid-PET, and the primary
10% of patients. In the same year, Roche terminated devel- outcome is AD Composite Score (ADCOMS), a cognitive
opment of its RG7129 in Phase I, without explanation. composite score. Secondary outcomes include change in hip-
pocampal volume (volumetric MRI) and CSF Aβ from base-
53.3.1.1.1 VERUBECESTAT/MK-8931 line. This trial is estimated to be completed in 2019.
In Phase I trials in healthy volunteers, the Merck BACE1
inhibitor MK-8931 reduced CSF Aβ by over 90% without 53.3.1.1.3 AZD3293
dose-limiting side effects. Following the withdrawal of its BACE1 inhibitor (LY2886721),
MK-8931 is currently in Phase II/III trials in both mild- Eli Lilly teamed up with Astra Zeneca in a Phase II/III trial of
to-moderate AD (EPOCH) and prodromal AD (APECS). their compound, AZD3293. In Phase I studies of AZD3293,
In December 2013, Merck announced that the EPOCH CSF Aβ levels were reduced by 75% and took 2–3 weeks to
study had met its initial safety requirements, following a return to baseline after treatment. Named AMARANTH,
3-month safety substudy of the first 200 patients. The EPOCH this large Phase II/III trial will compare AZD3293 (20 mg or
study is now continuing recruitment towards the proposed 50 mg orally daily) to placebo in 1551 participants with MCI
1960 patients. The primary outcomes are change in cognitive due to AD or mild AD, over 2 years. The screening phase will
(Alzheimer’s Disease Assessment Scale-Cognition [ADAS- include biomarker evidence of AD (CSF or PET). Primary
Cog]) and functional (Alzheimer’s Disease Cooperative Study outcome is CDR-SB, and secondary measures include ADAS-
– activities of daily living [ADCS-ADL]) measures from Cog, ADCS-ADL, Functional Activities Questionnaire
baseline. Secondary measures include change in the sum of (FAQ), NPI, as well as change in CSF biomarkers, functional
boxes of the Clinical Dementia Rating (CDR-SB), total hip- and amyloid PET and MRI. The study is estimated to be com-
pocampal volume (volumetric magnetic resonance imaging pleted by May 2019.
[MRI]), Neuropsychiatric Inventory (NPI) and Mini-Mental
State Examination (MMSE) score. The study also includes two 53.3.1.1.4 BI1181181
optional substudies examining change in CSF biomarkers and Boehringer Ingelheim recently reported results of two
total tau, brain amyloid burden on 18F-flutemetamol PET. Phase I trials of its BACE inhibitor, BI1181181. This com-
The APECS study began in November 2013. It aims to pound also shows promise; it was well-tolerated, with a
recruit 1500 people diagnosed with amnestic mild cognitive greater than 80% reduction in CSF Aβ in healthy controls.
impairment (aMCI) with positive 18F-flutemetamol PET In 2015, BI1181181 was placed on a temporary clinical hold
amyloid scans (prodromal AD), to receive either 12 or 40 mg to investigate skin reactions seen in some participants in in
MK-8931 daily for 2 years. The primary outcome measure a subsequent ascending dose study.
558 Dementia

53.3.1.1.5 LY3202626 To date, at least six agents influencing the γ-secretase


LY3202626 (Lilly) is another small-molecule BACE1- enzyme have entered human studies. Two of these,
inhibitor. In 2016 it entered Phase II in participants with semagacestat and avagacestat, advanced to Phase III trials,
mild AD (with an amyloid biomarker). The study’s primary but ultimately were unsuccessful.
outcome measure is change in uptake seen on Tau PET as a
53.3.1.2.1 SEMAGACESTAT
marker of disease progression.
Semagacestat (previously known as LY450139) was noted
53.3.1.1.6 THALIDOMIDE to lower CSF Aβ in a Phase I trial in healthy individuals
Thalidomide has diverse properties including immunomod- (Bateman, 2009). However, in Phase II trials in mild-to-
ulation and anti-angiogenesis. It was marketed as an over- moderate AD, there was an effect seen with plasma but not
the-counter anti-emetic in the 1950s and was responsible for with CSF Aβ (Fleisher et al., 2008).
approximately 10,000 cases of phocomelia (congenital limb Two large Phase III trials of semagacesat involving over
malformation) worldwide, prompting its withdrawal. It has 3000 AD patients, IDENTITY-1 and IDENTITY-2 were
since shown efficacy in several autoimmune conditions, as terminated prematurely due to lack of efficacy and adverse
well as multiple myeloma and other cancers. effects. The active treatment was associated with a signifi-
It has effects on numerous inflammatory mediators cantly greater risk of skin cancer and infections, as well as
including tumour necrosis factor-α (TNF-α), which is impli- gastrointestinal symptoms. In addition, ADAS-Cog and
cated in AD. It also appears to reduce the activity of BACE1. ADCS-ADL outcomes were worse in the treatment groups
In transgenic mice, chronic administration of thalido- than placebo (Doody et al., 2013).
mide led to decreased activation of microglia and astro-
cytes, as well as reductions in both quantity and activity 53.3.1.2.2 AVAGACESTAT
of BACE1, with subsequent reduction in total and fibrillar Avagacestat (BMS-708163) is an arylsulfonamide γ-secretase
Aβ (He et al., 2013). A Phase II/III study of thalidomide in inhibitor, reported to selectively target the enzyme’s APP
patients with mild-to-moderate AD (MMSE 12–26) with substrate, sparing Notch. In animal models and Phase I tri-
CSF and plasma biomarker outcome measures was com- als, it was found to reduce CSF Aβ levels without Notch-
menced in 2010; however, recruitment has proved challeng- related toxicity.
ing and no data are available to date. Two Phase II trials were conducted in 2009 – one in
53.3.1.1.7 MINOCYCLINE mild-to-moderate AD and another in prodromal AD (using
CSF biomarkers for enrichment). The first, involving 209
The tetracycline antibiotic, minocycline, also inhibits BACE1
participants from across 41 sites, showed a higher rate of
and has anti-inflammatory properties demonstrated in mouse
adverse effects (skin cancer – including non-melanoma skin
models (Ferretti et al., 2012). It is currently in Phase II (mino-
cancers – and gastrointestinal symptoms) and poorer cog-
cycline 200 mg/400 mg/placebo, n = 480) in participants with
nitive outcomes in those receiving highest doses, similar to
very mild AD, with cognitive and functional end points.
semagacestat.
Out of 100 AD participants receiving avagacestat, three
γ-Secretase inhibition/modulation
53.3.1.2  (compared with 0/29 placebo) developed vasogenic edema
γ-Secretase is a complex of four proteins – presenilin, nica- (VE). This suggests that some target engagement (i.e. mobi-
strin, Aph-1 and Pen-2 – that cleaves the transmembrane lization of Aβ) may have occurred as a response to the
region of APP to produce Aβ (Panza et al., 2009). It also has treatment (see Section 53.3.1.7), although the CSF results
numerous substrates apart from APP, including some (e.g. reported were not statistically significant (Coric et al., 2012).
Notch) that are vital to embryogenesis, cell differentiation The second trial was halted following an interim analysis of
and communication. If the components of γ-secretase can 263 participants, which also demonstrated higher rates of
be differentially inhibited, this may enable lowering of Aβ skin and gastrointestinal effects, with no change in rate of
production, with sparing of its other functions, and avoid- progression to dementia (Coric et al., 2013).
ance of unwanted toxicities.
New molecules targeting γ-secretase may not fully inhibit 53.3.1.2.3 TARENFLURBIL
the enzyme, but act more as a modulator of the enzyme’s Tarenflurbil (Flurizan) was originally developed as an
function. As γ-secretase is capable of cleaving APP at sev- anti-inflammatory with anti-amyloid effects. In fact, it
eral sites, its Aβ product may vary in length, from Aβ38 is primarily a selective amyloid-lowering agent through
to Aβ42. γ-Secretase modulators promote production of modulating γ-secretase, with no anti-inflammatory
shorter Aβ fragments over the aggregation-prone Aβ42. By effects (Weggen et al., 2003). Tarenflurbil decreased brain
sparing Notch, unwanted adverse effects are avoided, while levels of Aβ42 and improved spatial learning and mem-
maintaining therapeutic efficacy. Intriguingly, a substrate- ory performance in mouse models of AD (Weggen et al.,
labelling technique has suggested that some γ-secretase 2003; Kukar et al., 2007). A 3-week Phase I study in 48
modulators may also reduce Aβ aggregation as well as Aβ healthy volunteers using doses up to 800 mg twice daily
production (Kukar et al., 2008). were as well-tolerated as placebo and higher plasma drug
Drug treatments in development for Alzheimer’s disease 559

levels were related to lower plasma Aβ42 levels (Galasko α-Secretase enhancement
53.3.1.3 
et al., 2007). A 12-month Phase II study in 210 subjects
on stable doses of ChEIs again using doses up to 800 mg The α-secretase enzyme processes APP in a non-­
twice daily, showed a significantly lower rate of decline in amyloidogenic pathway by cleaving APP near the middle of
activities of daily living (ADCS-ADL) and global function the Aβ portion, generating the peptide sAPP-α, and prevent-
(CDR-SB) compared to placebo, along with an insignifi- ing Aβ formation. Thus, a potential AD therapy is α-secretase
cantly slower rate of cognitive decline (ADAS-Cog), in the enhancement (Chiang and Koo, 2014). Unfortunately there
milder AD patients (Wilcock et al., 2008). In a 12-month is only a limited amount of natural α-secretase so augmen-
extension all patients were treated with tarenflurbil and tation may not lead to a large reduction of Aβ. However, the-
when those treated for the full 24 months with the agent oretically it is possible to introduce extra α-secretase, akin to
were compared with the delayed start group, there was enzyme replacement therapy in deficiency disorders. Such
a significantly (p < 0.001) slower rate of decline on each replacement therapy could be genetically based.
of the primary outcome measures, suggesting a disease-
modifying effect. 53.3.1.3.1 ETAZOLATE
Unfortunately, the 18-month Phase III trial results Etazolate (EHT-0202, Exonhit) is a pyrazolopyridine-
using 800 mg tarenfurbil twice daily were negative. In one derivative α-secretase activator EHT-0202, which also has
of the two large studies, in 1684 randomized participants, affinity for multiple other receptor-binding sites including
tarenfurbil had no beneficial effect on either of the two GABA A, adenosine A1 and A2 and phosphodiesterase (PDE)
primary outcomes: difference in change from baseline to E4. It has neuroprotective properties in vitro and increases
month 18 versus placebo for the ADAS-Cog and for the levels of sAPP-α (the ‘non-amyloidogenic pathway’). In
ADCS-ADL. There was also no significant benefit seen Phase IIa (n = 159, mild-to-moderate AD, adjunct to ChEI),
in any of the secondary outcomes (Green et al., 2009). EHT0202 (40 mg or 80 mg twice daily) etazolate was gen-
The second large Phase III trial was also negative. It is erally well-tolerated, although at higher doses there were
highly unlikely that this drug will be further developed increased CNS AEs and withdrawals. No significant cogni-
for AD. tive effects were seen, although the study was not powered
for efficacy (Vellas et al., 2011).
53.3.1.2.4 EVP-0962
EVP-0962 (FORUM Pharmaceuticals) is another NSAID- 53.3.1.3.2 ACITRETIN
derivative γ-secretase modulator. In vitro, it increases lev- Acitretin (Stiefel laboratories) is a vitamin A derivative
els of CSF Aβ-38 while reducing Aβ42, without altering (retinoid) currently indicated for treatment of psoriasis. It
the total Aβ level or affecting Notch. Treatment with EVP- also increases expression of the α-secretase, a disintegrin
0962 in Tg2576 transgenic mice resulted in reductions in and metalloproteinase 10 (ADAM10). In a small Phase II
Aβ aggregates and plaques, reduced neuroinflammation study, 21 participants with mild-to-moderate AD (MMSE
(astrocyte and microglial activation) and reversed memory 14–27) were randomized to acitretin 30 mg/day or placebo
deficits (Rogers et al., 2012). Phase I and II trials have been for 4 weeks. Those on drug had significant increases in CSF
undertaken in healthy adults and in those meeting National sAPP-α levels compared with the placebo group (difference
Institute on Aging – Alzheimer’s Association (NIA-AA) cri- 0.38, 95% confidence interval [CI] 0.03–0.72, p = 0.035) and
teria for MCI due to AD. Primary outcomes are tolerability it was well-tolerated (Endres et al., 2014).
and safety as well as concentration and rate of synthesis of
Aβ in CSF. 53.3.1.4 RAGE antagonists
53.3.1.2.5 NIC5-15/PINITOL RAGE is a multi-ligand receptor on cell surfaces. Ligands
NIC5-15/Pinitol (James J. Peters Veterans Affairs Medical recognized by RAGE include Aβ, which is first released
Center/National Center for Complementary and Alternative from APP extracellularly, so uptake into cells may be inhib-
Medicine/Humanetics Corporation) is a cyclic sugar alco- ited by RAGE antagonism, reducing intracellular amyloid
hol (in the same class as sweeteners sorbitol and xylitol) load and the inflammatory effects of Aβ (Deane, 2012).
that is found naturally in legumes, soy and pine trees. It
has insulin-sensitising and anti-inflammatory proper- 53.3.1.4.1 
A ZELIRAGON/TTP488/PF-04494700
ties and may also influence γ-secretase activity (Do et al., Azeliragon/TTP488 is a small-molecule RAGE antagonist,
2008). originally developed by TransTech Pharma for diabetic
Although no clinical trials data have been published, neuropathy, and subsequently licensed to Pfizer. In APP
Phase IIa results were presented in 2009, reporting good transgenic mice, TTP488 treatment was associated with a
tolerability and cognitive benefits in a very small trial of 15 significant reduction in inflammatory markers (TNF-α,
mild-to-moderate AD patients. A second Phase IIa trial is in TGF-β and IL-1) and CNS Aβ deposition (Sabbagh et al.,
progress (n = 40, mild-to-moderate AD), to assess pharma- 2011). In humans, it progressed to a large 18-month Phase
cokinetics/pharmacodynamics, cognitive and biomarker IIb trial, which recruited 399 patients randomised to one of
(plasma Aβ) outcomes. two oral TTP488 dosing regimes (60 mg for 6 days followed
560 Dementia

by 20 mg/day; 15 mg for 6 days followed by 5 mg/day) or (or deleterious). Additionally, full-length Aβ immuniza-
placebo. The trial was terminated early after interim analy- tion may be over-activating the immune system, caus-
ses suggested increased adverse outcomes (falls, confusion, ing the meningoencephalitis seen (Orgogozo et al., 2003)
cognitive decline) in the higher-dose group and lack of effi- and compromising potential clinical improvements from
cacy (but no safety concerns) in the lower-dose group. Later plaque removal.
analyses revealed slight decreased decline on the ADAS- Thus, more recent active immunization approaches
Cog in the lower-dose group at 18 months, but difference have utilized Aβ fragments that are less likely to activate
in CR-SB, MMSE or ADCS-ADL and no change in CSF unwanted (T-cell-mediated) immune responses.
biomarkers or hippocampal volume (Galasko et al., 2014). ACC-001, also known as vanutide cridificar (Janssen/
A Phase III study of TTP488 in mild AD began recruit- Pfizer/Wyeth/Elan), used N-terminal fragments of Aβ
ment in 2016, with ADAS-Cog and CDR-SB as co-primary (residues 1–7), whereas the T-cell lymphocyte response seen
endpoints. with AN1792 targeted Aβ towards the C-terminus (residues
15–42).
53.3.1.5 Active immunization (vaccination) 53.3.1.5.2 ACC-001
Antibodies against Aβ can be induced by injecting Aβ or its In preclinical trials, vaccination with ACC-001 led to the
fragments, to stimulate an immune response. The antibod- production of anti-Aβ antibodies but did not generate
ies generated may modify the amyloid cascade, preventing a T-cell-mediated response. It also reduced plaques and
AD progression (Schenk et al., 1999). The first trial of the improved cognition in transgenic mice (Panza et al., 2014).
active immunization against Aβ began after transgenic ani- Several Phase II trials have been conducted and results were
mal studies showed that such an approach prevented depo- presented. One study included 125 participants with mild-
sition of amyloid (if used early) and reduced amyloid load (if to-moderate AD, receiving six intramuscular injections
used later), leading to clinical benefits including prevention of either placebo, 3 mg or 10 mg ACC-001 with the adju-
of memory loss (Morgan et al., 2000). vant QS-21, with 2-year follow-up. Ninety percent of those
receiving the active vaccine produced a measurable antibody
53.3.1.5.1 AN-1792 response, and levels of Aβ in plasma correlated with anti-
A Phase I clinical trial of immunotherapy of patients with body titres, suggesting it was being cleared from the brain.
AD, using full-length synthetic Aβ42 as the antigen (AN- Correspondingly, there was a dose-dependent reduction in
1792), showed a highly variable development of antibodies accrual of brain β-amyloid (measured with florbetapir PET)
(Bayer et al., 2005) but no significant clinical benefits (though in treated subjects, although at trend-level significance only.
the trial was not powered for clinical end-points). A subse- There was also some suggestion of influence on downstream
quent Phase IIa study was halted when 6% of the patients processes, with a (non-significant) reduction in levels of CSF
developed a sterile meningoencephalitis (Orgogozo et al., p-tau. As seen with other agents, the treatment group had
2003), but showed no major differences in cognitive outcomes greater brain atrophy at higher doses, though the signifi-
when antibody responders were compared with the placebo cance of this is unclear. Six percent of the active group also
group after 1 year, despite evidence of high serum antibodies developed imaging evidence of VE (also known as Amyloid-
to Aβ42 in a group who received active treatment (Hock et al., Related Imaging Abnormalities-oEdema, ARIA-E [see
2003). Follow-up 4 years later demonstrated small differences Section 53.3.1.6.1]), and there were no improvements seen
in functional decline in those with a positive antibody titre in cognition (although not powered to detect cognitive out-
response only (25/129 participants) but no significant differ- comes). Despite some biomarker evidence of efficacy, clini-
ence in cognitive end points (Vellas et al., 2009). cal development has been discontinued (Ketter et al., 2014).
Neuropathological examination of 10 Phase I participants
who had received active immunization revealed removal 53.3.1.5.3 AD02
of amyloid plaque proportional to the antibody response. As with ACC-001, AD02 (Affiris/GSK) mimics the
There was no reduction in neurofibrillary tangles seen within N-terminal (residues 1–6) of Aβ, without the T-cell epit-
neuronal cell bodies; however, reduction in p-tau was noted ope. In Phase I trials to 12 months it was generally safe and
within neuronal processes (Boche et al., 2010). well-tolerated, with no meningoencephalitis. A Phase II
The finding that plaque removal is not enough to halt trial of AD02 in 332 patients with early AD, however, was
progressive neurodegeneration in AD poses challenges negative on its primary and secondary outcome measures.
to the amyloid hypothesis. It is possible that a specific Intriguingly, although data are yet to be published, partici-
amount of amyloid amyloid may be needed to initiate, pants receiving one of the placebo formulations (adjuvant
but not to maintain, progressive neurodegeneration. It is without vaccine) demonstrated superior performance to
also hypothesized that it is oligomeric Aβ, rather than the all of the other groups – including a second placebo group
fibrillar Aβ in plaques, that is responsible for most neuro- and three receiving the active AD02 treatment (including
degeneration in AD and that as immunization had failed with the adjuvant). Participants administered this pla-
to reduce oligomeric Aβ (or indeed increases this as Aβ cebo formulation (now identified as alum and renamed as
in plaques is mobilized), this approach may be ineffective ‘AD04’) demonstrated less cognitive decline (composite of
Drug treatments in development for Alzheimer’s disease 561

ADAS-Cog and ADCS-ADL) and hippocampal atrophy over nasopharyngitis were common. The vaccine does not stim-
18 months. Vaccines are commonly administered along with ulate an Aβ-specific T-cell response, and there have been no
an adjuvant, to stimulate an immune response and improve cases of meningoencephaitis or VE (although one case of
generation of antibodies. Alum is an aluminium salt (typi- intracerebral haemorrhage has been reported, not related
cally potassium aluminium sulphate) used as an adjuvant in to antibody titres). The study authors suggest that CAD106
many vaccines. While trials of AD02, including an exten- may potentially be amenable to long-term administration in
sion study, have been terminated, Affiris announced plans AD (Winblad et al., 2014).
to attempt to replicate the AD004 findings in its own trial To further investigate this hypothesis, in collaboration
(Schneeberger et al., 2014). This concept has been suggested with the Banner Alzheimer’s Institute, Novartis is planning
already in animal models – Frenkel et al. (2008) administered to use CAD106 as a treatment arm in a Phase II/III trial
an intranasal adjuvant (‘Protollin’, derived from lipopolysac- recruiting more than 1300 cognitively normal individual
charide and Neisseria meningitidis outer membrane proteins) aged 60–75, at genetic risk of AD (APOE ε4 homozygotes).
to transgenic mice, resulting in reduction in soluble and
fibrillar Aβ. Other techniques of immune stimulation may 53.3.1.5.5 ACI024
also be effective (see Section 53.3.1.9.1). ACI024 is a liposomal vaccine, with N-terminal Aβ frag-
ments (Aβ1–15), attached to palmitoylated lysine residues,
53.3.1.5.4 CAD106 incorporated into a phospholipid bilayer. In preclinical
Perhaps the most advanced active vaccine still in active tri- studies, it induced high titres of oligo-specific anti-Aβ anti-
als is CAD106 (Panza et al., 2014). This vaccine contains bodies, without causing cellular inflammation, increases in
multiple copies of the N-terminus Aβ epitope (residues 1–6), pro-inflammatory cytokines or microbleeds. In addition, it
conjugated to a carrier protein (180 copies of the Q-β coat decreased concentrations of Aβ40 and Aβ42 and improved
protein) that confers additional immunogenicity. cognition (Hickman et al., 2011).
In animal studies, CAD106 was demonstrated to induce In humans, a Phase I/II trial of up to 198 AD participants
anti-Aβ antibodies, without provoking a T-cell-mediated is currently recruiting. Individuals are to be screened using
direct toxicity and reduced amyloid accumulation. It was β-amyloid PET (florbetaben [FBB]), on stable doses of acetyl-
most effective when administered to the transgenic mice cholinesterase medication. Primary outcome measures will
prior to the appearance of Aβ plaques (Wiessner et al., 2011). include safety outcomes, antibody response titres and change
Several Phase I and II trials have been conducted to date. in neuropsychological test battery (NTB) at 12 months, with
In one Phase I trial, 58 participants with mild-to-­ secondary end points including CSF Aβ and tau, volumetric
moderate AD (MMSE 16–26) received three doses of MRI and change in brain amyloid on FBB PET.
CAD106 (50 or 150 μg) over 100 days. They showed dose-
dependent generation of anti-Aβ immunoglobulin G (IgG) 53.3.1.5.6 UB-311
titres, with 82% at 150 μg and 67% at 50 μg reaching a United Biochemical’s UB-311 uses two synthetic T-helper (Th)
pre-specified ‘response’ threshold. Total plasma Aβ con- epitopes (UBITh®), bound to the N-terminal Aβ1–14 fragment.
centration increased with treatment, whereas free plasma It also uses a proprietary delivery system (CpG oligonucle-
Aβ decreased (implying mobilization of brain Aβ, with otide) that promotes Th type 2 (regulatory T-cell) responses
increased proportion of antibody-bound Aβ in plasma) in preference to the Th type 1 (pro-inflammatory) response.
(Winblad et al., 2012). In a very small Phase I study with an observational extension
Further Phase II studies have been conducted, including phase, 19 Taiwanese patients with mild-to-moderate AD aged
PET and pharmacogenomic substudies, and both subcu- 50–80, were administered three 300 μg doses of UB-311 intra-
taneous and intramuscular administration. Similar to the muscularly (weeks 0, 4 and 12). All participants demonstrated
Phase I trials, about 90% of participants developed adequate Aβ1–14 specific antibody responses, which were still detect-
antibody responses and those with the highest antibody able at week 48, with no serious adverse effects. In addition, in
titres demonstrated the highest increases in total plasma older subjects with mild AD (n = 6; age ≥60 years with baseline
Aβ. In vitro studies of antibodies taken from participants MMSE ≥ 20) there were small improvements in ADAS-Cog,
during these studies demonstrate that there is improved ADCS–Clinical Global Impression of Change (ADCS-CGIC)
antibody maturation (increased affinity to target) seen with and MMSE, although the numbers reported were very small.
time and repeated administrations. Intramuscular admin- A Phase IIa study is planned.
istration was better tolerated and generated a greater anti-
body response than subcutaneous administration. 53.3.1.5.7 LU AF20513
There were no differences seen in CSF biomarkers with One drawback limiting the efficacy of active immunization
treatment, although the authors contend that the timing of is that the immune response declines with ageing, producing
sampling was chosen for safety reasons and was not optimal fewer antibodies and requiring repeated doses. To address
to detect biomarker changes (Winblad et al., 2014). this challenge, Lundbeck has developed Lu AF20513, which
Following up to seven administrations, and 2.5 years’ combines Aβ1–12 with Th epitopes from the tetanus toxin,
follow-up, safety is reported as favourable, although mild aiming to stimulate existing memory-Thcells to promote
adverse effects including injection site erythema and B-cell production of antibody. Preclinical studies have
562 Dementia

had promising results, with robust antibody generation protocol MRI surveillance, with 7 asymptomatic and 5 hav-
and reduction in Aβ40 and Aβ42 (soluble and Aβ plaques) ing reported transient AEs (including confusion, headache
(Winblad et al., 2014). or visual disturbance) in the preceding weeks.

53.3.1.5.8 V950 53.3.1.6.2 AMYLOID-RELATED IMAGING


Merck has also completed a Phase I trial with its multivalent ABNORMALITIES (ARIA)
Aβ peptide vaccine, V950; however, no further trials have In response to these findings and in accordance with guid-
been initiated. ance issued by the U.S. Food and Drug Administration
(FDA), the Alzheimer’s Association Research Roundtable
53.3.1.6 Passive immunization convened a workgroup in July 2010. The term ‘Amyloid-
Related Imaging Abnormalities’ (ARIA) was coined, incor-
An approach that bypasses the need to respond to an antigen porating microhaemorrhage and haemosiderosis (ARIA-H)
and, variably, produce antibodies is to inject the antibod- and VE and effusions (ARIA-E).
ies themselves. This can be done by using pooled immuno- A subsequent review of all MRI images from all Phase II
globulin, which contain anti-Aβ antibodies, or developing participants identified these lesions in 17% of those treated,
monoclonal antibodies (mAbs) against Aβ. These mAbs, of whom 78% were asymptomatic, with coincident micro-
usually produced from animal immunoglobulin, can be bleeds or haemosiderosis noted in 47%. ARIA-E was more
humanized and modified in other ways (e.g. by altering the common with higher doses of bapineuzumab and in APOE
Fc or ‘backbone’ component) that can impact on efficacy ε4 homozygotes. There was no association between ARIA-E
and reduce unwanted immune/inflammatory effects. and age, gender or baseline white matter disease on MRI,
Currently mAbs in clinical trials are directed against and presence of ARIA-H at baseline did not increase risk
different conformational epitopes (binding sites) com- of incident ARIA-E (although participants with multiple
prised by different regions of the Aβ peptide (e.g. the C-or microbleeds were excluded from participation).
N-terminus or the midportion). This provides different effi- The cases identified during the bapineuzumab trial were
cacy against the various forms of Aβ, from monomers (­single perhaps the most prominent, although ARIA-E have also
units) through soluble aggregated oligomers/­ protofibrils been reported with another Aβ immunotherapeutic agent
(dimers, tetramers etc.) to insoluble fibrillar (plaque) Aβ. (solanezumab) (Chen et al., 2012) and a γ-secretase inhibi-
Unfortunately, while there have been several prominent tor (Freiherr et al., 2013). Similar phenomena had also been
negative clinical trials to date, some have provided glim- described with active immunotherapy in humans and ani-
mers of positivity from encouraging subgroup analyses. mal models, and rare spontaneous case reports associated
Others are yet to release results or are still under way. with neurological symptoms and evidence of CAA (Gold
et al., 2010; Sato et al., 2011; Moon et al., 2012; McClean
53.3.1.6.1 BAPINEUZUMAB et al., 2014; Shah et al., 2014).
Bapineuzumab is a humanized form of a murine mAb Although ARIA-E commonly preceded or coincided
directed against the N-terminus (residues 1–5), binding to with ARIA-H, the two findings were not necessarily co-
both soluble and fibrillar Aβ. Preclinical studies demon- located, suggesting a generalised disruption of vascular
strated that 3D6 (the murine mAb) could bind to and reduce integrity, rather than a focal insult. It is hypothesized that
burden of amyloid plaques, mitigate of Aβ-induced synap- removal of amyloid from vascular deposits may lead to
totoxicity and improve behaviour. causing transient leakiness and subsequent VE. An alterna-
In Phase II (124 participants; mild-to-moderate AD; 6 tive hypothesis is that there is failure of saturable paravas-
infusions; 78 weeks follow-up) trial, there were no differ- cular Aβ clearance mechanisms, by massive mobilization of
ences seen with the two pre-specified primary end points soluble from sequestered Aβ. Hence, waste that is otherwise
(ADAS-Cog and disability assessment for dementia [DAD]); soluble (e.g. Aβ) accumulates, causing altered vascular per-
however, post hoc analyses suggested benefit in APOE ε4 meability and leakage of plasma and blood products; see
non-carriers and in those completing all six infusions. Figure 55.6 (Li et al., 2010).
Biomarker substudies showed some reduction in brain Aβ Two large Phase III trials were conducted, involving 2452
burden (11C-PiB PET), a trend to reduced CSF p-tau (p = patients with mild-to-moderate AD (one in APOE ε4 carri-
0.056), but no difference in CSF Aβ or total tau. MRI vol- ers, n = 1121 and one in non-carriers, n = 1331) to receive
umetric analyses showed less brain volume loss in treated intravenous bapineuzumab or placebo every 13 weeks for
APOE ε4 non-carriers compared with placebo (p = 0.004). 78 weeks.
Bapineuzumab was generally well-tolerated; however, in Unfortunately, there was no difference between treat-
the higher-dose groups, MRI abnormalities consistent with ment and placebo groups in the primary outcomes of
VE were noted in 15 participants (3 Phase I and 12 Phase II). ADAS-Cog or DAD at 78 weeks in either study. Biomarker
Three presented with symptoms including confusion, head- results did, however, show less accumulation of Aβ (11C-PiB
ache and gait disturbance, prompting off-protocol MRIs. PET) and a reduced CSF p-tau in APOE ε4 carriers but not
One was treated with intravenous corticosteroids with reso- non-carriers. No treatment difference in volumetric MRI
lution of symptoms. The remaining 12 were identified in per was seen in either group.
Drug treatments in development for Alzheimer’s disease 563

So why did bapineuzumab fail? Some hypothesize that reduction in cognitive decline in individuals with mild AD
the safety concerns meant that lower-than-efficacious doses (MMSE 20–26) and a trend to reduced decline in function
were used. Others have suggested that like AN1792, the (ADCS-ADL).
treatment may have worked, but was initiated too late, past Although these differences are small and of limited clini-
the point at which neurodegeneration could be reversed cal significance, they were seen by many as an encouraging
or substantially modified. It should also be noted that a signal, targeting the amyloid hypothesis in AD may, in fact
high proportion of those in the PiB substudy fell below the still hold promise.
threshold attributed for a ‘positive’ amyloid scan (6.5% of The biomarker response outcomes showed increases in
carriers and 36.1% of non-carriers), suggesting possible total CSF Aβ42 and Aβ40 and increased plasma Aβ (bound
non-AD pathology in these individuals — despite meeting to antibody), supportive of the ‘peripheral sink’ hypothesis.
clinical criteria for AD-type dementia. This may partially Consistent with prior animal models, despite evidence of
explain the negative biomarker results in the APOE ε4- increasing peripheral Aβ, there was no measurable change in
negative group. If this was extrapolated to the wider trial fibrillar Aβ (using Amyvid-PET). There were also no changes
population, this may have significant impact on overall in CSF p-tau or MRI brain or hippocampal volumes.
efficacy. Of note, as with the bapineuzumab Phase III trial, about
Despite the negative outcomes, there are several lessons one in four participants did not demonstrate measurable
learned from the bapineuzumab study that has shaped sub- florbetapir (Amyvid) retention on PET imaging, which
sequent clinical trial design in AD. The phenomenon of would limit the effectiveness of the intervention.
ARIA is now better characterized, and central MRI moni- Following from these results, Expedition-3, a trial of
toring to assess for incident events has been widely adopted. solanezumab in mild AD with biomarker screening (Amyvid
Second, the use of CSF and PET biomarkers for enrichment PET) is under way, with results expected in late 2016.
or screening of clinical trial populations, a criticism of the Solanezumab has also been selected for use in two sec-
bapineuzumab study, is now a common practice. ondary prevention trials in asymptomatic (or very mildly
symptomatic) individuals at risk of AD: the Dominantly
53.3.1.6.3 PF-05236812/AAB-003 Inherited Alzheimer’s Network–Trials Unit (DIAN–TU)
To counter its adverse safety profile, Pfizer has modified and Anti-Amyloid in Asymptomatic Alzheimer’s Disease
the Fc component of bapineuzumab to dampen its effect on (A4) trials, both currently recruiting. DIAN-TU is a 5-year
microglial activation and hence, reduce incidence of ARIA Phase II/III trial, which is a collaboration between Eli Lilly,
(Crespi et al., 2014). The product, PF-05236812/AAB-003, is Roche and the Alzheimer’s Association. This is using solan-
being tested in a Phase I trial of 88 mild-to-moderate AD ezumab or gantenerumab (see Section 53.3.1.6.5) in 210
patients with subsequent open-label extension. Outcomes asymptomatic or mildly symptomatic APP/PS1/PS2 muta-
will include safety measures (including ARIA), as well as tion carriers, each responsibly for autosomal-dominant
cognitive and biomarker changes. forms of AD with consistent age at onset across generations.
Outcome measures will include biomarkers (at 2 years) and
53.3.1.6.4 SOLANEZUMAB cognitive outcomes (selected to be particularly sensitive to
Although bapineuzumab was targeted against the early cognitive changes in AD).
N-terminus of Aβ, solanezumab, derived from the mouse The A4 Study trial will apply solanezumab in a 3-year
mAb m266a, is directed against the mid-portion (Aβ16–24). trial of 1000 individuals, age 65 or older, with biomarker
It has affinity for small Aβ oligomers, but does not bind evidence of brain amyloid deposition (amyloid PET), but
to fibrillar Aβ plaques. As only a small proportion (0.1%) normal cognition (i.e. NIA-AA-defined Stage 2 or 3 preclin-
crosses the BBB, its predominant mechanism of action ical AD[Sperling et al., 2011]).
is by capturing Aβ40 and Aβ42 in the plasma, increasing
efflux of Aβ from the brain and (potentially) decreasing 53.3.1.6.5 GANTENERUMAB
deposited Aβ. The Roche IgG1 mAb, gantenerumab, binds to both N-terminal
Phase I and II trials demonstrated favourable safety and and adjacent central Aβ sequences. Although bapineuzumab
tolerability, with no episodes of ARIA. It proceeded to two and solanezumab are humanized murine antibodies, gan-
large Phase III studies, the EXPEDITION-1 and -2, involv- tenerumab is an entirely human antibody, selected from a
ing 2052 people with mild-to-moderate AD, to receive 400 synthetic human antibody library (huCAL®).
mg solanezumab intravenously each month for 80 weeks. This compound acts by recruiting microglia and macro-
Although the infusions were generally well-tolerated, phages to degrade fibrillar Aβ, both in parenchyma and ves-
ARIA-E occurred in 1%, the studies were negative in their sels. It also neutralizes inhibitory effects of oligomeric Aβ42
primary cognitive and functional end points (ADAS-Cog11 in vitro. It has the advantage of convenience of administra-
and ADCS-ADL). However in the EXPEDITION-1 study, tion by monthly subcutaneous injection (Ostrowitzki et al.,
a pre-specified secondary analysis in mild AD (MMSE 2012).
20–26), demonstrated reduced cognitive decline com- Phase I trials of 308 participants (healthy control vol-
pared with placebo. A subsequent analysis of pooled data unteers or AD patients) have been completed, with admin-
of mild AD participants from both studies suggested a istration either subcutaneous or intravenous infusion.
564 Dementia

In general, gantenerumab was well-tolerated and safe, how- The first, named ‘ABBY’ recruited individuals with mild-
ever, two of six participants in the highest-dose group dem- to-moderate AD to receive 300 mg of crenezumab subcu-
onstrated ARIA (Ostrowitzki et al., 2012). Over 6 months, taneously every 2 weeks (n = 122/62 placebo) or 15 mg/kg
this group also showed −35.7% mean difference in brain intravenously monthly (n = 163, 84 placebo). The second,
amyloid change compared to placebo. ‘BLAZE’ was a 91-patient biomarker substudy, with similar
Subsequently a Phase II/III trial commenced in 770 indi- treatment groups, using florbetapir PET as the primary out-
viduals with prodromal AD, as well as a Phase III study in come. Both were completed in 2014 and preliminary data
1000 patients with (clinically diagnosed) mild AD. presented.
The former, termed ‘SCarlet RoAD’, enrolled people Similar to the EXPEDITION-1 and -2 trials of solan-
aged over 50, with amnestic MCI (using the Free and Cued ezumab, ABBY, while negative in its primary end points
Selective Reminding Test [FCSRT-IR]), MMSE above 24 (ADAS-Cog12 and CDR-SB) demonstrated trends to
and CDR of 0.5 (mild or questionable dementia), with a cognitive differences in subgroup analyses of mild AD
positive disease biomarker (using amyloid PET). This was (MMSE 20–26), which reached statistical significance when
the first Phase III trial to utilize the Dubois research criteria restricted to those with MMSE of 22–26.
for prodromal AD (presence of episodic memory impair- Larger effects were seen with intravenous compared with
ment with biomarker evidence of pathology) (Dubois subcutaneous administration, which received about half the
et al., 2007). Co-primary end points for the trial were the effective dose.
CDR-SB and change in brain amyloid burden (using PET). The biomarker substudy, BLAZE did not show any differ-
Unfortunately, in December 2014, Roche announced that ence in its primary end point (brain Aβ by florbetapir PET);
SCarlet RoAD had failed a pre-specified interim futility however, there were significant increases in CSF Aβ with
analysis. This suggests that at the time of the analysis, the treatment over time compared with placebo. Considerable
intervention and placebo arms did not differ sufficiently to intra-scan variability was noted, prompting post hoc analysis
warrant the trial continuing. However, there were no new using different PET uptake standardization techniques (using
safety concerns. white matter, rather than cerebellar grey matter, as reference
Although previous trials have been set later in the regions to normalize the images). With these techniques, a
dementia trajectory and relied on clinical diagnosis alone, trend to reduced Aβ accumulation was seen with the intra-
SCarlet RoAD was applied in prodromal disease with a rig- venous, but not with subcutaneous treatment group. In both
orous screening process with biomarker support. Although ABBY and BLAZE, crenezumab was well-tolerated overall,
the reasons for study failure remain to be announced, there and there was only one single case of ARIA-E. There is now
have been some suggestions. As with bapineuzumab, gan- an open-label extension of 361 participants, to run until 2016.
tenerumab primarily targets fibrillar Aβ and not soluble Crenezumab is also being tested in preclinical Familial
species (e.g. oligomers) that may have greater impact clini- AD in the Alzheimer’s Prevention Initiative (API).
cal presentation. Second, as with bapineuzumab, gan- Commenced in 2013, this is an adaptive trial in pre-symp-
tenerumab may be associated with risk of ARIA at higher tomatic individuals carrying an autosomal-dominant
doses. So it is possible that while the doses used in Scarlet presenilin mutation to receive bimonthly subcutaneous
RoAD did not impact on safety, they may not have been Crenezumab (at higher dose than used in ABBY/BLAZE).
sufficient to demonstrate a therapeutic response. A third The study aims to recruit 300 participants, the major-
explanation could be that even though gantenerumab may ity of whom will be from a large kindred from Medellin,
work, the intervention’s timing (i.e. MCI due to AD) may be Colombia, with some also from the United States with simi-
still too late to impact on clinical outcome, and still-earlier lar mutations. The primary outcome is change in a cognitive
intervention is necessary (i.e. in at-risk individuals with composite score, with secondary outcomes including safety
normal cognition). measures, time to MCI and CSF/imaging biomarkers. It is
Although SCarlet RoAD is being terminated, a trial in set to run until 2020.
mild AD continues (Marguerite Road), and gantenerumab
is also being applied opposite to solanezumab in one of the 53.3.1.6.7 BAN2401
arms of the DIAN–TU, which is due to run until 2017. BAN2401 (Eisai/BioArctic) is a humanized IgG1 mAb that
binds to and neutralizes large, soluble toxic Aβ oligomers,
53.3.1.6.6 CRENEZUMAB also known as protofibrils. It has 1000-fold higher selec-
Crenezumab (MABT510A, AC Immune/Genentech/Roche) tivity for protofibrils over Aβ monomers. In cell culture,
is an IgG4 mAb, which recognizes both oligomeric and BAN2401 lowered the binding of Aβ protofibrils to hip-
fibrillar Aβ with high affinity, and it also has low affinity for pocampal neurones and neutralized neurotoxicity, and in
the single-unit Aβ monomers. It was designed to reduced mouse models, the murine form mAb158 reduced proto-
effector T-cell response, limiting complement activation fibril levels in CSF by 53%, without change in monomeric
and inflammation, while still stimulating microglial clear- Aβ142 (Tucker et al., 2015).
ance of Aβ (Adolfsson et al., 2012). After Phase I, studies In Phase I trials, BAN2401 was well-tolerated at all doses
showed the drug was well-tolerated, with no ARIA; higher (up to 10 mg/kg fortnightly and no episodes of ARIA were
doses were used in the Phase II trials. reported (Lannfelt et al., 2014).
Drug treatments in development for Alzheimer’s disease 565

The drug is currently in a Phase II trial, with an adap- aggregation into plaque, and is more resistant to clear-
tive study design testing five different doses of intravenous ance than Aβ42 (Gunn et al., 2010). In preclinical studies,
BAN2401 in biomarker-confirmed MCI due to AD and the murine equivalent, mE8, has been shown to clear Aβ
early AD (using amyloid-PET). Following recruitment of the plaques without causing microhaemorrhage (Demattos
first 200 participants, frequent interim analyses will dictate et al., 2012). A Phase I study in prodromal or mild AD with
group allocation of additional participants joining the study florbetapir PET screening on entry was commenced in 2013,
up to a recruitment target of 800. The primary outcome mea- with results expected in 2015.
sure will be cognitive composite, with secondary outcomes Other companies no doubt also have mAbs under devel-
including change in brain β amyloid burden (PET) and hip- opment, as these approaches target the core of the amyloid
pocampal volume (volumetric MRI) over 18 months. hypothesis. Failure would cast an even greater shadow over
this anti-amyloid approach, although a role in prevention is
53.3.1.6.8 PONEZUMAB still possible even if therapy of established AD is unsuccessful.
Ponezumab (PF-04360365, Pfizer/Rinnat) is a human-
ized IgG2A mAb against the free C-terminal amino acids 53.3.1.7 Polyclonal antibodies
33–40 of Aβ 140 that progressed to Phase II trials in mild-
to-moderate AD (La Porte et al., 2012). Safety outcomes Human immunoglobulin contains polyclonal anti-Aβ
were acceptable, with no ARIA reported; however, the drug antibodies (IgG and IgM classes) that bind to both oligo-
failed to demonstrate a significant effect on brain or CSF meric and fibrillar Aβ and three companies have trialled
Aβ, and its development for AD has been discontinued. human immunoglobulin infusions for the treatment of
There was evidence of a dose-dependent increase in plasma AD (Octagam, Octapharma; Gamunex, Grifols Biologicals;
Aβ40 however, which has prompted initiation of a Phase II Gammagard, Dexter Healthcare). Intravenous immuno-
study of ponezumab in patients with CAA (predominantly globulin preparations are already utilized for a range of
composed of Aβ40) (Burstein et al., 2013). other immune-mediated conditions, and it has been pre-
There are still more mAbs in Phase I/II development yet viously observed that individuals treated with immuno-
to reach Phase III trials, which show promise. These include globulin therapy for other reasons have a reduced risk of
BIIB037/aducanumab (Biogen), LY3002813 (Eli Lilly), subsequent AD (Fillit et al., 2009).
MEDI1814 (AstraZeneca) and SAR228810 (Sanofi).
53.3.1.7.1 GAMMAGARD
53.3.1.6.9 BIIB037/ADUCANUMAB The most advanced agent in trials is Gammagard (Baxter),
BIIB037/aducanumab (Biogen) has been developed by ‘reverse which was tested in two Phase III trials. The first was
translational medicine’, in that it is a naturally occurring fully run in conjunction with the U.S. ADCS group and tested
human IgG1 mAb against Aβ, acquired from cognitively Gammagard, 400 or 200 mg/kg against placebo over 18
healthy elderly donors. Phase Ib trial in biomarker-positive months (n = 390, mild-to-moderate AD). Unfortunately, the
participants with prodromal or mild AD (amyloid-PET) is study failed to show a difference in its primary (cognitive)
under way, and Biogen recently announced results of pre- end points, although there were some trends to improve-
specified interim analyses. They reported a dose- and time- ment in pre-specified subgroups (moderate AD, APOE
dependent reduction in brain Aβ (florbetapir-PET) at 26 and 54 ε4 carriers) and dose-dependent biomarker responses
weeks, which corresponded with a significant slowing of dete- (decrease in CSF Aβ42). Following the release of these out-
rioration in clinical measures (MMSE and CDR-SB). For the comes, the second Phase III trials were terminated.
MMSE, the placebo group declined by 3.1 points at 12 months,
compared with 0.6 points in the highest-dose group (p < 0.05). 53.3.1.7.2 OCTAGAM
For the CDR-SB, the change in score was 2.0 (placebo) and 0.6 Octapharma’s agent Octagam has completed Phase II tri-
(for 10 mg/kg aducanumab), respectively (p < 0.05). als in MCI patients, with initial results suggesting reduced
The incidence of ARIA-E was quite high, ranging from 5 atrophy over 12 months. However, no cognitive differences
to 55%, and was highest in APOE ε4 carriers and at higher emerged and no further trials have been announced. The
doses. Most participants with ARIA were asymptomatic or Grifols Biologicals agent, Gamunex, is currently still in
reported mild, transient symptoms only, and treatment was Phase II/III, estimated to run to 2016.
continued at lower doses. APOE ε4 carriers treated with
the highest dose (10 mg/kg) were most likely to discontinue 53.3.1.8 New modes of delivery of
treatment (35%). The other most common AE reported was immunotherapy
headache, in 22% of the intervention group compared with
5% of placebo. Biogen plans a Phase III trial in the near future A significant drawback of immunotherapy is the need to
(Biogen Press Release, 2015). infuse the treatment, usually intravenously. Aβ fragments
delivered mucosally (e.g. via the nose) have elicited an anti-
53.3.1.6.10 LY3002813 body response in mice, avoiding the need for parenteral
LY3002813 binds to a pyroglutamate-bound form of Aβ administration. An oral Aβ vaccine is also being developed,
(p3-42), which promotes formation of oligomers and but efficacy is not yet in the public domain.
566 Dementia

Other methods of delivery of Aβ, or mAbs, include uti- (Aisen et al., 2011). It has not been tested in preclinical or
lizing filamentous viral bacteriophages and modifying an prodromal AD, but tramiprosate is currently available over
adenovirus to include a ‘payload’ of antigen or antibody. the counter as a supplement to assist memory (Vivimind) in
the United States.
53.3.1.9 Other immunotherapies 53.3.1.10.2 ELND005/ADZ-103/SCYLLO-INOSITOL
Anti-Aβ DNA vaccination is a promising approach that is ELND005/ADZ-103/Scyllo-inositol is related to the natu-
being trialled. Additionally, an antibody has been devel- rally occurring inositol (found in fruit) that binds to and
oped from a subject with the British APP mutation that inhibits neurotoxic oligomeric Aβ42, and prevents aggrega-
interacts with various β-pleated protein sheets. This anti- tion and plaque formation. It also lowers brain myo-inositol
body potentially targets both amyloid and tau (as neurofi- levels, which are higher in AD, Down’s Syndrome and bipo-
brillary tangles) as both are β-sheets and it may thus have lar disorder, and correlate with behavioural and psycho-
benefits beyond those of individual anti-amyloid or anti-tau logical symptoms of dementia (BPSD) (Shinno et al., 2007).
therapies (Lambracht-Washington et al., 2013). In transgenic mice, ELND005 reduces Aβ plaque burden
and improves learning deficits (Salloway et al., 2011). In
53.3.1.9.1 GRANULOCYTE-MACROPHAGE COLONY- a Phase II trial, 353 patients with mild-to-­moderate AD
STIMULATING FACTOR (GM-CSF) AND received 500, 2000 or 4000 mg daily or placebo for 78 weeks.
GRANULOCYTE COLONY–STIMULATING Increased infections and deaths were seen in the higher-
FACTOR (G-CSF) dose groups and these were terminated early. At 500 mg,
treatment with ELND005 was deemed safe, but did not
Sargramostim and Filgrastim/Neupogen are synthetic
result in any significant difference in cognition or function
preparations of the haemopoietic growth factors, GM-CSF
(NTB, ADCS-ADL) overall. It was however measureable in
and G-CSF. These agents are indicated for bone marrow
CSF, and CSF Aβ42 level decreased significantly (p = 0.009).
stimulation post-transplant in haematological malignan-
There were significant differences in NTB scores when the
cies. These agents are proposed to enhance the innate
mild AD group were analysed separately, and interestingly,
immune system, thereby stimulating microglia to take up
this group also had slightly less emergence of neuropsychi-
and clear Aβ. Second, as soluble Aβ in the peripheral cir-
atric symptoms on the NPI (Neuropsychiatric Inventory). It
culation binds to circulating erythrocytes (via complement
is unclear whether these effects may be due to the drug’s AD
opsonin 3b), they may also increase peripheral clearance by
disease-modifying properties or due to the lowering of myo-
promoting erythropoiesis (Boyd et al., 2010).
inositol. Further Phase II trials have looked at ELND005’s
Observational data suggest that haematology patients
role in treatment of neuropsychiatric symptoms in bipolar
treated with G-CSF + GM-CSF have better cognitive out-
disorder (add-on therapy, terminated early due to financial
comes than those on G-CSF alone (Jim et al., 2012). A pilot
reasons) Down’s syndrome and in moderate–to-severe AD
study of eight mild-to-moderate AD patients treated with
(n = 400, 12 + 18 weeks, still recruiting). Results presented
G-CSF for 5 days showed improvements on a single epi-
from the study in Down’s syndrome (n = 23, ELND005 250
sodic memory task, but no change in CSF Aβ1–42 (Sanchez-
mg/500 mg/placebo for 4 weeks) have been presented. A
Ramos et al., 2012). Sarmograstin/GM-CSF is currently
greater proportion of those on the high-dose regime par-
in several Phase II trials in patients with AD, MCI and
ticipants showed improvements in NPI score than with
Parkinson’s disease (PD).
low-dose or placebo – although the numbers overall were
very small (7/8 compared with 0/4 and 1/3, respectively)
53.3.1.10 Amyloid aggregation inhibitors (Transition Therapeutics Media Release, 2014).

53.3.1.10.1 TRAMIPROSATE 53.3.1.10.3 CURCUMIN
Glycosaminoglycans (GAGS) are components of proteo- Curcumin is found in the spice turmeric and has antioxi-
glycans, which are incorporated into the amyloid plaque. dant, anti-inflammatory and anti-aggregation properties.
Tramiprosate (Alzhemed) is a GAG-mimetic patented vari- Curcumin binds Aβ and reduces amyloid plaque burden
ant of taurine that binds to Aβ and inhibits fibril formation. (Garcia-Alloza et al., 2007) and tau aggregation in mice (Ma
In vitro, tramiprosate binds soluble Aβ, inhibits aggregation et al., 2013). A Phase II trial in AD was negative, with some
and neurotoxicity. In a Phase II trial, tramiprosate reduced gastrointestinal adverse symptoms, and it failed to show any
brain levels of Aβ42 but did not improve cognition scores biomarker changes (Ringman et al., 2012). However, a trial of
compared to placebo (Aisen et al., 2006). The drug pro- curcurmin in combination with exercise (aerobic and non-
ceeded to an 18-month Phase III trial in mild-to-moderate aerobic yoga) is in progress in MCI, with clinical, blood and
AD, which was negative on primary end points. There was fludeoxyglucose-PET (FDG-PET) biomarker end points.
a trend towards slowing of decline on the ADAS-Cog but
no difference in the CDR-SB. There was with significant 53.3.1.10.4 PBT-2
reduction in hippocampal atrophy in participants tak- Aβ binds copper and zinc and some metal-chelating com-
ing tramiprosate, but there are no other biomarker results pounds affect Aβ aggregation, particularly into the most toxic
Drug treatments in development for Alzheimer’s disease 567

oligomeric species. Original studies with clioquinol, a ­copper– plaques in mice by 50% within 72 hours (Cramer et al.,
zinc chelator, showed reduced Aβ deposition in a mouse 2012). Subsequent attempts to replicate these findings have
model of AD (Cherny et al., 2001). A structural analogue, met with varied success although two have corroborated an
PBT-2, has completed a 12-week Phase II trial in 78 subjects effect on soluble Aβ (Veeraraghavalu et al., 2013). Bexarotene
(­placebo, 50 mg and 250 mg daily) (Lannfelt et al., 2008). The acts on the retinoid X receptor to increase ApoE levels and
trial was not powered for cognitive end points but did show a promote ApoE lipidation, both of which assist with ApoE-
significant benefit of the 250 mg dose, compared to placebo, mediated trafficking of Aβ. However, its benefit may vary
on verbal fluency and working memory. There was also a according to APOE genotype – i.e. it appears to correct the
highly significant (p = 0.0060) reduction in CSF Aβ42 with the pathological loss-of-function of APOE ε4 but make little
250 mg dose. A subsequent Phase III trial was completed in difference in mice expressing APOE ε3 (Tai et al., 2014).
42 prodromal AD patients over 12 months (MCI with a posi- A Phase I trial in healthy adults, and a Phase IIa trial
tive 11C-PiB PET scan). Results presented to date indicate that (BEATAD) is currently under way, the latter using change
there was no difference seen in the primary outcome (PiB PET in amyloid (florbetapir PET) as the primary outcome.
binding). The active group did have a reduction in PiB stan- Importantly, it has been known to cause lipid and triglyceride
dardized uptake value ratio (SUVR) (Aβ burden), although abnormalities and pancreatitis, so safety monitoring is critical.
unusually, the SUVR went down in the placebo group as well.
There was a trend to reduction in hippocampal atrophy with 53.3.1.11.3 PHENSERINE AND POSIPHEN
treatment; however, cognitive outcomes were also negative. Phenserine has both cholinesterase-inhibiting and anti-
The drug was safe and well-tolerated; however, it is unclear amyloid effects (reduces APP expression) and preclinical
whether it will have the opportunity to go on to larger trials results were promising (Lahiri et al., 2007). Subsequent
better powered for clinical benefits. Phase II and III trials were negative (albeit trends to cogni-
tive and CSF benefits), and it was withdrawn from devel-
53.3.1.10.5 COLOSTRININ opment (Winblad et al., 2010). Its positive enantiomer,
Colostrinin is a proline-rich polypeptide found in sheep Posiphen, is currently under evaluation. It does not cause
colostrum, which affects amyloid aggregation and may pre- cholinergic effects, potentially enabling higher doses to be
vent apoptosis of neurones. A small open-label study sug- used. In a Phase I study, treatment with Posiphen resulted
gests possible clinical benefit and the peptide components of in significant CSF biomarker improvements (Maccecchini
colostrinin are under investigation (Janusz and Zabłocka, et al., 2012). A 10-day course of Posiphen in MCI patients
2010). Like curcumin, it may be favourably regarded due to led to normalization of CSF tau- and p-tau levels. Posiphen
its ‘nutraceutical’ categorization. also lowers α-synuclein, which also makes it a compound of
interest for PD (Ross et al., 2013).
53.3.1.10.6 CARVEDILOL
Carvedilol is a nonselective antagonist of the α- and 53.3.1.11.4 CHF 5074
β-adrenergic receptors.It is currently indicated for man- A NSAID-derivative initially promoted as a γ-secretase
agement of congestive heart failure, ischaemic heart dis- modulator, CHF 5074 is now understood to have several
ease/angina and hypertension. In TgCRND8 and Tg2576 additional mechanisms of action, activating microglia and
transgenic mouse models of AD, carvedilol attenuates Aβ reducing aberrant cell cycle events. A 12-week Phase II
oligomerization and formation of fibrillar plaques, as well as trial in 96 participants with MCI showed improvements
preserving dendritic spine density and attenuating special in CSF measures of inflammation (CD40l and TNF-
memory deterioration (Wang et al., 2011). Trials in humans α) and trends to cognitive benefits in APOE ε4 carriers.
(mild AD) are under way, with cognitive and CSF-Aβ out- Withdrawals due to diarrhoea occurred in both Phases I
come measures. and II (Ross et al., 2013). The Phase II has been extended
(open label) to 90 weeks.
53.3.1.11 Other anti-amyloid approaches 53.3.1.11.5 PQ912 (PROBIODRUG AG)
PQ912 is a small-molecule antagonist of glutamyl cyclase,
53.3.1.11.1 APOE ε4 GENE MODULATION which generates the aggregation-prone and neurotoxic
The association between APOE ε4 and AD risk has led pyroglutamate Aβ (see Section 53.3.1.7.8). Phase I trials have
to early phase trials of agents that modulate or reduce ε4 suggested that it is well-tolerated, but there are no data on
expression or activity. One group of agents that inhibit efficacy to date (Perez-Garmendia et al., 2013).
APOE ε4 domain interactions looks promising, but large
human trials are awaited (Chen et al., 2011). 53.3.1.11.6 LEVETIRACETAM
Levetiracetam is an anticonvulsant medication that has
53.3.1.11.2 BEXAROTENE (TARGRETIN) shown cognitive benefits in AD patients with seizures
Bexarotene is already FDA approved for use in humans, for (Cumbo et al., 2010). MCI subjects treated with leveti-
cutaneous T-cell lymphoma. It gathered interest in 2012 racetam demonstrated improved memory performance
when a preclinical study reported clearance of amyloid and reduced pathological neuronal hyperactivity in the
568 Dementia

hippocampus (Bakker et al., 2012). Recently, levetirace- AADvac-1 is another active vaccine against pathological
tam has been shown to act on synaptic vesicle protein 2A tau fragments (Axon Neuroscience) that is also in Phase I
(SV2A), which regulates neuronal endocytosis. In APOE development.
ε4 carriers, there is enhanced endocytosis of BACE1
and APP within endosomes, which provides ideal con-
ditions for BACE1 cleavage of APP to Aβ40 and Aβ42. 53.3.2.2 Anti-aggregation approaches for
Modification of SVA2 activity via levetiracetam in trans- tau
genic mice suppresses this APOE ε4-dependent increase
53.3.2.2.1 METHYLTHIOMINIUM CHLORIDE
in Aβ40 and Aβ42 production (Rhinn et al., 2013). Two
(REMBER/TRX0014) AND TRX0237
early phase studies of levetiracetam on cognition and epi-
leptiform activity in AD are currently recruiting, one of There has been a completed trial of the first therapeutic agent
which is targeting early-onset AD (age at symptom onset targeting tau, utilizing a drug derived from the dye used to
<70 years). stain neurofibrillary tangles in neuropathological stud-
ies. The agent primarily inhibits tau aggregation, although
53.3.1.11.7 CT1812 (Cognition Therapeutics) other reported effects include antioxidant and increasing Aβ
In vitro studies suggest that binding of Aβ oligomers to spe- clearance. The 84-week Phase II study in 321 subjects showed
cific receptors at neuronal synapses may be an important significant benefits, compared to placebo, in the cognitive
step leading to loss of neuronal function in AD. CT1812 is (ADAS-Cog) and global (CDR) primary end points but only
a small molecule antagonist at sigma-2/PGRMC1 that dis- for the lower doses (Wischik et al., 2014). The 100 mg (high-
places binding of Aβ oligomers from neurones in vitro, and dose) formulation may have had formulation deficiencies
restores cognition n AD transgenic mice. It is currently in (cross-linking with the gelatine capsule), which have sub-
early phase studies in humans. sequently been addressed. Neuroimaging end points were
also promising, including single-photon emission computed
53.3.1.11.8 CSF DRAINAGE/COGNISHUNT™ tomography (SPECT) in 125 subjects (with no decline in per-
It is possible that continuous CSF drainage will remove Aβ fusion seen with the 60 mg dose, compared to a decline in
from the brain, essentially creating an Aβ gradient. A ran- the placebo group) and FDG-PET in 19 subjects (improved
domized controlled trial (RCT) of 215 subjects utilized a metabolism in the actively treated group). The agent is poorly
sham (occluded) shunt as the control arm. At 9 months, absorbed and diarrhoea occurred in some treated subjects.
there was no significant difference in cognitive or global end TRx0237 is the second-generation formulation of
points between the two groups. There were 12 CNS infec- Rember, which is reported to have improved bioavailabil-
tions. It is unlikely that this approach will be subjected to ity and tolerability. It is currently in Phase III for mild-to-
further trials (Silverberg et al., 2008). moderate AD, mild dementia (all-cause, MMSE > 20) and
behavioural variant FTD.
53.3.2 TAU
53.3.2.3 Targeting tau
By 2050, there will be 600 million people with significant hyperphosphorylation
tau pathology (BRAAK stage II or above) – not all with AD.
Anti-tau therapy could have widespread application for Several phosphorylation inhibitors have been developed, as
people with AD, some FTLDs and MCI. have agents that dephosphorylate tau.

53.3.2.3.1 TIDEGLUSIB
53.3.2.1 Anti-tau immunotherapy
Tideglusib is an inhibitor of glycogen synthase kinase 3
A number of potential targets exist for immunotherapy (GSK-3), which phosphyorylates tau. In mice, it reduced p-tau
against tau, including the microtubule-binding region and neurodegeneration, and improved memory deficits. It
of tau and the pathogenic p-tau (as opposed to non-­ has undergone Phase II human trials for AD and progressive
phosphorylated tau). supranuclear palsy (PSP), a tauopathy. Unfortunately both
applications failed in their primary end points (subanalysis
53.3.2.1.1 ACI-35 at lower dose in mild AD suggested slight benefit) and its
AC Immune recently announced it will begin human tri- development has been discontinued (Lovestone et al., 2014;
als of ACI-35, a liposome vaccine containing synthetic Tolosa et al., 2014).
p-tau fragments within a lipid bilayer. In tau transgenic
mice, vaccination with ACI-35 resulted in generation of IgG 53.3.2.3.2 LITHIUM
antibodies against p-tau, without T-cell actuation, gliosis Lithium inhibits tau phosphorylation via GSK-3 and may
or increasing inflammation. The antibodies produced were have other beneficial roles including improving mitochon-
able to reduce soluble and aggregated tau in vitro (Theunis drial function, increasing levels of neurotrophins (e.g.
et al., 2013). A Phase I trial in humans (mild-to-moderate brain-derived neurotrophic factor [BDNF]) and reduc-
AD) is currently under way. ing apoptosis (Forlenza et al., 2012). Some observational
Drug treatments in development for Alzheimer’s disease 569

studies have also suggested a reduced incidence of demen- 53.3.3 NEURONES AND SYNAPSES
tia in patients with bipolar affective disorder treated with
lithium, compared with other agents (Gerhard et al., 2015). 53.3.3.1 Neurotrophic therapies
Several small open-label or case–control studies in mild-to-
moderate AD have not shown benefit; however, a small RCT 53.3.3.1.1 NERVE GROWTH FACTOR
in amnestic MCI (n = 45) showed lower conversion to AD The first trial of NGF via direct intraventricular injection
compared with placebo, which correlated with reductions into the CSF was halted due to a high proportion of sub-
in CSF p-tau levels (Forlenza et al., 2012). jects experiencing intense axial (back) pain. This may have
been due to the presence of NGF receptors in the spinal
53.3.2.3.3 SELENIUM cord (Eriksdotter Jönhagen et al., 1998). A variation on this
Selenium (as sodium selenate) activates the enzyme PP2A direct neurorestorative approach was examined in a Phase I
phosphatase, which dephosphorylates tau. It is shown to trial in eight patients with mild AD (Tuszynski et al., 2005).
lower levels of p-tau and total tau and improve memory Fibroblasts were extracted from their fingers, genetically
and learning in transgenic mice (Corcoran et al., 2010). It modified to express human NGF, then injected directly
is being tested against placebo and vitamin E in a Phase III back into the cholinergic nucleus basalis of their forebrain.
prevention study in cognitively healthy men (PREADVISE, After a mean follow-up of 22 months in six subjects, no long-
n = 7547); however, no results have been presented as yet. term adverse effects were seen but two (non-anaesthetized)
A small 24-week Phase IIa trial (n = 40) was completed in subjects moved abruptly during the procedure, causing
Australia but sodium selenate at doses of 10 mg and 320 μg subcortical haemorrhage. All subsequent procedures were
showed no effect on CSF biomarkers including both total performed under general anaesthesia. The MMSE and
and p-tau (VEL015- Velacor). ADAS-Cog showed improvements in the rate of decline and
serial FDG-PET scans showed significant (p < 0.05) wide-
53.3.2.3.4 METFORMIN spread increases in metabolism after the procedure. Brain
The insulin-sensitizing antidiabetic drug, metformin is autopsy from one subject showed robust growth responses
also a PP2A phosphatase agonist. It may also reduce AD to the NGF expression.
risk by improving glucose sensitivity and metabolic profile A more recent variation on this theme involved ste-
(see Section 53.2.8); however in vitro results are conflict- reotactic implantation of an encapsulated cell biodelivery
ing, as it may also increase BACE1 transcription. A trial device containing engineered retinal pigment cells into
of metformin in MCI (n = 80) is recently completed (as yet the basal forebrain. In a small Phase I study (n = 6) in AD
unpublished) and another in MCI and AD is in progress patients, the procedure was reported as ‘safe, well-tolerated
(Yarchoan and Arnold, 2014). and feasible’, with cognitive (CDR, instrumental activities
of daily living [IADLs]) and biomarker (CSF Aβ 1–42, hip-
53.3.2.3.5 ABT-957 (ABBVIE) pocampal atrophy) improvements seen in 50% of partici-
The enzyme calpain is involved in a number of pathways to pants (Ferreira et al., 2015).
neurodegeneration, including mediating Aβ-induced neu- CERE-110/AAV2-NGF (Ceregene/Sangiamo) uses a viral
rotoxicity and activating the tau kinase, cyclin-dependent vector that expresses human NGF, which is injected into the
kinase 5 (CDK5). It may also be involved in pathologi- nucleus basalis of Meynert. In Phase I (mild-to-moderate
cal cleavage pathways of the α-synuclein protein in Lewy AD, n = 6), it was well-tolerated and cognitive improve-
body disorders. Inhibition of calpain in tau transgenic mice ments were reported, as well as increases in brain metabo-
reduced hyperphosphorylation of tau and prevented neuro- lism using FDG-PET. The technique is currently in Phase II.
degeneration (Rao et al., 2014). ABT-957, a calpain antago-
nist, began Phase I trials in 2014. 53.3.3.1.2 XALIPRODEN
Xaliproden is a neurotrophic factor enhancer and 5-HT1A
agonist that demonstrated neuroprotective and neuro-
53.3.2.4 Enhancing microtubular stability restorative effects in preclinical studies. It proceeded to
large Phase III clinical trials (>1300 patients each) in sub-
53.3.2.4.1 EPOTHILONE D
jects with AD. Results were negative, although a smaller
Epothilone D (BMS-241027, Bristol-Myers Squibb) clears substudy (n = 329) showed less hippocampal atrophy in the
tau pathology and improves cognition and behaviour in active group (Porzner et al., 2009).
transgenic tauopathy mice (Barten et al., 2012), but a Phase I
study was terminated early due to lack of target engagement. 53.3.3.1.3 CEREBROLYSIN
A peptide mixture extracted from pig brains, Cerebrolysin
53.3.2.4.2 TPI 287 has neurotrophic activity (increases pro-NGF) and
TPI 287 (Cortice Biosciences) is a derivative from the taxane reduced Aβ deposition in animal models. A recent meta-
family of cancer drugs. It is in Phase I for AD, and other analysis of six RCTs in mild- and moderate-AD subjects
tauopathies including corticobasal degeneration (CBD) (n = 784) compared Cerebrolysin (30 mL intravenous
and PSP. infusion daily for 4 weeks) with measures at 4 weeks and
570 Dementia

6 months. By standardizing outcome scales from the dif- 53.3.3.2.1 STEM CELL THERAPY IN OTHER CENTRAL
ferent studies, they found benefit for cerebrolysin in global NERVOUS SYSTEM DISEASES
clinical change at week 4 (odds ratio [OR] 3.32, 95% CI Early human studies in animal models and humans with
1.20–9.21) and 6 months (OR 4.98, 95% CI 1.37–18.13). PD grafted (postmitotic) foetal dopaminergic neurones into
However, a significant cognitive function benefit was seen the striatum. While this achieved clinical improvement in
at week 4 only (standardized mean difference −0.40, 95% some subjects, lack of availability of foetal tissue has limited
CI −0.66to −0.13) but not at 6 months (not reported). AEs its utility (Antigoni Ekonomou, 2010).
profile was similar to placebo, and it has been studied Under strict guidelines, ESCs for research have been
both as monotherapy and in combination with ChEI. A sourced from in vitro fertilization programmes. A trial of
Cochrane review of RCTs in vascular dementia (six stud- ESCs (oligodendrocyte progenitor cells [OPCs], OPC1) to
ies, n = 597) also was supportive, although also benefits treat neurologically complete thoracic spinal cord injury
over placebo were of modest magnitude only (one-point was commenced in 2010, following promising preclini-
difference in MMSE, four-point difference in ADAS-Cog). cal findings in rats. Only 5 of the planned 10 participants
It is commercially available for treatment of AD in several were recruited, and the study sponsor, Genen, discon-
countries and is being trialled in acute traumatic brain tinued this field of development. Their stem cell therapy
injury and stroke (Alvarez et al., 2006, 2011; Chen et al., portfolio has been purchased and is being expanded
2013; Gauthier et al., 2014). through Asterias Biotherapeutics. Although no results
have been published, they report successful delivery of
53.3.3.1.4 T-817MA
OPC1 cells, with no serious AEs, and potential radio-
T-817MA is an oral neurotrophic agent that promote neu- logical benefits, although no measurable neurological
rite growth and protects synapses against Aβ-induced neu- improvements were seen. A subsequent Phase I/IIa esca-
rotoxicity (Takamura et al., 2014). A large Phase IIa trial lating dose study in cervical cord injury is to recruit in
(n = 373) showed trends to improved cognition and func- 2015 (Asterias Biotherapeutics, 2015).
tion, which will be tested in another trial as add-on therapy For multiple sclerosis, autologous bone marrow-derived
in AD (Schneider et al., 2013). haemopoietic stem cells have also been used as an add-on
to immunosuppression to mitigate immune-mediated neu-
53.3.3.2 Stem cell therapies ropathology. Both radiological and clinical benefits are
reported, although larger RCTs are required (Burt et al.,
Stem cells are self-renewing cells that can replicate and 2015).
differentiate to produce other diverse cell types. Stem cell
therapy provides neural progenitor cells, with the aim of
replacing damaged nerves (neurogenesis). Its proposed 53.3.3.2.2 STEM CELL THERAPY IN ALZHEIMER’S
applications in the CNS include treatment of nerve injury DISEASE
(e.g. spinal cord injury) and neurodegenerative disorders There is evidence for benefit of NSC and MSC therapies
(e.g. PD, AD). For comprehensive reviews, see Antigoni in animal models of AD, including reduction of expres-
Ekonomou (2010), Fan et al. (2014) and Li et al. (2015). sion of APP, BACE1 and Aβ; attenuation of BACE1 and
Stem cells may be derived from several sources, includ- γ-secretase activity; increased expression of BDNF and
ing neural stem cells (NSCs) from mature or foetal brain neprilysin (enhancing degradation of Aβ); reduction in
tissue, mesenchymal stem cells (MSCs) from umbilical pro-inflammatory cytokines and inflammation; improve-
cord blood or bone marrow and cells from the developing ment in cholinergic transmission and reversal of mem-
embryo (embryonic stem cells [ESCs]). Somatic cells may ory deficits (Yun et al., 2013; Li et al., 2015). However,
also be treated to reprogramme their activity to produce the unfriendly microenvironment may compromise the
pluripotent stem cells. With stem cell therapy, there are survival and ‘successful’ differentiation of the NSCs into
inherent risks of immune rejection (with donor or non-self neurones. In AD, presence of APP and Aβ fragments may
cells) and development of tumours (with pluripotent cell result in reduced NSC proliferation, reduced neurogen-
lines) (Antigoni Ekonomou, 2010; Li et al., 2015). esis, and preferential production of astrocytes. Provision
Endogenous NSCs are also present in adult brains, and of additional neurotrophins, e.g. BDNF, glial-derived
ongoing neurogenesis occurs in select regions, including neurotrophic factor (GDNF) and vascular endothelial
the hippocampus. However, these cells are few in number growth factor (VEGF) may counter these negative effects
and this activity is reduced in neurodegenerative disease. (Li et al., 2015).
Treatments or factors shown to restore or promote this There are no current trials of NSCs or ESCs in humans
neurogenesis in vitro and in animal models include allo- with AD, although a Phase I trial of intravenous infusion
pregnanolone (an endogenous neurosteroid), and intrinsic of human umbilical cord blood-derived MSCs in 30 AD
factors such as environment and physical activity (Li et al., participants has commenced in China. Outcomes are safety
2015). (AEs) and cognitive (ADAS-Cog and Clinician’s Interview
Research with foetal and ESCs poses great ethical and Based Impression Of Change [CIBIC]). Results are expected
supply challenges, and use of this tissue is tightly regulated. by end of 2015.
Drug treatments in development for Alzheimer’s disease 571

53.3.4 INSULIN-SENSITIZING amino acid metabolism, improving insulin signalling


TREATMENTS AND PPAR-γ and pancreatic β-cell function. It also influences metabo-
AGONISTS lism to favour ‘brown fat’ (calorie burning) and decrease
‘white fat’ (calorie storing). A small Phase IIa study in
Several therapeutic approaches to improving insulin and mild-to-moderate AD (n = 29), MCDC-0160 maintained
IGF signalling in AD are currently in clinical trials. or increased glucose metabolism using FDG-PET in ante-
rior and posterior cingulate, parietal lateral temporal
53.3.4.1 PPAR-γ agonists and medial temporal cortices (regions affected by AD)
(Shah et al., 2014). It is also being assessed for benefit as
The thiazolidinedione (TZD) group of PPAR-γ receptor ago- an alternative insulin-sensitizer in diabetes, and in PD,
nists (glitazones), increase tissue sensitivity to glucose, reg- where it may have neuroprotective effects on dopaminer-
ulate lipid metabolism and increase mitochondrial activity, gic neurones.
as well as reducing inflammatory responses by inhibiting
nuclear factor-κB and downregulating BACE1 promotion 53.3.4.2 Glucagon-like peptide-1 (GLP-1)
(Gold et al., 2010). At low doses, they may also effect amy- analogues and GLP-1 receptor
loid clearance by increasing LRP1 expression (Moon et al.,
agonists
2012).
Incretin hormones are released by the gut in response to oral
53.3.4.1.1 ROSIGLITAZONE
ingestion of glucose. They act to facilitate insulin signalling,
Rosiglitazone is a PPAR-γ agonist that increases peripheral but also have neurotrophic and anti-inflammatory effects
insulin sensitivity and is approved for clinical use in type (McClean and Hölscher, 2014). A number of GLP-1 ago-
2 (T2DM). In transgenic mice, it decreases Aβ42 plaque nists or analogues are currently approved for use in T2DM,
burden, mitigates Aβ-associated inflammation, synaptic including liraglutide (Victoza), exendin-4 (Exenatide/
dysfunction and cognition. In Phase II (n = 323), cognitive Byetta) and lixisenatide (Lyxumia).
benefits were seen in APOE ε4-negative individuals, but not
in APOE ε4 carriers. Subsequently a large Phase III study 53.3.4.2.1 LIRAGLUTIDE
(n = 693) stratified by APOE ε4 status was negative (Gold Liraglutide is a GLP-1 receptor agonist currently approved
et al., 2010). Side effects included peripheral oedema and for T2DM. In APP/PS1 mice, liraglutide reduced Aβ plaque
cardiac failure. load and inflammation by one-third, while increasing IDE
levels and improving object recognition and memory per-
53.3.4.1.2 PIOGLITAZONE formance (McClean and Hölscher, 2014). A Phase IIb study
Pioglitazone is another PPAR-γ agonist that is approved for in mild AD (Dubois criteria, MMSE > 22, CDR global score
use in T2DM that has been trialled in AD. It has been tested 0.5–1) is currently recruiting, to test liraglutide (1.8 mg sub-
in MCI and AD with small Phase I/II trials and has shown cutaneous injection daily) against placebo. It will run for
positive results in diabetics, but not in non-diabetics. In one 12 months, with primary outcome measure change in brain
open-label study of AD patients with T2DM, pioglitazone metabolism using FDG-PET. Secondary measures include
15–30 mg improved regional cerebral blood flow, stabilised change in cognitive ADAS exec, MRI volumes, microglial
plasma Aβ40/42 ratio and improved cognition (Sato et al., activation and CSF markers.
2011). Pioglitazone is currently being tested at low doses
(0.8 mg daily about 2% of the therapeutic dose in diabetes) in 53.3.4.2.2 EXENATIDE
a large multicentre study of cognitively normal individuals Exendin-4 (Byetta) is a GLP-1 analogue, derived from the
aimed at preventing progression to MCI (TOMMORROW venom of the Gila monster, a shy Central American lizard
Study). Aiming to enrol 5800 participants, this study will that can survive long periods of fasting.
assign individuals to intervention or placebo according to In AD transgenic (3xTg-AD) mice, exendin-4 signifi-
their genetic risk for AD using APOE and TOMM40 geno- cantly reduced brain Aβ levels (Li et al., 2010) and in rats
types. The primary end point will be the time to conversion with experimental hyperglycaemia and oxidative stress
to MCI due to AD. Exendin-4 downregulates GSK-3β, reducing tau hyperphos-
This trial is administering pioglitazone at doses much phorylation (Chen et al., 2012) and improving cognition.
lower than that usually prescribed for diabetes, suggest- A Phase II trial of Exendin-4 (twice daily subcutaneous
ing that it aims to target the LRP1 hypothesis (Moon et al., injections) in AD (MMSE > 20, n = 230) is under way. This
2012) rather than insulin resistance. trial uses CSF Aβ for participant screening and as a second-
ary outcome measure.
53.3.4.1.3 MSDC-0160
MCDC-0160 (Metabolic Solutions) is a novel oral insulin- 53.3.4.2.3 INTRANASAL INSULIN (DETEMIR,
sensitizer that acts at the mitochondrial target of thiazoli- LEVEMIR, HUMULIN, NOVOLIN)
dinedione (mTOT) receptor in the inner mitochondrial Improved brain insulin signalling correlates with mem-
membrane. mTOT coordinates carbohydrate, lipid and ory function in healthy adults, AD and MCI (Freiherr
572 Dementia

et al., 2013). The challenge has been to deliver insulin (donepezil) was terminated prematurely following a futility
effectively to the brain, without causing peripheral effects. analysis.
Several small trials of intranasal delivery of insulin (vari- Both MK0249 (Merck) and PF-03654746 (Pfizer) have
ous formulations) have been conducted (Benedict et al., been studied in AD, schizophrenia and attention-deficit
2004; Reger et al., 2008; Craft et al., 2012). One Phase I hyperactivity disorder (ADHD), all with negative findings
clinical trial in MCI (n = 64) or early AD (n = 40) par- on cognition. It is unclear whether they will be developed
ticipants administered 20 or 40 IU of intranasal insulin further.
over 4 months. The treatments were well-tolerated and
they showed improvements in primary efficacy measures 53.3.5.1.1 5-HT6 RECEPTOR ANTAGONISM
(ADAS-Cog and ADCS-ADL) over placebo. Subsets also
5-HT6 receptor is localized almost exclusively in the CNS,
had CSF (n = 23) and FDG PET (n = 40). FDG hypome-
particularly in the cerebral cortex and hippocampus.
tabolism was attenuated in the treatment groups, and
Antagonism enhances cholinergic and glutamatergic neu-
although CSF = Aβ did not differ at group level, change in
rotransmission and may be complementary as adjunct ther-
CSF = Aβ42 and Aβ42/tau ratio correlated with change in
apies to ChEIs (Upton et al., 2008).
cognitive performance (Craft et al., 2012). A Phase II/III
Two agents, RVT-101 (formerly SB-742457) (Axovant,
study of intranasal insulin in AD and aMCI is currently
formerly GSK) and LU AE58054/idalopirdine (Lundbeck)
recruiting (n = 240, the ‘Study of Nasal Insulin in the Fight
both led to improvements in cognition and global function,
against Forgetfulness, SNIFF’).
with stronger findings in moderate-to-severe AD, and when
used as add-on therapy with donepezil. Lundbeck is test-
53.3.5 NEUROTRANSMITTERS AND ing idalopirdine in three large Phase III trials (n ~ 2500 in
RECEPTORS total) in AD as adjunct to ChEIs. GSK tested SB-742457 to
Phase II. Under Axovant, SB-742457 has been renamed and
Most therapies directed at neurotransmitters and their will be tested to Phase III in mild-to-moderate AD, again as
receptors are symptomatic rather than disease modify- add-on to donepezil. Trials in Lewy body dementia are also
ing, although the distinction between these two classes of planned.
effects is less clear than originally thought. Modifying syn- A Pfizer agent, PF-05212377, is currently being tested
aptic activity may have neuroprotective effects, although in Phase II as add-on therapy to donepezil in patients with
it has been difficult to demonstrate this with the ChEIs. mild-to-moderate AD with neuropsychiatric symptoms.
Several new neurotransmitter approaches are being tri- There are also at least four other 5-TH6 antagonists in
alled and some may ultimately improve symptoms in Phase I (Schneider, 2014).
the short term while also having longer-term effects on
neurodegeneration. α-Amino-3-hydroxy-5-methyl-4-
53.3.5.2 
isoxazolepropionic acid (AMPA)
53.3.5.1 Histamine 3 (H3) receptor receptor modulation
antagonists
Reduction in number of the glutamatergic AMPA recep-
The H3 receptor is preserved even in severe AD, and tors is an early finding in AD, accompanied by loss of den-
drugs that antagonize it increase levels of acetylcholine, dritic spines and synapses. Modulation of AMPA receptor
noradrenaline and dopamine in the cingulate cortex and function is suggested to improve alertness and concentra-
hippocampus. Several compounds are being evaluated for tion, facilitate long-term potentiation (required for memory
cognitive benefits through H3 receptor antagonism, which encoding) and they have been postulated as beneficial in
improves cognition, attention and alertness in animal AD, ADHD and sleep disorders.
models (Vohora and Bhowmik, 2012). GSK239512IT has Eli Lilly’s LY451395 was tested to Phase II after preclini-
undergone a Phase II study (mild-to-moderate AD, MMSE cal studies demonstrated reversal of age-associated memory
16–24, 16 weeks, n = 196), which demonstrated modest but deficits in mice; however in humans with mild-to-moderate
significant improvement in episodic memory performance AD, there was no benefit seen in cognition or neuropsychi-
(but not executive function or ADAS-Cog). Side effects atric symptoms.
included headache, dizziness and sleep disturbance (Grove Cortex Pharmaceuticals have developed many
et al., 2014). ‘AMPAkines’, including several that have been discontinued –
A Phase II trial of the Sanofi compound SAR110894 CX-516/Ampalex (lack of efficacy), CX-691/Farampator
(mild-to-moderate AD, MMSE 15–25, n = 290, 24 weeks) (inconsistent results). CX717 was rejected by FDA for AD
was negative in all primary and secondary end points, due to toxicity concerns but was subsequently trialled in
except for a significant improvement in functional score Europe for respiratory depression. CX1632, acquired by
(ADCS-ADL) compared with placebo. Servier S47445, is being tested as an add-on to donepezil in
A Phase II study of the Abbot H3 antagonist, ABT-288, patients with mild-to-moderate AD with depressive symp-
in mild-to-moderate AD versus placebo and active placebo toms (Aisen et al., 2012).
Drug treatments in development for Alzheimer’s disease 573

53.3.5.3 Acetylcholinesterase inhibition dose, head-to-head with donepezil and placebo (n = 274,


mild-to-moderate AD, MMSE 10–24, 12 weeks). It was well-
53.3.5.3.1 HUPERZINE-A tolerated, and there were trends to cognitive benefit with
Huperzine-A is derived from the club moss, Huperzia high-dose ABT-126 that were similar in magnitude to the
serrata. A traditional Chinese remedy for fever and donepezil group (ADAS-Cog mean difference [standard
inflammation, Huperzine-A, is a potent inhibitor of acetyl- error]: −1.2 [0.9], p = 0.09 versus −1.4 [0.9], p = 0.06) (Gault
cholinesterase. In animal models, it also reduces soluble and et al., 2013).
insoluble β-amyloid, reduces tau hyperphosphorylation and
53.3.5.4.3 TC-1734/AZD3480
decreases levels of iron (implicated in free radical formation
and oxidative damage) (Huang et al., 2014) Several trials One agent, TC-1734/AZD3480, a selective agonist of the
in China in the 1990s suggested cognitive benefits in AD, α4β2 and α2β2 receptors progressed to Phase II trials.
and different formulations have since been tested to Phase A large Phase IIb study was negative in its primary out-
II, with varying results (Yang et al., 2013). Huperzine-A is come, but showed modest effects in moderate (but not
approved for treatment of AD in China and is available over mild) AD, comparable with donepezil (Frölich et al., 2011).
the counter as a neutraceutical in the United States. Given Subsequent head-to-head study with donepezil did not
that its mechanism of action is similar to that of the FDA- evidence superiority, however, and development has been
approved ChEIs, it is not recommended for use in combina- discontinued.
tion with any of these agents.
53.3.5.4.4 GLN-1062 (MEMOGAIN)
53.3.5.3.2 LADOSTIGIL GLN-1062 is a pro-drug of the ChEI, galantamine, with
Ladostigil is an inhibitor of acetylcholinesterase that improved bioavailability in the brain (>15 fold), without the
also inhibits monoamine oxidase A and B (MAO-A and same dose-limiting side effects seen with the others within
-B) and has neuroprotective and antioxidant properties this class (Maelicke et al., 2010). A Phase Ia in young and
(Weinreb et al., 2012). It is currently in a Phase IIb trial in elderly healthy subjects compared GLN-1062 to donepezil
MCI (n = 200). Interim results announced in a media release 10 mg and galantamine 16 mg. Nausea was reported with
in 2014 asserted no safety concerns, with positive efficacy galantamine at 16 mg, but GLN-1062 was well-tolerated
trends. Final results will be reported in 2016. up to the highest dose (44 mg). Also, GLN-1062 improved
vigilance and short-term memory performance in the both
53.3.5.4 Nicotinic receptor agonists and young and elderly healthy volunteers, compared with done-
modulators pezil and galantamine, which had no effect (Neurodyn
media release, 2014).
Galantamine, a ChEI used in AD, also has nicotinic recep-
tor activity. Several nicotinic agents (predominantly act-
ing on the α7-nicotinic receptor) have been developed for 53.3.5.5 Muscarinic receptor agonists
AD. These agents may have other neurotransmitter benefits
beyond increasing levels of acetylcholine. 53.3.5.5.1 AF267B
M1 muscarinic agonists such as AF267B increase non-­
53.3.5.4.1 ENCENICLINE/EVP-6124 amyloidogenic APP processing (promote production of
Encenicline/EVP-6124 is an α7 receptor potentiator that is sAPPα over Aβ). In a mouse model AF267B attenuated both
believed to increase the receptor’s responsiveness to acetyl- plaque and tangle formation (Salloway et al., 2008). It went
choline. It was well-tolerated in Phase I and II, and cognitive to Phase I in AD and in Sjögren’s syndrome (dry mouth) but
and functional benefits were seen at higher doses (ADAS- no further trials have been registered.
Cog and CDR-SB, respectively). It proceeded to Phase III
trials (COGNITIV AD) in mild-to-moderate AD (with or 53.3.5.5.2 MK-7622
without ChEIs), and schizophrenia (Deardorff et al., 2015). Merck has a muscarinic agonist, MK-7622, in Phase II cur-
These studies were placed on hold in 2015 due to severe gas- rently, as an adjunct to donepezil in mild-to-moderate AD.
trointestinal side effects in AD participants. Interestingly, Multiple dose regimes will be assessed, with change in
the two Phase III studies in participants with schizophre- ADAS-Cog11 the primary clinical end points.
nia continued to completion, with no severe gastrointesti- Earlier trials with direct muscarinic receptor agonists
nal effects reported. Unfortunately, however, neither study were abandoned largely due to unacceptable adverse effects
showed positive results on the co-primary endpoints of cog- (these drugs included milameline, xanomeline and talsa-
nition or function. clidine). There are several muscarinic receptor subtypes
and it is possible that an effective therapy with an accept-
53.3.5.4.2 ABT-126 able adverse effects profile will emerge. The Anavex agent,
ABT-126 (ABBVie) is another α7 nicotinic receptor ­agonist, ANAVEX2-73 has both M1 agonist and sigma-1 agonist
which has been tested in a Phase II study, high and low properties (see Section 53.3.7.2).
574 Dementia

53.3.6 ANTI-INFLAMMATORY THERAPIES 53.3.7.2 ANAVEX2-73 (Anavex Life Sciences


Corp.)
53.3.6.1 Non-steroidal anti-inflammatory
drugs ANAVEX2-73 (tetrahydro-N, N-dimethyl-2, 2-diphenyl-
3-furanmethanamine) is a mixed muscarinic receptor
Alzheimer recognized the presence of activated microg- ligand and a sigma-1 receptor (s1R) agonist.
lia cells around plaques and tangles and many other M1-4 agonists preferentially activate the neuroprotec-
markers of inflammation have been documented. Large tive/non-amyloidogenic pathway (see Section 53.3.5.5).
epidemiological studies have demonstrated a lower The σ1 protein is a molecular chaperone, located on the
prevalence of AD in long-term users of NSAIDs and endoplasmic reticulum (ER) at sites in communication
this led to a large number of therapeutic trials of these with mitochondria. It regulates calcium exchange between
agents in AD, from 1993 to 2004. All were negative and ER and mitochondria and is important for regulation of
any further development of existing agents (including metabolism, production of reactive oxygen species and
steroids, NSAIDs and hydroxychloroquine) is unlikely cell apoptosis. Promoting σ1 activity has beneficial effects
(Woodward, 2007). on Ca 2+ homeostasis and signal transduction and protects
mitochondrial function (Lahmy et al., 2013, 2015).
53.3.6.1.1 ETANERCEPT In preclinical trials, ANAVEX2-73 was shown to be
Involvement of the pro-inflammatory cytokine tumour protective against oxidative stress and mitochondrial
necrosis factor-α (TNF-α) in the pathogenesis of AD has dysfunction, reduced tau hyperphosphorylation and led
long been suspected. Etanercept is a biological antagonist to memory and neuroprotective benefits (Lahmy et al.,
of TNF-α and when delivered by perispinal administration 2015). A Phase I study in healthy controls has been com-
it improved cognitive function in a small number of cases pleted, and a Phase II two-phase crossover study of oral
from a single centre (Tobinick and Gross, 2008). It is pos- and intravenous ANAVEX2-73 in mild-to-moderate AD is
sible that perispinal delivery may not be necessary; a sub- recruiting.
sequent Phase II trial of subcutaneous etanercept (50 mg
weekly) versus placebo (n = 41) also showed reduced rate of 53.3.8 ANTIOXIDANTS
decline in cognition and ADLs at 6 months. Further trials
are suggested (Holmes et al., 2014). Α-TOCOPHEROL (VITAMIN E)
53.3.8.1 
53.3.6.1.2 MASITINIB Following encouraging results from a trial of the antioxi-
Masitinib is a selective tyrosine kinase inhibitor that influ- dant Vitamin E in patients with moderate-to-severe AD
ences the differentiation and activation of mast cells, which (Sano et al., 1997), there was a negative trial of Vitamin E
are involved inflammatory processes throughout the body in subjects with MCI – the therapy did not delay progres-
and within the CNS. It has also been applied in inflamma- sion to AD over 3 years (Petersen et al., 2005). Vitamin E
tory disorders such as asthma and rheumatoid arthritis. In has been retested alone and in combination with meman-
a small Phase II study (n = 34) in mild-t-moderate AD as tine in mild-to-moderate AD. Improvements in ADCS-
adjunct therapy, there were significant differences seen in ADL (function) and caregiver time were noted, and a trend
ADAS-Cog, ADCS-ADL and MMSE compared with pla- to improvement in ADAS-Cog but not MMSE or NPI.
cebo. However, AEs were more common (65% versus 38% Intriguingly, memantine was no better than placebo in this
in placebo) with oedema, rash and gastrointestinal symp- group, and the combination of Vitamin E and memantine
toms most frequent (Piette et al., 2011). A Phase III trial has did worse still. At doses to 2000 IU, the rates of AEs and
commenced. discontinuation was similar across all groups (Dysken et al.,
2014). Concern about increased mortality risk of Vitamin E
at high doses has limited study of this antioxidant as an AD
53.3.7 MITOCHONDRIAL AGENTS therapy (Miller et al., 2005).
53.3.7.1 Latrepirdine (dimebon)
53.3.8.2 RESVERATROL
Latrepirdine was developed as an antihistamine (5HT6
antagonist) but also is a weak ChEI, a weak NMDA antag- Resveratrol is a polyphenolic compound present in seeds
onist, and improves mitochondrial function. A Phase and skin of many plants, including grapes and berries. It is
II trial (n = 193) in AD was overwhelmingly positive for also found in dark chocolate, tea and red wine.
the primary and all secondary end points at 6 months, As a potent antioxidant and a direct free radical scav-
and also after an additional 6-month blinded extension enger, resveratrol may also have other neuroprotective
(Doody et al., 2008). However, the drug failed to meet any effects, including reducing iron accumulation and decreas-
of its end points in two large Phase III trials and has been ing production of Aβ (Rege et al., 2014). In cognitively nor-
discontinued. mal obese humans, it induces metabolic changes similar to
Drug treatments in development for Alzheimer’s disease 575

the effect of caloric restriction (activates ­sirtuin-1, lowers elderly subjects with ‘age-associated memory impairment’,
blood pressure [BP], improves glucose tolerance, reduces it was well-tolerated and led to improvements in long-term,
inflammatory markers) and improves brain glucose but not short-term memory. No results have been published;
metabolism, functional connectivity and cognitive mea- however, it is listed as still being in development according
sures. In AD (Phase II, MMSE 14–26, n = 119, 12 months), to the Dart NeuroScience (2014) website.
resveratrol altered CSF Aβ40 and reduced brain vol- The α-secretase activator Etazolate/EHT-0202, described
umes, but had no effect on CSF Aβ42, p-tau, hippocam- in (Section 53.3.1.3.1), also inhibits PDE-4.
pal volume or cognition. A large six-arm Phase IV trial of
dietary modification, which includes resveratrol in over- 53.3.9.2 Statins (3-hydroxy-3-
weight MCI participants (body mass index [BMI] 25-35)
is ongoing. methylglutaryl-coenzyme A [HMG-
CoA] reductase inhibitors)
53.3.9 OTHER AGENTS A significantly lower prevalence of AD in those using statins
(HMG-CoA reductase inhibitors) has been found in most
53.3.9.1 Phosphodiesterase inhibition epidemiological studies, irrespective of cholesterol levels.
Statins affect APP processing by γ-secretase so any efficacy
The cyclic nucleotide PDEs incorporate 11 families of
in AD could be due to decreased Aβ production. Despite
enzymes (over 100 variants) that break down cyclic adenos-
epidemiological findings, a Cochrane review of statin ther-
ine monophosphate (cAMP) and cyclic guanosine mono-
apy for dementia (three trials included 748 participants) did
phosphate (cGMP), two second messengers, that regulate
not identify any cognitive benefits in treatment of possible
diverse cellular processes by promoting gene transcription.
or probable AD (McGuinness et al., 2014).
The expression of different PDE isoforms varies between
tissues, including with the CNS, and as such, several rep- 53.3.9.2.1 ATORVASTATIN
resent potential targets in AD. Dependent on isoform and
A small study of atorvastatin in 63 individuals showed some
site, either symptomatic or disease-modifying effects are
promising results (Sparks et al., 2005) but a large Phase III
suggested (García-Osta et al., 2012).
trial using the 80 mg dose for those with AD (LeaDe trial)
53.3.9.1.1 PF-04447943 (Jones et al., 2008) was negative.
The Pfizer drug PF-04447943 is a high affinity inhibitor 53.3.9.2.2 SIMVASTATIN
of PDE-9A and this action should elevate cGMP, in turn
Simvastatin is one of the most highly lipophilic of the HMG-
improving synaptic function and stabilizing vulnerable
CoA reductase inhibitors and so can pass the BBB. Early stud-
synapses. Although preclinical data were promising, trials
ies of simvastatin in AD suggested some disease-modifying
to Phase II have not shown any difference in cognition or
effects, with alterations in CSF biomarkers seen (Simons
function (Heckman et al., 2014).
et al., 2002; Sjögren et al., 2003). However, a subsequent large
53.3.9.1.2 CILOSTAZOL trial in mild-to-moderate AD had no benefit on symptom
progression, despite lowering cholesterol (Sano et al., 2011).
The PDE-3 inhibitor Cilostazol, is a vasodilator currently
Statins may still be of benefit when applied in cognitively
available for treatment of peripheral vascular disease.
normal individuals. A small Phase IV study (normal con-
PDE-3 is also upregulated in cerebral vessels in AD and
trols, n = 23) compared 12 weeks’ treatment with simvastatin
CAA. In mouse models Cilostazol improved Aβ clearance
against pravastatin, which is less lipophilic and does not
by promoting arterial pulsation-driven perivascular drain-
cross the BBB). Simvastatin lowered CSF p-tau, but did not
age (Maki et al., 2014). It has shown some efficacy in early-
alter Aβ isoforms (Riekse et al., 2006). A 4-month trial study
phase trials, but data are mixed (Heckman et al., 2014).
of simvastatin versus placebo (n = 57) for 4 months showed
53.3.9.1.3 MK-0952 no difference in CSF biomarkers, but did not improve verbal
fluency and working memory in treated subjects (Carlsson
PDE-4 has been a target of interest in AD, as inhibiting PDE-4
et al., 2008). Further trials in cognitively normal individu-
leads to increased availability of cAMP, important in mediat-
als and prodromal AD are in progress. Statins may yet also
ing step in the consolidation of long-term memory. However,
have a role as add-on therapy, but only positive trial results
many PDE-4 inhibitors to date have caused dose-limiting
could support this usage.
nausea and vomiting. Merck has developed MK-0952 a PDE-4
inhibitor, which was well-tolerated in animal models and did
improve memory. Phase II trials were initiated in 2006, but no 53.3.9.3 CONTINUOUS POSITIVE AIRWAYS
results have been published (Heckman et al., 2014). PRESSURE (CPAP)
Observational studies suggest links between sleep-­
53.3.9.1.4 HT-0712 disordered breathing and dementia risk, as well as with
HT-0712 is another PDE-4 inhibitor that has positive effects cardiovascular and cerebrovascular disease. Obstructive
on memory in preclinical studies. In a Phase IIa study in sleep apnoea (OSA) causes chronic intermittent hypoxia
576 Dementia

and increase oxidative stress, which are each associated Elevated levels of the glucocorticoid hormone cortisol
with increased β-amyloid and p-tau in animal models. are associated with cognitive dysfunction and may be neu-
Untreated OSA and excessive daytime sleepiness are asso- rotoxic. Xanamem (UE2343) inhibits 11β-hydroxysteroid
ciated with increased prevalence of cognitive impairment dehydrogenase type 1 (11β-HSD1), which converts inactive
(particularly attention and executive function) and risk of cortisone into the active cortisol. Preclinical trials suggest
future cognitive decline. Further, in one study of cognitively benefits on cognition and Aβ plaque deposition, and the
normal elderly, severity of sleep-disordered breathing cor- drug is now in Phase II (XanADu) in mild AD (MMSE
related with CSF Aβ and tau, although the directions and 20-26, CDR 0.5-1.0). Co-primary endpoints include the
significance varied by APOE genotype (Osorio et al., 2014). ADCOMS cognitive composite score and the ADAS-Cog.
A trial of CPAP in AD patients with OSA improved mem-
ory, executive function and processing speed (Ancoli-Israel
53.3.9.4 Homocysteine-lowering therapies
et al., 2008), although there were no biomarker outcome
measures. Two further trials in AD and one in MCI have Homocysteine levels are higher in AD patients than in age-
been commenced. As well as attempting to confirm cog- matched controls (Seshadri et al., 2002) and this may relate
nitive benefits of CPAP in AD, they will collect biomarker to reduced DNA repair or cerebrovascular effects of homo-
measures (neuroimaging and CSF) to look for evidence of cysteine, but trials of the homocysteine-lowering agents,
disease modification. folate and vitamins B12 and B6, in AD have been negative
(Kwok et al., 2011).
53.3.9.4 NILVADIPINE
Nilvadipine is a dihydropyridine calcium channel inhibitor 53.3.9.5 Neutraceuticals and medical foods
that is in clinical use as an antihypertensive medication in
Europe and Japan. In an open-label study of 55 AD partici- There are several ‘neutralceuticals’ or medical foods cur-
pants, nilvadipine had benefits on cognition over 6 weeks rently in development or commercially available for AD.
treatment (Kennelly et al., 2011). In a small study of hyper- According to FDA, medical foods are ‘… formulated to
tensive MCI participants, nilvadipine warded off cognitive be consumed … under the supervision of a physician …
decline and improved cerebral blood flow (Hanyu et al., for the specific dietary management of a disease for which
2007). A large multicentre trial in mild-to-moderate AD distinctive nutritional requirements, based on recognized
(n = 500) is ongoing. scientific principles, are established …’ Notably, medi-
cal foods are not required to undergo pre-market FDA
53.3.9.5 REPETITIVE TRANSCRANIAL MAGNETIC approval (FDA, 2016).
STIMULATION (rTMS)
Transcranial magnetic stimulation is a technique, which 53.3.9.5.1 SOUVENAID
can non-invasively measure cortical activity, with altera- Souvenaid, containing Fortasyn Connect, is a medical food
tions in AD seen in accordance with underlying neu- rich in precursors and cofactors for formation of neuronal
rodegeneration. Preliminary studies also suggest that membranes, including choline, phospholipids, docosahexae-
administered repetitively, rTMS can also influence corti- noic acid (DHA), folic acid and selenium. A multicentre
cal excitability and neuroplasticity, although larger tri- ‘proof-of-concept trial’ (n = 225, mild AD, 12 weeks) showed
als are required to confirm these results (Nardone et al., benefit in one (Wechsler Memory Scale – Revised), but not
2014). another co-primary end point (ADAS-Cog) (Kamphuis et al.,
2011a). Two subsequent trials were performed, the Souvenir
53.3.9.3 Hormone therapy II (mild AD, 24 weeks, n = 259) and S-Connect (mild-to-
moderate AD, on conventional treatment, 24 weeks, n = 527)
Driven by observational evidence in population stud- studies. Souvenir II demonstrated significant benefits over
ies (females) and studies of treatments for prostate cancer placebo in its primary end point, the NTB memory function
(males), the potential therapeutic role for hormones in those Z-score (p = 0.02, Cohen’s d = 0.21). It also showed improved
with AD has led to several trials. Hormone replacement electroencephalogram (EEG) functional connectivity mea-
therapy has not been effective for the treatment of AD in sures with Souvenaid treatment (Scheltens et al., 2012). By
females and most recently the ‘COSTAR’ study of tamoxifen/ contrast, the S-Connect study did not demonstrate any dif-
raloxifene was negative. Leuprolide, which affects levels of ference in ADAS-Cog in mild-to-moderate AD on cholines-
luteinizing hormone (Casadesus et al., 2004) was ineffective terase treatment (Shah et al., 2013).
in two large 52-week Phase III trials conducted by Voyager The treatment is usually well-tolerated. Weight gain
Pharmaceuticals despite earlier promising Phase II results. is reported, however, this is a benefit as development of
Dehydroepiandrosterone (DHEA) dione has also been dementia in AD is commonly associated with weight loss.
trialled in AD subjects, and again results were negative. It In Souvenir I, post hoc analyses reported greatest functional
appears very unlikely that any hormonal therapy for AD benefits in those with lower pretreatment BMI (Kamphuis
will be available within the next decade. et al., 2011b)
Drug treatments in development for Alzheimer’s disease 577

A small study has tested Souvenaid in behavioural 53.3.9.5.4 DOCOSAHEXAENOIC ACID


variant FTD, with reported improvements in behavioural DHA is the most abundant long-chain fatty acid in the
symptoms and social cognition skills (Pardini et al., 2015). brain. Epidemiological evidence suggest that high DHA
Souvenaid is also being evaluated in prodromal AD: the consumption is associated with lower risk of AD, and in
LipiDiDiet study has recruited around 300 participants and animal models, DHA lowers amyloid and p-tau levels. It
the results of the 2-year trial, with cognitive (NTB, ADAS- is suggested that it may improve cardiovascular health as
Cog), functional (ADCS-ADL), neuroimaging (MRI), blood well as influencing AD pathology. However, a large ran-
and CSF biomarkers outcomes (Freund-Levi et al., 2012), domized, double-blind, placebo-controlled trial of DHA
will be reported in 2015. in mild-to-moderate AD (n = 402, MMSE 14–26) showed
no effect on cognitive decline overall, although there was
a suggestion of benefit in APOE ε4 non-carriers, with
53.3.9.5.2 KETOSYN (AXONA/AC-1202) AND AC-1204
slower cognitive decline seen in this subgroup (Quinn
In AD there is impaired uptake and utilization of glucose et al., 2010).
in vulnerable brain regions. AC-1202 and AC-1204 are DHA/Omega 3 is being tested in the large secondary
formulations of caprylic acid (found in coconut oil) and prevention Multi-domain Alzheimer Preventive Trial
medium-chain triglycerides, which are converted in the (MAPT) – which also includes a multi-domain behavioural
liver to ‘ketone bodies’ (β-hydroxobutyrate, acetoacete and intervention arm (nutritional, physical and cognitive train-
acetone). These compounds cross the BBB and can provide ing) (Ousset, 2010).
an alternative energy source for mitochondrial metabolism,
even with a carbohydrate-rich diet.
A large multicentre Phase II trial of AC-1202 (n = 152, 53.3.9.5.5 GINKGO BILOBA/EGB761
mild-to-moderate AD) showed cognitive differences ini- Ginkgo biloba is a traditional Chinese medicine that has
tially (at 45 days) but these did not persist to study end multiple suggested mechanisms of action in AD includ-
(90 days). However, in pre-specified analyses stratified by ing improving mitochondrial metabolism, inhibiting Aβ
ε4 status, there were five-point differences seen in ADAS- aggregation and reducing blood viscosity/increasing perfu-
Cog in ε4 non-carriers at 90 days (p = 0.008) and cogni- sion. A recent meta-analysis of seven RCTs of Ginkgo biloba
tive improvement correlated with higher serum levels of extract EGb761 in AD (n = 2684) suggested symptomatic
the ketone body, β-hydroxobutyrate. Unfortunately, 23% of benefit, although a large study (n = 2854) of its use in demen-
the active group discontinued from the trial, with gastro- tia prevention was negative (Vellas et al., 2012; Gauthier and
intestinal adverse effects in common (diarrhoea, dyspepsia, Schlaefke, 2014).
flatulence) (Henderson et al., 2009). Instead of pursuing
a Phase III trial, AC-1202 has been brought to market as a
medical food under the tradename Axona. 53.3.9.6 Newer symptomatic treatments
NOURISH AD, a Phase II/III trial of AC-1204 (a in behavioural and psychological
­follow-up formulation of AC-1202) is under way in mild-to-­ symptoms of dementia
moderate AD (n = 480, 26 weeks).
53.3.9.6.1 RO40602522/RG1577
RO40602522 (Evotech AG/Roche) is a centrally act-
53.3.9.5.3 EPIGALLOCATECHIN-3-GALLATE (ECCG)
ing inhibitor of MAO-B, an enzyme that inactivates
EGCG is the main biologically active polyphenol in green monoamines such as dopamine. MAO-B expression is
tea. In vitro, it has numerous beneficial attributes, includ- higher in AD, which may contribute to neurodegenera-
ing antioxidant, anti-inflammatory and antiatherogenic tion, as its activity generates reactive oxygen species. In
properties. It also influences growth-factor-mediated addition, reduced dopaminergic function in AD has
pathways, protein kinase pathways, has anti-aggregation been linked with behavioural symptoms including apa-
effects on Aβ and possibly promotes α-secretase activ- thy and ADL impairment. It is currently being tested in
ity. Unfortunately, its in vivo effects are less consistent, Phase II in AD as an adjunct to a ChEI or memantine
which may be due to inability to attain therapeutic con- (Mayflower RoAD, MMSE 13–20), with change in the
centrations within the brain (Mereles and Hunstein, ADAS-Cog-11 (cognitive behaviour subscale) the primary
2011). A pilot study in Down’s syndrome (n = 3 1, age < 29 outcome measure.
years, 3 months’ treatment) resulted in better immediate
and working memory performance, but poorer executive
function (psychomotor speed) (De la Torre and Dierssen, 53.3.9.6.2 BREXIPIPRAZOLE/OPC-34712
2012). Brexipiprazole/OPC-34712 is a quinolinone partial ago-
EGCG is currently in Phase II/III in AD patients with nist of the dopamine receptor D2, related to the atypi-
and without concomitant donepezil therapy. It is also being cal antipsychotic aripiprazole. It also has partial agonist
tested at Phase II/III in Down’s syndrome and multiple sys- effects against serotonin 5-HT1a receptors. Its primary
tem atrophy. indication is in schizophrenia and as add-on therapy in
578 Dementia

depression; however, it is also being tested in two Phase may ‘fast-track’ drugs for AD to become available within
III studies for symptoms of agitation in moderate-to- the next few years (e.g. liraglutide, exendin-4, levetirace-
severe AD. tam). Availability and affordability are likely to be limiting
factors in the uptake of new therapies.
53.3.9.6.3 AVP-923/NUEDEXTRA (AVANIR) The application of therapeutics for AD is complex and
AVP-923/Nuedextra (Avanir) is a combination of dextro- depends not only on disease stage and severity but also on
methorphan (a weak NMDA antagonist) and quinidine (an individual clinical (e.g. diabetic or non-diabetic) or phar-
antiarrhythmic that increases the bioavailability of dextro- macogenomic differences (e.g. some, like levetiracetam,
methorphan). It is currently also in evaluation for pseudo- may work in APOE ε4 carriers but not in non-carriers).
bulbar effect in multiple sclerosis and motor neurone disease, Life course risk factors including physical activity, cogni-
dyskinesia in PD and neuropathic pain. A Phase II study tive reserve and diet are also being assessed in several large,
tested its efficacy against agitation in AD, with benefit on the randomized clinical trials internationally, results of which
NPI aggression/agitation domain. ultimately may have even greater impact on dementia prev-
alence than pharmacological therapies. In addition, the fre-
53.3.9.6.4 PRAZOCIN quent presence of multiple neuropathologies in those with
dementia (Scheltens et al., 2012) may necessitate multiple
Prazocin is an α-1 adrenoreceptor antagonist, which has therapeutic agents targeting the individual pathologies.
vasodilatory effects on vascular smooth muscle. It is in Indeed, it may be as important to target the contributory
clinical use for hypertension and benign prostatic hyper- pathologies as the clinical features.
trophy. In APP23 transgenic mice, prazocin mediated an AD therapy may be used similarly to therapies for
anti-­inflammatory response and improved memory deficits other chronic diseases – a mixture of disease-modifying
(Katsouri et al., 2013). α-1Adrenergic blockade may also atten- and symptomatic therapies used both in combination and
uate Aβ-mediated cerebral vasoconstriction and improve sequentially, over the course of the disease.
cerebral perfusion (Haase et al., 2013). Administration of The prospect of applying preventative or preclinical
prazocin 1–6 mg daily in probable AD with agitation/aggres- interventions in AD is tantalizing. However, dementia prev-
sion (n = 22) resulted in significant improvements in NPI, alence rates are likely to continue to increase worldwide,
Brief Psychiatric Rating Scale (BPRS) and CGIC. A larger AD even once disease-modifying agents become available.
trial for agitation (target n = 120) is recruiting. Continued efforts into identifying and implementing novel
53.3.9.6.5 CIRCADIN/MELATONIN approaches for symptoms and behaviour in AD dementia
also remain imperative.
Melatonin secretion from the pineal gland declines in
AD, with sleep–wake cycle disturbance and nocturnal
awakening a common contributor to carer fatigue. In REFERENCES
addition to restoring sleep–wake phase, in animal mod-
ADAPT Research Group, Martin, B.K., Szekely, C.
els, melatonin reduces Aβ aggregation/oligomerization,
et al. (2008). Cognitive function over time in the
inhibits tau hyperphosphorylation, regulates α-secretase
Alzheimer’s disease anti-inflammatory prevention trial
(to promote non-amyloidogenic pathway) and has anti-
(ADAPT): Results of a randomized, controlled trial of
oxidant properties (Zhang et al., 2015). Early small open-
naproxen and celecoxib. Archives of Neurology, 65
label or placebo-controlled trials have hinted at cognitive
(7): 896–905.
benefits of melatonin, although a large trial was negative
Adolfsson, O., Pihlgren, M., Toni, N. et al. (2012). An effec-
for improvement in sleep. More recently, a Phase II study
tor-reduced anti-β-amyloid (Aβ) antibody with unique
of prolonged-release melatonin as add-on to donepezil
aβ binding properties promotes neuroprotection and
(+/− memantine) in mild-to-moderate AD (n = 80) dem-
glial engulfment of Aβ. Journal of Neuroscience, 32
onstrated significant positive effects on cognition and
(28): 9677–9689.
sleep, most marked in those reporting poor sleep at study
Aisen, P.S., Cummings, J. and Schneider, L.S. (2012).
outset (Wade et al., 2014). Circadin is a slow release mela-
Symptomatic and nonamyloid/tau based pharmaco-
tonin preparation.
logic treatment for Alzheimer disease. Cold Spring
Harbor Perspectives in Medicine, 2 (3): a006395.
Aisen, P.S., Gauthier, S., Ferris, S.H. et al. (2011).
Tramiprosate in mild-to-moderate Alzheimer’s disease –
53.4 THE NEAR FUTURE: THERAPIES IN A randomized, double-blind, placebo-controlled,
DEMENTIA multi-centre study (the Alphase Study). Archives of
Medical Science, 7 (1): 102–111.
Despite a number of high-profile disappointments, there Aisen, P.S., Saumier, D., Briand, R. et al. (2006). A Phase
remain many extremely promising treatments that may yet II study targeting amyloid-beta with 3APS in mild-
make the jump into clinical use. Application of therapies to-moderate Alzheimer disease. Neurology, 67 (10):
that have already survived Phase III for other indications 1757–1763.
Drug treatments in development for Alzheimer’s disease 579

Alvarez, X.A., Cacabelos, R., Laredo, M. et al. (2006). www.biogenidec.com/press_release_details.aspx?


A 24-week, double-blind, placebo-controlled study of ID=14712&Action=1&NewsId=2486&M=NewsV2
three dosages of Cerebrolysin in patients with mild to &PID=61997. Accessed 21 March 2015.
moderate Alzheimer’s disease. European Journal of Boche, D., Donald, J., Love, S. et al. (2010). Reduction of
Neurology, 13 (1): 43–54. aggregated Tau in neuronal processes but not in the
Alvarez, X.A., Cacabelos, R., Sampedro, C. et al. (2011). cell bodies after Abeta42 immunisation in Alzheimer’s
Combination treatment in Alzheimer’s disease: results disease. Acta Neuropathologica (Berlin), 120 (1): 13–20.
of a randomized, controlled trial with cerebrolysin Boyd, T.D., Bennett, S.P., Mori, T. et al. (2010). GM-CSF
and donepezil. Current Alzheimer Research, 8 (5): upregulated in rheumatoid arthritis reverses cognitive
583–591. impairment and amyloidosis in Alzheimer mice. Journal
Ancoli-Israel, S., Palmer, B.W., Cooke, J.R. et al. (2008). of Alzheimer’s Disease, 21 (2): 507–518.
Cognitive effects of treating obstructive sleep apnea Broadstock, M., Ballard, C. and Corbett, A. (2014). Latest
in Alzheimer’s disease: A randomized controlled study. treatment options for Alzheimer’s disease, Parkinson’s
Journal of the American Geriatrics Society, 56 (11): disease dementia and dementia with Lewy bod-
2076–2081. ies. Expert Opinion on Pharmacotherapy, 15 (13):
Antigoni Ekonomou, S.L.M. (2010). Stem cells and demen- 1797–1810.
tia. The Journal of Quality Research in Dementia, Burstein, A.H., Zhao, Q., Ross, J. et al. (2013). Safety and
Issue 1; www.alzheimers.org.uk/site/scripts/documents_ pharmacology of ponezumab (PF-04360365) after
info.php?documentID=77&pageNumber=7. Accessed a single 10-minute intravenous infusion in subjects
14 June 2016. with mild to moderate Alzheimer disease. Clinical
ASPREE Investigator Group (2013). Study design of Neuropharmacology, 36 (1): 8–13.
ASPirin in Reducing Events in the Elderly (ASPREE): Burt, R.K., Balabanov, R., Han, X. et al. (2015). Association
A randomized, controlled trial. Contemporary Clinical of nonmyeloablative hematopoietic stem cell trans-
Trials, 36 (2): 555–564. plantation with neurological disability in patients with
Asterias Biotherapeutics (2016). AST-OPC1 Clinical relapsing-remitting multiple sclerosis. JAMA, 313 (3):
Program (website). Available at www.asteriasbiothera- 275–284.
peutics.com/pipeline/ast-opc1/. Accessed 14 June 2016. Carlsson, C.M., Gleason, C.E., Hess, T.M. et al. (2008).
Bakker, A., Krauss, G.L., Albert, M.S. et al. (2012). Effects of simvastatin on cerebrospinal fluid biomark-
Reduction of hippocampal hyperactivity improves cog- ers and cognition in middle-aged adults at risk for
nition in amnestic mild cognitive impairment. Neuron, Alzheimer’s disease. Journal of Alzheimer’s Disease, 13
74 (3): 467–474. (2): 187–197.
Barten, D.M., Fanara, P., Andorfer, C. et al. (2012). Casadesus, G., Zhu, X., Atwood, C.S. et al. (2004).
Hyperdynamic microtubules, cognitive deficits, and Beyond estrogen: Targeting gonadotropin hormones
pathology are improved in tau transgenic mice with in the treatment of Alzheimer’s disease. Current
low doses of the microtubule-stabilizing agent BMS- Drug Targets-CNS and Neurological Disorders, 3 (4):
241027. Journal of Clinical Neuroscience, 32 (21): 281–285.
7137–7145. Chen, H.-K., Ji, Z.-S., Dodson, S.E. et al. (2011).
Bateman, R.J., Siemers, E.R., Mawuenyega, K.G., Wen, G., Apolipoprotein E4 domain interaction mediates
et al. (2009). A gamma-secretase inhibitor decreases detrimental effects on mitochondria and is a potential
amyloid-beta production in the central nervous sys- therapeutic target for Alzheimer disease. Journal of
tem. Annals of Neurology, Jul;66(1): 48–54. Biological Chemistry, 286 (7): 5215–5221.
Bateman, R.J., Xiong, C., Benzinger, T.L.S. et al. (2012). Chen, S., Liu, A., An, F. et al. (2012). Amelioration of
Clinical and biomarker changes in dominantly inher- neurodegenerative changes in cellular and rat models
ited Alzheimer’s disease. New England Journal of of diabetes-related Alzheimer’s disease by exendin-4.
Medicine, 367 (9): 795–804. Age (Dordrecht, Netherlands), 34 (5): 1211–1224.
Bayer, A.J., Bullock, R., Jones, R.W. et al. (2005). Chen, N., Yang, M., Guo, J. et al. (2013). Cerebrolysin for
Evaluation of the safety and immunogenicity of vascular dementia. Cochrane Database Systematic
synthetic Abeta42 (AN1792) in patients with AD. Reviews, (1): CD008900.
Neurology, 64 (1): 94–101. Cherny, R.A., Atwood, C.S., Xilinas, M.E. et al. (2001).
Benedict, C., Hallschmid, M., Hatke, A. et al. (2004). Treatment with a copper-zinc chelator markedly
Intranasal insulin improves memory in humans. and rapidly inhibits beta-amyloid accumulation in
Psychoneuroendocrinology, 29 (10): 1326–1334. Alzheimer’s disease transgenic mice. Neuron, 30 (3):
Biogen (2015). Biogen Idec Presents Positive Interim 665–676.
Results from Phase 1B Study of Investigational Chiang, K. and Koo, E.H. (2014). Emerging therapeu-
Alzheimer’s Disease Treatment Aducanumab (BIIB037) tics for Alzheimer’s disease. Annual Review of
at 2015 AD/PD Conference (webpage). Available at Pharmacology Toxicology, 54: 381–405.
580 Dementia

Corcoran, N.M., Martin, D., Hutter-Paier, B. et al. (2010). Do, G.-M., Choi, M.-S., Kim, H.-J. et al. (2008). Soy pinitol
Sodium selenate specifically activates PP2A phos- acts partly as an insulin sensitizer or insulin mediator
phatase, dephosphorylates tau and reverses memory in 3T3-L1 preadipocytes. Genes and Nutrition, 2 (4):
deficits in an Alzheimer’s disease model. Journal of 359–364.
Clinical Neuroscience, 17 (8): 1025–1033. Doody, R.S., Gavrilova, S.I., Sano, M. et al. (2008). Effect
Coric, V., Salloway, S., van Dyck, C. et al. (2013). A phase of dimebon on cognition, activities of daily living,
II study of the gamma-secretase inhibitor avagace- behaviour, and global function in patients with mild-
stat (BMS-708163) in predementia Alzheimer’s dis- to-moderate Alzheimer’s disease: A randomised,
ease. Alzheimer’s and Dementia: The Journal of the double-blind, placebo-controlled study. Lancet, 372
Alzheimer’s Association, 9 (Suppl. 4): S283. (9634): 207–215.
Coric, V., van Dyck, C.H., Salloway, S. et al. (2012). Doody, R.S., Raman, R., Farlow, M. et al. (2013). A phase
Safety and tolerability of the γ-secretase inhibitor 3 trial of semagacestat for treatment of Alzheimer’s
avagacestat in a phase 2 study of mild to moderate disease. New England Journal of Medicine, 369 (4):
Alzheimer disease. Archives of Neurology, 69 (11): 341–350.
1430–1440. Dubois, B., Feldman, H.H., Jacova, C. et al. (2007).
Craft S. (2005). Insulin resistance syndrome and Research criteria for the diagnosis of Alzheimer’s
Alzheimer’s disease: Age- and obesity-related effects disease: Revising the NINCDS-ADRDA criteria. The
on memory, amyloid, and inflammation. Neurobiology Lancet Neurology, 6 (8): 734–746.
of Aging, Dec;26 Suppl 1: 65–9. Dysken, M.W., Sano, M., Asthana, S. et al. (2014). Effect
Craft, S., Baker, L.D., Montine, T.J. et al. (2012). Intranasal of vitamin E and memantine on functional decline in
insulin therapy for Alzheimer disease and amnestic Alzheimer disease: The Team-AD VA cooperative ran-
mild cognitive impairment. Archives of Neurology, 69 domized trial. JAMA, 311 (1): 33–44.
(1): 29–38. Endres, K., Fahrenholz, F., Lotz, J. et al. (2014).
Cramer, P.E., Cirrito, J.R., Wesson, D.W. et al. (2012). Increased CSF APPs-α levels in patients with
ApoE-directed therapeutics rapidly clear β-amyloid Alzheimer disease treated with acitretin. Neurology,
and reverse deficits in AD mouse models. Science, 335 83 (21): 1930–1935.
(6075): 1503–1506. Eriksdotter Jönhagen, M., Nordberg, A., Amberla, K.
Crespi, G.A.N., Ascher, D.B., Parker, M.W. and Miles, L.A. et al. (1998). Intracerebroventricular infusion of nerve
(2014). Crystallization and preliminary X-ray diffraction growth factor in three patients with Alzheimer’s
analysis of the fab portion of the Alzheimer’s disease disease. Dementia and Geriatric Cognitive Disorders,
immunotherapy candidate bapineuzumab complexed 9 (5): 246–257.
with amyloid-β. Acta Crystallographica Section F, Fan, X., Sun, D., Tang, X. et al. (2014). Stem-cell challenges
Structural Biology and Crystallization Communications, in the treatment of Alzheimer’s disease: A long way
70 (Pt 3): 374–377. from bench to bedside. Medical Research Reviews, 34
Cumbo, E. and Ligori, L.D. (2010). Levetiracetam, (5): 957–978.
lamotrigine, and phenobarbital in patients with epi- Ferreira, D., Westman, E., Eyjolfsdottir, H. et al. (2015).
leptic seizures and Alzheimer’s disease. Epilepsy and Brain changes in Alzheimer’s disease patients
Behavior, 17 (4): 461–466. with implanted encapsulated cells releasing nerve
Dart NeuroScience (2014). Dart Neuroscience Drug growth factor. Journal of Alzheimer’s Disease, 43 (3):
Programs. Available at www.dartneuroscience.com/ 1059–1072.
DrugPrograms.php. Accessed 20 January 2015. Ferretti, M.T., Allard, S., Partridge, V. et al. (2012).
De la Torre, R. and Dierssen, M. (2012). Therapeutic Minocycline corrects early, pre-plaque neuroinflam-
approaches in the improvement of cognitive perfor- mation and inhibits BACE-1 in a transgenic model of
mance in Down syndrome: Past, present, and future. Alzheimer’s disease-like amyloid pathology. Journal of
Progress in Brain Research, 197: 1–14. Neuroinflammation, 9: 62.
Deane, R.J. (2012). Is RAGE still a therapeutic target for Fillit, H., Hess, G., Hill, J. et al. (2009). IV immunoglobu-
Alzheimer’s disease? Future Medicinal Chemistry, 4 (7): lin is associated with a reduced risk of Alzheimer
915–925. disease and related disorders. Neurology, 73 (3):
Deardorff, W.J., Shobassy, A. and Grossberg, G.T. (2015). 180–185.
Safety and clinical effects of EVP-6124 in subjects with Fleisher, A.S., Raman, R., Siemers, E.R. et al. (2008). Phase
Alzheimer’s disease currently or previously receiving 2 safety trial targeting amyloid beta production with
an acetylcholinesterase inhibitor medication. Expert a gamma-secretase inhibitor in Alzheimer disease.
Review of Neurotherapeutics, 15 (1): 7–17. Archives of Neurology, 65 (8): 1031–1038.
Demattos, R.B., Lu, J., Tang, Y. et al. (2012). A plaque- Forlenza, O.V., de Paula, V.J., Machado-Vieira, R. et al.
specific antibody clears existing β-amyloid plaques in (2012). Does lithium prevent Alzheimer’s disease?
Alzheimer’s disease mice. Neuron, 76 (5): 908–920. Drugs and Aging, 29 (5): 335–342.
Drug treatments in development for Alzheimer’s disease 581

Freiherr, J., Hallschmid, M., Frey, W.H. et al. (2013). Gold, M., Alderton, C., Zvartau-Hind, M. et al. (2010).
Intranasal insulin as a treatment for Alzheimer’s dis- Rosiglitazone monotherapy in mild-to-moderate
ease: A review of basic research and clinical evidence. Alzheimer’s disease: Results from a randomized,
CNS Drugs, 27 (7): 505–514. double-blind, placebo-controlled phase III study.
Frenkel, D., Puckett, L., Petrovic, S. et al. (2008). A nasal Dementia and Geriatric Cognitive Disorders, 30 (2):
proteosome adjuvant activates microglia and pre- 131–146.
vents amyloid deposition. Annals of Neurology, 63 (5): Green, R.C., Schneider, L.S., Amato, D.A. et al. (2009).
591–601. Effect of tarenflurbil on cognitive decline and activities
Freund-Levi, Y., Visser, P.J., Kivipelto, M. et al. (2012). of daily living in patients with mild Alzheimer dis-
The LipiDiDiet study: Rationale and study design. ease: A randomized controlled trial. JAMA, 302 (23):
Alzheimer’s and Dementia: The Journal of the 2557–2564.
Alzheimer’s Association, 8 (Suppl. 4): S602. Grove, R.A., Harrington, C.M., Mahler, A. et al. (2014).
Frölich, L., Ashwood, T., Nilsson, J. and Eckerwall A randomized, double-blind, placebo-controlled,
G; Sirocco Investigators. (2011). Effects of 16-week study of the H3 receptor antagonist,
AZD3480 on cognition in patients with mild-to- GSK239512 as a monotherapy in subjects with mild-
moderate Alzheimer’s disease: A phase IIb dose- to-moderate Alzheimer’s disease. Current Alzheimer
finding study. Journal of Alzheimer’s Disease, 24 (2): Research, 11 (1): 47–58.
363–374. Gunn, A.P., Masters, C.L. and Cherny, R.A. (2010).
Galasko, D., Bell, J., Mancuso, J.Y. et al. (2014). Clinical trial Pyroglutamate-Aβ: Role in the natural history of
of an inhibitor of RAGE-Aβ interactions in Alzheimer Alzheimer’s disease. The International Journal of
disease. Neurology, 82 (17): 1536–1542. Biochemistry and Cell Biology, 42 (12): 1915–1918.
Galasko, D.R., Graff-Radford, N., May, S. et al. (2007). Haase, N., Herse, F., Spallek, B. et al. (2013). Amyloid-β
Safety, tolerability, pharmacokinetics, and Abeta levels peptides activate α1-adrenergic cardiovascular recep-
after short-term administration of R-flurbiprofen in tors. Hypertension, 62 (5): 966–972.
healthy elderly individuals. Alzheimer Disease and Hanyu, H., Hirao, K., Shimizu, S. et al. (2007). Favourable
Associated Disorders, 21 (4): 292–299. effects of nilvadipine on cognitive function and
Garcia-Alloza, M., Borrelli, L.A., Rozkalne, A. et al. (2007). regional cerebral blood flow on SPECT in hyperten-
Curcumin labels amyloid pathology in vivo, disrupts sive patients with mild cognitive impairment. Nuclear
existing plaques, and partially restores distorted Medicine Communication, 28 (4): 281–287.
neurites in an Alzheimer mouse model. Journal of Hardy, J. and Selkoe, D.J. (2002). The amyloid hypoth-
Neurochemistry, 102 (4): 1095–1104. esis of Alzheimer’s disease: Progress and problems
García-Osta, A., Cuadrado-Tejedor, M., García-Barroso, on the road to therapeutics. Science, 297 (5580):
C. et al. (2012). Phosphodiesterases as therapeu- 353–356.
tic targets for Alzheimer’s disease. ACS Chemical He, P., Cheng, X., Staufenbiel, M. et al. (2013). Long-term
Neuroscience, 3 (11): 832–844. treatment of thalidomide ameliorates amyloid-like
Gault, L., Ritchie, C., Robieson, W. et al. (2013). Efficacy pathology through inhibition of β-secretase in a
and safety of the alpha7 agonist ABT-126 in mild-to- mouse model of Alzheimer’s disease. PLoS One, 8 (2):
moderate Alzheimer’s dementia. Alzheimer’s and e55091.
Dementia: The Journal of the Alzheimer’s Association, Heckman, P.R.A., Wouters, C. and Prickaerts, J. (2014).
9 (Suppl. 4): S138. Phosphodiesterase inhibitors as a target for cogni-
Gauthier, S., Proaño, J.V., Jia, J. et al. (2015). Cerebrolysin in tion enhancement in aging and Alzheimer’s disease:
mild-to-moderate Alzheimer’s disease: A meta-­analysis A translational overview. Current Pharmaceutical
of randomized controlled clinical trials. Dementia and Design, 21 (3): 317–331.
Geriatric Cognitive Disorders, 39 (5–6): 332–47. Henderson, S.T., Vogel, J.L., Barr, L.J. et al. (2009). Study
Gauthier, S. and Schlaefke, S. (2014). Efficacy and toler- of the ketogenic agent AC-1202 in mild to moderate
ability of Ginkgo biloba extract EGb 761® in dementia: Alzheimer’s disease: A randomized, double-blind,
A systematic review and meta-analysis of random- placebo-controlled, multicenter trial. Nutrition and
ized placebo-controlled trials. Clinical Interventions in Metabolism, 6: 31.
Aging, 9: 2065–2077. Hickman, D.T., López-Deber, M.P., Ndao, D.M. et al. (2011).
Gerhard, T., Devanand, D.P., Huang, C. et al. (2015). Sequence-independent control of peptide confor-
Lithium treatment and risk for dementia in adults with mation in liposomal vaccines for targeting protein
bipolar disorder: population-based cohort study. The misfolding diseases. Journal of Biological Chemistry,
British Journal of Psychiatry, 207 (1): 46–51. 286 (16): 13966–13976.
Ghiso, J., Shayo, M., Calero, M. et al. (2004). Systemic Hock, C., Konietzko, U., Streffer, J.R. et al. (2003).
catabolism of Alzheimer’s Abeta40 and Abeta42. Antibodies against beta-amyloid slow cognitive
Journal of Biological Chemistry, 279 (44): decline in Alzheimer’s disease. Neuron, 38 (4):
45897–45908. 547–554.
582 Dementia

Holmes, C., Butchart, J., Wolfe, L. et al. (2014). The safety with Mild-to-Moderate Alzheimer’s Disease. Barcelona,
and tolerability of etanercept in Alzheimer’s disease Spain: The Journal of Prevention of Alzheimer’s
(steadi-09): A phase II double blind randomised pla- Disease.
cebo controlled trial. Alzheimer’s and Dementia: The Kukar, T.L., Ladd, T.B., Bann, M.A. et al. (2008). Substrate-
Journal of the Alzheimer’s Association, 10 (Suppl. 4): targeting gamma-secretase modulators. Nature, 453
S273. (7197): 925–929.
Hu, X., Zhou, X., He, W. et al. (2010). BACE1 deficiency Kukar, T., Prescott, S., Eriksen, J.L. et al. (2007). Chronic
causes altered neuronal activity and neurodegenera- administration of R-flurbiprofen attenuates learning
tion. The Journal of Neuroscience, 30 (26): 8819–8829. impairments in transgenic amyloid precursor protein
Huang, X.-T., Qian, Z.-M., He, X. et al. (2014). Reducing mice. BMC Neuroscience, 8: 54.
iron in the brain: A novel pharmacologic mechanism of Kwok, T., Lee, J., Law, C.B. et al. (2011). A randomized
huperzine A in the treatment of Alzheimer’s disease. placebo controlled trial of homocysteine lowering to
Neurobiology of Aging, 35 (5): 1045–1054. reduce cognitive decline in older demented people.
Iliff, J.J. and Nedergaard, M. (2013). Is there a cerebral Clinical Nutrition, 30 (3): 297–302.
lymphatic system? Stroke, 44 (Suppl. 1): S93–S95. La Porte, S.L., Bollini, S.S., Lanz, T.A. et al. (2012).
Janusz, M. and Zabłocka, A. (2010). Colostral proline-rich Structural basis of C-terminal β-amyloid peptide bind-
polypeptides – Immunoregulatory properties and ing by the antibody ponezumab for the treatment of
prospects of therapeutic use in Alzheimer’s disease. Alzheimer’s disease. Journal of Molecular Biology, 421
Current Alzheimer Research, 7 (4): 323–333. (4–5): 525–536.
Jim, H.S., Boyd, T.D., Booth-Jones, M. et al. (2012). Lahiri, D.K., Chen, D., Maloney, B. et al. (2007). The
Granulocyte macrophage colony stimulating factor experimental Alzheimer’s disease drug posi-
treatment is associated with improved cognition in phen [(+)-phenserine] lowers amyloid-beta pep-
cancer patients. Brain Disorders and Therapy, 1 (1): tide levels in cell culture and mice. Journal of
1000101. Pharmacology and Experimental Therapeutics, 320
Jones, R.W., Kivipelto, M., Feldman, H. et al. (2008). (1): 386–396.
The Atorvastatin/Donepezil in Alzheimer’s Disease Lahmy, V., Long, R., Morin, D. et al. (2015). Mitochondrial
Study (LEADe): Design and baseline characteris- protection by the mixed muscarinic/σ1 ligand
tics. Alzheimer’s and Dementia: The Journal of the ANAVEX2-73, a tetrahydrofuran derivative, in Aβ25–35
Alzheimer’s Association, 4 (2): 145–153. peptide-injected mice, a nontransgenic Alzheimer’s
Jonsson, T., Atwal, J.K., Steinberg, S. et al. (2012). A muta- disease model. Frontiers in Cellular Neuroscience,
tion in APP protects against Alzheimer’s disease and 8: 463.
age-related cognitive decline. Nature, 488 (7409): 96–99. Lahmy, V., Meunier, J., Malmström, S. et al. (2013).
Kamphuis, P.J.G.H., Verhey, F.R.J., Olde Rikkert, M.G.M. Blockade of tau hyperphosphorylation and Aβ1–42
et al. (2011a). Efficacy of a medical food on cognition generation by the aminotetrahydrofuran derivative
in Alzheimer’s disease: Results from secondary analy- ANAVEX2-73, a mixed muscarinic and σ1 recep-
ses of a randomized, controlled trial. The Journal of tor agonist, in a nontransgenic mouse model of
Nutrition, Health and Aging, 15 (8): 720–724. Alzheimer’s disease. Neuropsychopharmacology, 38
Kamphuis, P.J.G.H., Verhey, F.R.J., Olde Rikkert, M.G.M. (9): 1706–1723.
et al. (2011b). Effect of a medical food on body mass Lambracht-Washington, D. and Rosenberg, R.N. (2013).
index and activities of daily living in patients with Anti-amyloid beta to tau-based immunization:
Alzheimer’s disease: Secondary analyses from a Developments in immunotherapy for Alzheimer dis-
randomized, controlled trial. The Journal of Nutrition, ease. ImmunoTargets and Therapy, 2: 105–14.
Health and Aging, 15 (8): 672–676. Lannfelt, L., Blennow, K., Zetterberg, H. et al. (2008).
Katsouri, L., Vizcaychipi, M.P., McArthur, S. et al. (2013). Safety, efficacy, and biomarker findings of PBT2 in tar-
Prazosin, an α(1)-adrenoceptor antagonist, prevents geting Abeta as a modifying therapy for Alzheimer’s
memory deterioration in the APP23 transgenic mouse disease: A phase IIa, double-blind, randomised,
model of Alzheimer’s disease. Neurobiology of Aging, placebo-controlled trial. The Lancet Neurology, 7 (9):
34 (4): 1105–1115. 779–786.
Kennelly, S.P., Abdullah, L., Paris, D. et al. (2011). Lannfelt, L., Möller, C., Basun, H. et al. (2014).
Demonstration of safety in Alzheimer’s patients Perspectives on future Alzheimer therapies: Amyloid-β
for intervention with an anti-hypertensive drug protofibrils – A new target for immunotherapy with
Nilvadipine: Results from a 6-week open label study. BAN2401 in Alzheimer’s disease. Alzheimer’s Research
International Journal of Geriatric Psychiatry, 26 (10): Therapy, 6 (2): 16.
1038–1045. Li, X.-Y., Bao, X.J. and Wang, R.-Z. (2015). Potential of
Ketter, N., Liu, E., Di, J. et al. (2014). A Phase 2 neural stem cell-based therapies for Alzheimer’s
Randomized Double-Blind Placebo-Controlled Study disease. Journal of Neuroscience Research, 93 (9):
of Vanutide Cridificar Vaccine (ACC-001) in Patients 1313–1324.
Drug treatments in development for Alzheimer’s disease 583

Li, Y., Duffy, K.B., Ottinger, M.A. et al. (2010). GLP-1 Nardone, R., Tezzon, F., Höller, Y. et al. (2014). Transcranial
receptor stimulation reduces amyloid-beta peptide magnetic stimulation (TMS)/repetitive TMS in mild
accumulation and cytotoxicity in cellular and animal cognitive impairment and Alzheimer’s disease. Acta
models of Alzheimer’s disease. Journal of Alzheimer’s Neurologica Scandinavica, 129 (6): 351–366.
Disease, 19 (4): 1205–1219. Neurodyn Inc. (2014). Positive Phase 1a outcome for
Liu, Y.-H., Wang, Y.-R., Xiang, Y. et al. (2014). Clearance Memogain® means potential new treatment for
of amyloid-beta in Alzheimer’s disease: Shifting Alzheimer’s disease (website). Available at www.
the action site from center to periphery. Molecular neurodyn-inc.com/positive-phase-1a-outcome-for-
Neurobiology, 51 (1): 1–7. memogain-means-potential-new-treatment-for-
Lovestone, S., Boada, M., Dubois, B. et al. (2014). A phase alzheimers-disease/. Accessed 20 January 2015.
II trial of tideglusib in Alzheimer’s disease. Journal of Orgogozo, J.-M., Gilman, S., Dartigues, J.-F. et al. (2003).
Alzheimer’s Disease, 45 (1): 75–88. Subacute meningoencephalitis in a subset of patients
Ma, Q.-L., Zuo, X., Yang, F. et al. (2013). Curcumin sup- with AD after Aβ42 immunization. Neurology, 61 (1):
presses soluble tau dimers and corrects molecular 46–54.
chaperone, synaptic, and behavioral deficits in aged Osorio, R.S., Ayappa, I., Mantua, J. et al. (2014).
human tau transgenic mice. Journal of Biological Interaction between sleep-disordered breathing and
Chemistry, 288 (6): 4056–4065. apolipoprotein E genotype on cerebrospinal fluid bio-
Maccecchini, M.L., Chang, M.Y., Pan, C. et al. (2012). markers for Alzheimer’s disease in cognitively normal
Posiphen as a candidate drug to lower CSF amyloid elderly individuals. Neurobiology of Aging, 35 (6):
precursor protein, amyloid-β peptide and τ levels: 1318–1324.
Target engagement, tolerability and pharmacokinetics Ostrowitzki, S., Deptula, D., Thurfjell, L. et al. (2012).
in humans. Journal of Neurology, Neurosurgery and Mechanism of amyloid removal in patients with
Psychiatry, 83 (9): 894–902. Alzheimer disease treated with gantenerumab.
Maelicke, A., Hoeffle-Maas, A., Ludwig, J. et al. (2010). Archives of Neurology, 69 (2): 198–207.
Memogain is a galantamine pro-drug having dramati- Ousset, P.-J. (2010). The multi-domain Alzheimer preven-
cally reduced adverse effects and enhanced efficacy. tive trial (MAPT): A new approach for the preven-
Journal of Molecular Neuroscience, 40 (1–2): 135–137. tion of Alzheimer’s disease- characteristics of the
Maki, T., Okamoto, Y., Carare, R.O. et al. (2014). study population. Alzheimer’s and Dementia: The
Phosphodiesterase III inhibitor promotes drainage Journal of the Alzheimer’s Association, 6 (Suppl. 4):
of cerebrovascular β-amyloid. Annals of Clinical and S146.
Translational Neurology, 1 (8): 519–533. Panza, F., Solfrizzi, V., Frisardi, V. et al. (2009). Beyond
Mereles, D. and Hunstein, W. (2011). Epigallocatechin- the neurotransmitter-focused approach in treating
3-gallate (EGCG) for clinical trials: More pitfalls than Alzheimer’s disease: Drugs targeting beta-amyloid and
promises? International Journal of Molecular Sciences, tau protein. Aging Clinical and Experimental Research,
12 (9): 5592–5603. 21 (6): 386–406.
McClean, P.L. and Hölscher, C. (2014). Liraglutide can Panza, F., Solfrizzi, V., Imbimbo, B.P. et al. (2014). Amyloid-
reverse memory impairment, synaptic loss and based immunotherapy for Alzheimer’s disease in the
reduce plaque load in aged APP/PS1 mice, a model time of prevention trials: The way forward. Expert
of Alzheimer’s disease. Neuropharmacology, 76: Review of Clinical Immunology, 10 (3): 405–419.
57–67. Pardini, M., Serrati, C., Guida, S. et al. (2015). Souvenaid
McGuinness, B., Craig, D., Bullock, R. et al. (2014). Statins reduces behavioral deficits and improves social
for the treatment of dementia. Cochrane Database cognition skills in frontotemporal dementia:
Systematic Reviews, (7): CD007514. A ­proof-of-­concept study. Neurodegenerative
Miller, E.R., Pastor-Barriuso, R., Dalal, D. et al. (2005). Disease, 15 (1): 58–62.
Meta-analysis: High-dosage vitamin E supplementa- Perez-Garmendia, R. and Gevorkian, G. (2013).
tion may increase all-cause mortality. Annals of Internal Pyroglutamate-modified amyloid beta peptides:
Medicine, 142 (1): 37–46. Emerging targets for Alzheimer’s disease immu-
Moon, J.H., Kim, H.J., Yang, A.H. et al. (2012). The effect notherapy. Current Neuropharmacology, 11 (5):
of rosiglitazone on LRP1 expression and amyloid 491–498.
β uptake in human brain microvascular endothelial Petersen, R.C., Thomas, R.G., Grundman, M. et al. (2005).
cells: A possible role of a low-dose thiazolidinedione Vitamin E and donepezil for the treatment of mild cog-
for dementia treatment. The International Journal of nitive impairment. New England Journal of Medicine,
Neuropsychopharmacology, 15 (1): 135–142. 352 (23): 2379–2388.
Morgan, D., Diamond, D.M., Gottschall, P.E. et al. (2000). Piette, F., Belmin, J., Vincent, H. et al. (2011). Masitinib as
A beta peptide vaccination prevents memory loss in an adjunct therapy for mild-to-moderate Alzheimer’s
an animal model of Alzheimer’s disease. Nature, 408 disease: A randomised, placebo-controlled phase 2
(6815): 982–985. trial. Alzheimer’s Research Therapy, 3 (2): 16.
584 Dementia

Porzner, M., Müller, T. and Seufferlein, T. (2009). SR Salloway, S., Sperling, R., Keren, R. et al. (2011). A phase
57746A/xaliproden, a non-peptide neurotrophic 2 randomized trial of ELND005, scyllo-inositol, in mild
compound: Prospects and constraints for the treat- to moderate Alzheimer disease. Neurology, 77 (13):
ment of nervous system diseases. Expert Opinion on 1253–1262.
Investigational Drugs, 18 (11): 1765–1772. Sanchez-Ramos, J., Cimino, C., Avila, R. et al. (2012).
Qiu, C., von Strauss, E., Bäckman, L. et al. (2013). Twenty- Pilot study of granulocyte-colony stimulating fac-
year changes in dementia occurrence suggest tor for treatment of Alzheimer’s disease. Journal of
decreasing incidence in central Stockholm, Sweden. Alzheimer’s Disease, 31 (4): 843–855.
Neurology, 80 (20): 1888–1894. Sano, M., Bell, K.L., Galasko. D. et al. (2011). A random-
Quinn, J.F., Raman, R., Thomas, R.G. et al. (2010). ized, double-blind, placebo-controlled trial of simv-
Docosahexaenoic acid supplementation and cogni- astatin to treat Alzheimer disease. Neurology, 77 (6):
tive decline in Alzheimer disease: A randomized trial. 556–563.
JAMA, 304 (17): 1903–1911. Sano, M., Ernesto, C., Thomas, R.G. et al. (1997). A con-
Rao, M.V., McBrayer, M.K., Campbell, J. et al. (2014). trolled trial of selegiline, alpha-tocopherol, or both as
Specific calpain inhibition by calpastatin prevents treatment for Alzheimer’s disease. The Alzheimer’s
tauopathy and neurodegeneration and restores disease cooperative study. New England Journal of
normal lifespan in tau P301L mice. Journal of Clinical Medicine, 336 (17): 1216–1222.
Neuroscience, 34 (28): 9222–9234. Sato, T., Hanyu, H., Hirao, K. et al. (2011). Efficacy of
Rege, S.D., Geetha, T., Griffin, G.D. et al. (2014). PPAR-γ agonist pioglitazone in mild Alzheimer disease.
Neuroprotective effects of resveratrol in Alzheimer Neurobiology of Aging, 32 (9): 1626–1633.
disease pathology. Frontiers in Aging Neuroscience, Scheltens, P., Twisk, J.W.R., Blesa, R. et al. (2012). Efficacy
6: 218. of Souvenaid in mild Alzheimer’s disease: Results from
Reger, M.A., Watson, G.S., Green, P.S. et al. (2008). a randomized, controlled trial. Journal of Alzheimer’s
Intranasal insulin administration dose-dependently Disease, 31 (1): 225–236.
modulates verbal memory and plasma amyloid-beta in Schenk, D., Barbour, R., Dunn, W. et al. (1999).
memory-impaired older adults. Journal of Alzheimer’s Immunization with amyloid-beta attenuates Alzheimer-
Disease, 13 (3): 323–331. disease-like pathology in the PDAPP mouse. Nature,
Rhinn, H., Fujita, R., Qiang, L. et al. (2013). Integrative 400 (6740): 173–177.
genomics identifies APOE ε4 effectors in Alzheimer’s Schneeberger, A., Hendrix, S., Mattner, F. et al. (2015).
disease. Nature, 500 (7460): 45–50. Results from a phase II study to assess the clinical
Riekse, R.G., Li, G., Petrie, E.C. et al. (2006). Effect of and immunological activity of AFFITOPE® AD02 in
statins on Alzheimer’s disease biomarkers in cere- patients with early Alzheimer’s disease (AD). Journal of
brospinal fluid. Journal of Alzheimer’s Disease, 10 (4): Prevention of Alzheimer’s Disease, 2(2): 103–114.
399–406. Schneider, L.S. (2014). Idalopirdine for Alzheimer’s dis-
Ringman, J.M., Frautschy, S.A., Teng, E. et al. (2012). Oral ease: Written in the stars. The Lancet Neurology, 13
curcumin for Alzheimer’s disease: Tolerability and effi- (11): 1063–1065.
cacy in a 24-week randomized, double blind, placebo- Schneider, L., Porsteinsson, A., Farlow, M. et al. (2013).
controlled study. Alzheimer’s Research Therapy, 4 (5): The neuroprotective and neurotrophic agent T-817MA
43. for Alzheimer’s disease: Randomized, double-blind,
Rogers, K., Felsenstein, K.M., Hrdlicka, L. et al. (2012). placebo-controlled proof-of-concept trial outcomes.
Modulation of γ-secretase by EVP-0015962 reduces Alzheimer’s and Dementia: The Journal of the
amyloid deposition and behavioral deficits in Tg2576 Alzheimer’s Association, 9 (Suppl. 4): S530–S531.
mice. Molecular Neurodegeneration, 7: 61. Seshadri, S., Beiser, A., Selhub, J. et al. (2002). Plasma
Ross, J., Sharma, S., Winston, J. et al. (2013). CHF5074 homocysteine as a risk factor for dementia and
reduces biomarkers of neuroinflammation in patients Alzheimer’s disease. New England Journal of
with mild cognitive impairment: A 12-week, double- Medicine, 346 (7): 476–483.
blind, placebo-controlled study. Current Alzheimer Shah, R.C., Kamphuis, P.J., Leurgans, S. et al. (2013). The
Research, 10 (7): 742–753. S-Connect study: Results from a randomized, controlled
Sabbagh, M.N., Agro, A., Bell, J. et al. (2011). trial of Souvenaid in mild-to-moderate Alzheimer’s dis-
PF-04494700, an oral inhibitor of receptor for ease. Alzheimer’s Research Therapy, 5 (6): 59.
advanced glycation end products (RAGE), in Alzheimer Shah, R.C., Matthews, D.C., Andrews, R.D. et al. (2014). An
disease. Alzheimer Disease and Associated Disorders, evaluation of MSDC-0160, a prototype mTOT modulat-
25 (3): 206–212. ing insulin sensitizer, in patients with mild Alzheimer’s
Salloway, S., Mintzer, J., Weiner, M.F. and Cummings, J.L. disease. Current Alzheimer Research, 11 (6): 564–573.
(2008). Disease-modifying therapies in Alzheimer’s Shinno, H., Inagaki, T., Miyaoka, T. et al. (2007). A
disease. Alzheimer’s and Dementia: The Journal of the decrease in N-acetylaspartate and an increase in myo-
Alzheimer’s Association, 4 (2): 65–79. inositol in the anterior cingulate gyrus are associated
Drug treatments in development for Alzheimer’s disease 585

with behavioral and psychological symptoms in transition-therapeutics-announces-results-from-


Alzheimer’s disease. Journal of the Neurological elnd005-clinical-study-in-adults-with-down-syn-
Sciences, 260 (1–2): 132–138. drome-283312281.html. Accessed 20 January 2015.
Silverberg, G.D., Mayo, M., Saul, T. et al. (2008). Tucker, S., Möller, C., Tegerstedt, K. et al. (2015). The
Continuous CSF drainage in AD: Results of a dou- murine version of BAN2401 (mAb158) selectively
ble-blind, randomized, placebo-controlled study. reduces amyloid-β protofibrils in brain and cerebro-
Neurology, 71 (3): 202–209. spinal fluid of tg-ArcSwe mice. Journal of Alzheimer’s
Simons, M., Schwärzler, F., Lütjohann, D. et al. (2002). Disease, 43 (2): 575–588.
Treatment with simvastatin in normocholesterolemic Tuszynski, M.H., Thal, L., Pay, M. et al. (2005). A phase
patients with Alzheimer’s disease: A 26-week random- 1 clinical trial of nerve growth factor gene therapy for
ized, placebo-controlled, double-blind trial. Annals of Alzheimer disease. Nature Medicine, 11 (5): 551–555.
Neurology, 52 (3): 346–350. Upton, N., Chuang, T.T., Hunter, A.J. and Virley, D.J.
Sjögren, M., Gustafsson, K., Syversen, S. et al. (2003). (2008). 5-HT6 receptor antagonists as novel cog-
Treatment with simvastatin in patients with Alzheimer’s nitive enhancing agents for Alzheimer’s disease.
disease lowers both alpha- and beta-cleaved amyloid Neurotherapeutics, 5 (3): 458–469.
precursor protein. Dementia and Geriatric Cognitive U.S. Food and Drug Administration (FDA) (2016).
Disorders, 16 (1): 25–30. Nutrition; Medical Foods – Draft Guidance for
Sparks, D.L., Sabbagh, M.N., Connor, D.J. et al. (2005). Industry: Frequently Asked Questions about
Atorvastatin for the treatment of mild to moderate Medical Foods; Second Edition (Website) Available
Alzheimer disease: Preliminary results. Archives of at www.fda.gov/Food/GuidanceRegulation/
Neurology, 62 (5): 753–757. GuidanceDocumentsRegulatoryInformation/
Sperling, R.A., Aisen, P.S., Beckett, L.A. et al. (2011). MedicalFoods/ucm054048.htm. Accessed 14 June 2016.
Toward defining the preclinical stages of Vassar, R., Kovacs, D.M., Yan, R. and Wong, P.C. (2009).
Alzheimer’s disease: Recommendations from The beta-secretase enzyme BACE in health and
the National Institute on Aging-Alzheimer’s Alzheimer’s disease: Regulation, cell biology, func-
Association workgroups on diagnostic guidelines tion, and therapeutic potential. The Journal of
for Alzheimer’s disease. Alzheimer’s and Dementia: Neuroscience, 29 (41): 12787–12794.
The Journal of the Alzheimer’s Association, Vellas, B., Black, R., Thal, L.J. et al. (2009). Long-term fol-
7 (3): 280–292. low-up of patients immunized with AN1792: Reduced
Summers, W.K., Majovski, L.V., Marsh, G.M. et al. (1986). functional decline in antibody responders. Current
Oral tetrahydroaminoacridine in long-term treatment Alzheimer Research, 6 (2): 144–151.
of senile dementia, Alzheimer type. New England Vellas, B., Sol, O., Snyder, P.J. et al. (2011). EHT0202 in
Journal of Medicine, 315 (20): 1241–1245. Alzheimer’s disease: A 3-month, randomized, placebo-
Tai, L.M., Koster, K.P., Luo, J. et al. (2014). Amyloid-β controlled, double-blind study. Current Alzheimer
pathology and APOE genotype modulate retinoid Research, 8 (2): 203–212.
X receptor agonist activity in vivo. Journal of Biological Veeraraghavalu, K., Zhang, C., Miller, S. et al. (2013).
Chemistry, 289 (44): 30538–30555. Comment on ApoE-directed therapeutics rapidly clear
Takamura, Y., Ono, K., Matsumoto, J. et al. (2014). Effects β-amyloid and reverse deficits in AD mouse models.
of the neurotrophic agent T-817MA on oligomeric Science, 340 (6135): 924.
amyloid-β-induced deficits in long-term potentiation Vellas, B., Coley, N., Ousset, P.-J. et al. (2012). Long-
in the hippocampal CA1 subfield. Neurobiology of term use of standardised Ginkgo biloba extract for
Aging, 35 (3): 532–536. the prevention of Alzheimer’s disease (GuidAge):
Theunis, C., Crespo-Biel, N., Gafner, V. et al. (2013). A randomised placebo-controlled trial. The Lancet
Efficacy and safety of a liposome-based vaccine Neurology, 11 (10): 851–859.
against protein Tau, assessed in tau.P301L mice that Villemagne, V.L., Burnham, S., Bourgeat, P. et al. (2013).
model tauopathy. PLoS One, 8 (8): e72301. Amyloid β deposition, neurodegeneration, and cognitive
Tobinick, E.L. and Gross, H. (2008). Rapid cogni- decline in sporadic Alzheimer’s disease: A prospective
tive improvement in Alzheimer’s disease following cohort study. The Lancet Neurology, 12 (4): 357–367.
perispinal etanercept administration. Journal of Vohora, D. and Bhowmik, M. (2012). Histamine H3 recep-
Neuroinflammation, 5: 2. tor antagonists/inverse agonists on cognitive and
Tolosa, E., Litvan, I., Höglinger, G.U. et al. (2014). A phase motor processes: Relevance to Alzheimer’s disease,
2 trial of the GSK-3 inhibitor tideglusib in progres- ADHD, schizophrenia, and drug abuse. Frontiers in
sive supranuclear palsy. Movement Disorders, 29 (4): Systems Neuroscience, 6: 72.
470–478. Wade, A.G., Farmer, M., Harari, G. et al. (2014). Add-on
Transition Therapeutics (2014). Transition Therapeutics prolonged-release melatonin for cognitive function
Announces Results from ELND005 Clinical and sleep in mild to moderate Alzheimer’s disease:
Study in Adults with Down Syndrome (website). A 6-month, randomized, placebo-controlled, multi-
Available at www.prnewswire.com/news-releases/ center trial. Clinical Interventions in Aging, 9: 947–961.
586 Dementia

Wang, J., Ono, K., Dickstein, D.L. et al. (2011). Carvedilol as Winblad, B., Giacobini, E., Frölich, L. et al. (2010).
a potential novel agent for the treatment of Alzheimer’s Phenserine efficacy in Alzheimer’s disease. Journal of
disease. Neurobiology of Aging, 32 (12): 2321. Alzheimer’s Disease, 22 (4): 1201–1208.
Warren, J.D., Rohrer, J.D. and Rossor, M.N. (2013). Winblad, B., Graf, A., Riviere, M.-E. et al. (2014). Active
Frontotemporal dementia. BMJ, 347: f4827–f4827. immunotherapy options for Alzheimer’s disease.
Weinreb, O., Amit, T., Bar-Am, O. and Youdim, M.B.H. Alzheimers Research Therapy, 6 (1): 7.
(2012). Ladostigil: A novel multimodal ­neuroprotective Wischik, C.M., Staff, R.T., Wischik, D.J. et al. (2014). Tau
drug with cholinesterase and brain-­selective mono- aggregation inhibitor therapy: An exploratory phase 2
amine oxidase inhibitory activities for Alzheimer’s study in mild or moderate Alzheimer’s disease. Journal
disease treatment. Current Drug Targets, 13 (4): of Alzheimer’s Disease, 44 (2): 705–720.
483–494. Woodward, M. (2007). Anti-inflammatory Drugs.
Weggen, S., Eriksen, J.L., Sagi, S.A. et al. (2003). Evidence Therapeutic Strategies in Dementia. Oxford, United
that nonsteroidal anti-inflammatory drugs decrease Kingdom: Atlas Medical Publishing, pp. 111–119.
amyloid beta 42 production by direct modulation Yang, G., Wang, Y., Tian, J. and Liu, J.-P. (2013). Huperzine
of gamma-secretase activity. Journal of Biological A for Alzheimer’s disease: A systematic review and
Chemistry, 278 (34): 31831–31837. meta-analysis of randomized clinical trials. PLoS One,
Wiessner, C., Wiederhold, K.-H., Tissot, A.C. et al. (2011). 8 (9): e74916.
The second-generation active Aβ immunotherapy Yarchoan, M. and Arnold, S.E. (2014). Repurposing dia-
CAD106 reduces amyloid accumulation in APP trans- betes drugs for brain insulin resistance in Alzheimer
genic mice while minimizing potential side effects. The disease. Diabetes, 63 (7): 2253–2261.
Journal of Neuroscience, 31 (25): 9323–9331. Yun, H.-M., Kim, H.S., Park, K.-R. et al. (2013). Placenta-
Wilcock, G.K., Black, S.E., Hendrix, S.B. et al. (2008). derived mesenchymal stem cells improve memory
Efficacy and safety of tarenflurbil in mild to moderate dysfunction in an Aβ1-42-infused mouse model of
Alzheimer’s disease: A randomised phase II trial. The Alzheimer’s disease. Cell Death and Disease,
Lancet Neurology, 7 (6): 483–493. 4 (12): e958.
Winblad, B., Andreasen, N., Minthon, L. et al. (2012). Zhang, L.-F., Zhou, Z.-W., Wang, Z.-H. et al. (2015). Coffee
Safety, tolerability, and antibody response of active and caffeine potentiate the antiamyloidogenic activ-
Aβ immunotherapy with CAD106 in patients with ity of melatonin via inhibition of Aβ oligomerization
Alzheimer’s disease: Randomised, double-blind, and modulation of the Tau-mediated pathway in N2a/
placebo-controlled, first-in-human study. The Lancet APP cells. Drug Design, Development and Therapy,
Neurology, 11 (7): 597–604. 9: 241–272.
(a) (b)

(c) (d)

Figure 11.1 Diffusion tensor imaging provides quantitative measures of (a) apparent diffusion coefficient (ADC) and
(b) ­fractional anisotropy (FA). Brighter signal means more average diffusion and more constrained diffusion for ADC and FA,
respectively. Note that grey and white matter do not differ in ADC but have very different FA values, such that FA provides
a map of the myelinated axons of white matter. By calculating the tensor information, the direction of diffusion can be
­determined in each voxel (c), from which tracts can be reconstructed (d). Panel (c) shows the principal diffusion direction in
the region including the genu of the corpus callosum depicted by the red box in panel (b). In panel (d), a tract of the forceps
minor (yellow) has been reconstructed by placing a single ‘seed’ voxel (blue) in the genu of the corpus callosum.
Figure 12.2 Typical pattern of 11C-PiB binding in AD. Representative sagittal and transaxial [11C]PiB PET images over-
layed on their respective MRI in a healthy elderly control subject (HC) (left) compared with an Alzheimer’s disease (AD)
patient (right). 11C-PiB retention is observed in frontal, temporal and parietal cortices as well as in the posterior cingulate/
precuneus areas, with relative sparing of occipital and sensorimotor cortex.
Figure 12.3 Aβ imaging in Alzheimer’s disease. Representative parametric sagittal, transaxial and coronal PET images
of Alzheimer’s disease (AD) patients assessed with either [11C]PiB, [18F]florbetaben, [18F]flutemetamol, [18F]florbetapir or
[18F]NAV4694. All AD patients harboured high Aβ burdens in the brain, reflected in marked radiotracer retention in
cortical and subcortical grey matter areas. [18F]Florbetapir (Amyvid®); [18F]flutemetamol (Vizamyl®) and [18F]florbetaben
(Neuraceq®) have already been approved for clinical use by the FDA and EMA.
PiB- HC

Misfolded
proteins PiB- MCI

PD

α-syn

PiB- DLB

PiB+ HC

PiB+ MCI

Aβ PiB+ DLB

AD

tau

FTLD

PrPsc spCJD

Figure 12.5 Aβ imaging in ageing and dementia. Representative 40–70 min post-injection transaxial 11C-PiB images along
the spectrum of some of the misfolded proteins associated with neurodegenerative diseases, encompassing the different
patterns of 11C-PiB retention in the brain, with – from top to bottom – a 73-year-old healthy PiB-negative control (PiB-HC)
subject (mini mental state examination [MMSE] 30), 83-year-old PiB-negative subject with mild cognitive impairment (PiB-
MCI) (MMSE 28), 61-year-old PiB-negative Parkinson’s disease (PD) patient (MMSE 27), 69-year-old PiB-negative patient
with dementia with Lewy Bodies (DLB; MMSE 24), 77-year-old PiB-positive healthy control (PiB+ HC) subject (MMSE 28),
82-year-old PiB-positive subject with mild cognitive impairment (PiB+ MCI) (MMSE 28), 78-year-old PiB-positive DLB
patient (PiB+ DLB) (MMSE 19), 76-year-old Alzheimer’s disease (AD) patient (MMSE 21), 59-year-old patient with fronto-
temporal lobe degeneration (FTLD; MMSE 20) and 59-year-old PiB-negative patient with confirmed sporadic Creutzfeldt-
Jakob disease (spCJD). PET images show clear differences when comparing cortical PiB retention in PiB-negative HC,
MCI, PD, spCJD and FTLD with PiB-positive HC, MCI, DLB or AD patients. Only non-specific PiB binding in white matter
is observed in PiB-negative HC, MCI, PD, spCJD and FTLD compared with PiB binding in cortical areas of AD and DLB
patients. All images are scaled to the same standardized uptake value ratio (SUVR) maximum.
HC AD

18F-flutemetamol 18F-flutemetamol
L R R L L R R L

PET-SUVRpons PET-SUVRpons
1.2 1.2
0.9 0.9
RT.LAT LT.LAT SUP INF RT.LAT LT.LAT SUP INF
0.7 0.7
0.4 0.4

RT.MED LT.MED POST ANT RT.MED LT.MED POST ANT

18F-THK5117 18F-THK5117
L R R L L R R L

PET-SUVRCbCtx PET-SUVRCbCtx
2.0 2.0
1.6 1.6
RT.LAT LT.LAT SUP INF RT.LAT LT.LAT SUP INF
1.2 1.2
0.8 0.8

RT.MED LT.MED POST ANT RT.MED LT.MED POST ANT

Figure 12.6 Aβ and tau imaging in ageing and dementia. Surface display PET images of the respective Aβ ([18F]flute-
metamol, top row) and tau ([18F]THK5117, bottom row) burden in an age-matched healthy control (HC) and an Alzheimer’s
disease (AD) patient.
Aβ production
Preclinical
Aβ aggregation
Aβ clearance (immunotherapy) ACI-24
Tau aggregation or phosphorylation Phenylthiazolylhydrazide
Cholinergic drugs
Anti-Aβ antibodies Phase 1
Others Rhodanines

RS-0406 Minocycline
Affitope AD-01*

Phase 2
GSK-933776 AL-208*
Affitope AD-02
SEN-1269 AL-108* Anti-tau
PF-04360365
MABT-5102A antibodies
ACC-001 MTC/LMT*
AN-1792* Phase 3 Nicotinamide
UB-311 IVIg
CAD-106 Valproate* N-031112
SP-233 Bapineuzumab (NP-12)* N-744
ELND-005
Solanezumab Phase 4 Lithium*
R-1450 NGF
PBT1* Tramiprosate* Donepezil† Antioxidants

Rivastigmine† SB-742457
Peptidic anti- EGCg
V-950 PBT2* Statins
aggregants Galantamine† PRX-03140*
NIC5-15 Tarenflurbil* Memantine† BDNF
Latrepirdine PUFAs*
E-2012*
Semagacestat TTP-488
Bryostatin-1 Phenserine*
Exebryl-1 Rosiglitazone* PF-04447943
MK-0752* EHT-0202*
EVP-6124
BMS-708163 Huperzine-A* Ladostigil
CHF-5074
AF-102B*
NGXsc Pioglitazone* AZD-3480*
CTS-21166* ABT-089* Talsaclidine* AF-267B

PF-3084014* M-30
GSM-1
Begacestat* Memoquin
Anti-β-secretase antibodies

BBS1

Figure 55.3 Drug development in Alzheimer’s disease: drugs being investigated for Alzheimer’s disease therapy,
reported according to the most advanced phase of study and main therapeutic properties (including data from studies
in vitro and animal models). Aβ, amyloid-β; BBS1, anti-β-site antibodies; BDNF, brain-derived neurotrophic factor; EGCg,
­epigallocatechin-3-gallate; IVIg, intravenous immunoglobulin; LMT, leuco-methylthioninium; MTC, methylthioninium chlo-
ride; NGF, nerve growth factor; NGXsc, NGX series compounds; PUFAs, polyunsaturated fatty acids; GSM, γ-secretase
modulator; RCT, randomized controlled trial. *RCTs in Alzheimer’s disease not ongoing. †Drugs approved for the treat-
ment of Alzheimer’s disease. (Reprinted from The Lancet Neurology, 9, Mangialasche, F. et al., Alzheimer’s disease: Clinical
trials and drug development, 702–716, 2010, with permission from Elsevier.)
Odds ratio of dementia
< 0.67 300
0.67–0.83 Miles
0.83–0.91
0.91–0.95
0.95–1.05
1.05–1.1
1.1–1.2
1.2–1.5
> 1.5

North

(a) (b)

Figure 55.7 Age-adjusted odds ratio of dementia in male (a) and female (b) Swedish twins with twin-level random effects
(likely to capture genetic and shared environmental variance) removed. (From Russ, T.C. et al., Epidemiology, 26, 263–270,
2015a. With permission.)
Odds ratio of dementia
< 0.67
0.67–0.83
0.83–0.91
0.91–0.95
0.95–1.05
1.05–1.1
1.1–1.2
1.2–1.5
>1.5

(a) (b)

100
Miles

North

(c) (d)

Figure 55.8 Age-adjusted odds ratio of dementia in the SMS1932 cohort: childhood – males (a), females (b); adulthood –
males (c), females (d). (From Russ, T.C. et al., Epidemiology, 26, 263–270, 2015a. With permission.)
Newcastle categorization of the major CV lesions
associated with cognitive impairment
I II III IV V VI

Large infarct or Multiple small or Strategic Cerebral Cerebral CV lesions


several infarcts microinfarcts infarcts hypoperfusion haemorrhage with AD pathology

Multi-infarct White matter Thalamus Hippocampal Lobar Mixed


dementia lesions hippocampus sclerosis ICH dementia
basal forebrain SAH

Figure 59.2 Schematic diagram of different cerebrovascular pathologies associated with dementia. The proposed
Newcastle categorization includes six subtypes (Kalaria et al., 2004). In all of the subtypes, the age of the vascular
lesion(s) should correspond with the time when disease began. The post-stroke cases are usually included in subtypes
I–III. While these may not be different from other published subtypes (Jellinger, 2008), they are practical and simple
to use. Cases with extensive white matter (WM) disease in the absence of significant other features are included under
SVD. Subtype I may result from large vessel occlusion (atherothromboembolism), artery-to-artery embolism or car-
dioembolism. Subtype II usually involves descriptions of arteriosclerosis, lipohyalinosis, hypertensive, arteriosclerotic,
amyloid or collagen angiopathy. Subtypes I–II and V may result from aneurysms, arterial dissections, arteriovenous
malformations and various forms of arteritis (vasculitis). CAA, cerebral amyloid angiopathy; ICH, intracerebral haemor-
rhage; SAH, subarachnoid haemorrhage. (Original diagram courtesy of Dr K Nagata, Akita, Japan.)
(a) (b)

(c) (d)

(e) (f )

Figure 59.4 Pathological features associated with SVD in VaD. Panels show examples of lacunes, small infarcts and micro-
infarcts: (a) typical cavitated lacunar lesions (arrow) in the putamen of a 65-year-old man. (b) Multiple infarcts in the basal
­ganglias of an 80-year-old man with vascular and neurofibrillary pathology. (c)–(e) Cerebral microvessels with variable
hyalinosis, perivascular rarefaction, microinfarcts and perivascular spaces in three different cases. Moderate gliosis in the
­surrounding region is also evident in case in (c); (f) small cortical infarct in a VaD case with severe CAA. A ­penetrating
­arteriole with CAA is seen in the middle of the infarct (arrow). Magnification bar: (a) = 1 cm, (b), (f) = 500 μm; (c), (d) = 100 μm,
(e) = 200 μm.
(a) (b)

(c) (d)

Figure 59.5 WM lesions visualized by conventional histopathological staining in an 85-year-old man diagnosed with vascular
encephalopathy. (a) >75% stenosis in the internal carotid artery 8 mm above the bifurcation. The narrowed lumen (arrow) is
seen. (b) WM rarefaction and myelin loss in the medial temporal lobe but sparing of U fibres; (c), post-mortem T2W magnetic
resonance image of a formalin-fixed block from the parietal lobe. Area of hypersignal can be seen in the WM (*); (d), H&E
stained section from the block in (c) showingsevere deep WM ­pallor in the area of hypertensity (*). A small cortical infract is
also seen (arrow). Magnification bar: (a) = 500 mm, (b) = 400 μm, (c) = 1 cm, (d) = 500 μm.
(a) (b) (d) (e)

50 μm 50 μm 20 μm

(c) (f )

50 μm 20 μm

100 μm 50 μm

Figure 62.1 Lewy bodies and Lewy pathologies in Parkinson’s disease. (a) and (b) Haematoxylin and eosin stained classic
Lewy bodies in pigmented dopaminergic neurones of the substantia nigra showing the typical core and halo structure.
Scale in (b) is equivalent for (a). (c)–(f). α-Synuclein immunoreactive pathologies showing (c) and (e) mature Lewy bodies,
(c) and (d) pale bodies and neuronal aggregates and (f) astrocytes containing aggregates.
Figure 65.2 Fluorodeoxyglucose positron emission tomography (FDG-PET) scan from a patient with FTD whose structural
imaging did not show convincing evidence of disproportionate frontal atrophy. Note the clear evidence of asymmetric
frontal hypometabolism (arrows).
(a)

(b)

(c)

Figure 65.3 Use of electroencephalography (EEG) in the investigation of suspected FTD. This 63-year-old male presented
with marked changes in personality and comportment, typical of those seen in FTD, that had evolved over the preceding
8 months. FDG-PET, shown as a parametric map of hypometabolism (a), revealed a marked prefrontal hypometabolism.
His EEG was grossly abnormal with a considerable delta-wave activity (b). This is not compatible with FTD. His diagnosis
was presumed to be an autoimmune encephalitis, and after treatment with high-dose methylprednisolone both his FTD-like
behavior and EEG (c) improved dramatically.
Primary progressive aphasia (PPA)

Sporadic PPA Familial PPA

GRN-associated C9orf72-associated
svPPA nfvPPA IvPPA
aphasia aphasia

Pathological subtypes

FTLD-TDP FTLD-TDP FTLD-TDP FTLD-tau FTLD-tau FTLD-tau Alzheimer


Type A Type B Type C CBD/PSP Pick’s GGT pathology

Figure 69.1 Clinico-pathological and clinico-genetic associations in primary progressive aphasia (PPA). PPA as a syndrome
has heterogeneous genetic and pathological associations. However, the importance of subtyping PPA is indicated by the
third row of boxes, which show in a schematic manner the pathological associations with semantic variant PPA (svPPA),
nonfluent variant PPA (nfvPPA), logopenic PPA (lvPPA) and with the familial progranulin (GRN)-associated or C9orf72-
associated form of PPA, where one pathological subtype tends to dominate. Each of the pathological subtypes are indi-
cated by a separate coloured box: frontotemporal lobar degeneration – TAR DNA binding protein (FTLD-TDP) types A to
C, FTLD-tau (CBD, corticobasal degeneration; PSP, progressive supranuclear palsy; GGT, globular glial tauopathy; and
Pick’s disease) and Alzheimer pathology.
Figure 69.2 Regional percentage differences in cortical thickness in posterior cortical atrophy (PCA) (N = 48) compared
with typical amnestic Alzheimer’s disease (tAD; N = 30) for the left and right hemispheres. The colour scale represents
magnitude of cortical thickness difference. Red and yellow (positive values) represent lower cortical ­thickness in PCA
subjects compared with tAD, whereas dark to light blue (negative values) represents greater cortical thickness. (From
Lehmann, M., et al., Cereb. Cortex, 21, 2122–2132, 2011a.)
54
Vitamins and complementary therapies used to
treat Alzheimer’s disease

DENNIS CHANG, GENEVIEVE Z. STEINER, DILIP GHOSH AND ALAN BENSOUSSAN

54.1 INTRODUCTION 54.2 HERBAL MEDICINE FOR AD

Alzheimer’s disease (AD) is the most common form of Herbal medicines have been used widely across many cul-
dementia, a leading cause of mental and physical disabil- tures to improve memory function and manage behav-
ity in elderly people. Current pharmaceutical manage- ioural and psychological symptoms. Specific herbs that
ment of AD offers only modest short-term symptomatic have attracted most attention include Ginkgo biloba,
relief and does not satisfactorily consider the magnitude Bacopa monnieri, Huperzia serrata, Panax ginseng,
of the disease burden on patients and the community as Curcuma longa, Melissa officinalis and Camellia sinensis.
a whole. Acetylcholinesterase (AChE) inhibitors or gluta- The mechanisms of action, adverse effects and herb–drug
mate receptor antagonists are typically used to manage the interactions of these herbal medicines for AD are summa-
behavioural and psychological symptoms of AD, but there rized in Table 54.2.
is limited evidence on their efficacy for patients beyond the Unlike pharmaceutical agents, herbal medicines often
first 6–12 months (Madhusoodanan and Ting, 2014) (see contain multiple bioactive components that can affect
Chapter 52). several therapeutic targets for diseases with multifacto-
Complementary and alternative medicine (CAM) is used rial/multisystem pathophysiological components, such as
extensively worldwide, with a national survey conducted by dementia. In addition, the complex chemical mixtures of
the National Center for Complementary and Alternative herbal medicines enhance therapeutic efficacy by facili-
Medicine (NCCAM) estimating approximately 33.2% of tating synergistic action (e.g. constituents interacting to
adults and 11.6% of children in the United States having improve their solubility and hence bioavailability) (Yang
used or use some form of CAM (Black et al., 2015; Clarke et et al., 2014).
al., 2015). Worldwide prevalence of CAM use is estimated to
be up to 74.8% (Frass et al., 2012). 54.2.1 GINKGO BILOBA
Treatment of ageing-related disorders with CAM dates
back to 5000 years in ancient China, where herbal remedies Ginkgo biloba leaf extract (from the ginkgo tree in China) is
were used to boost memory function and increase longevity. one of the most studied herbs. The principal active compo-
The common forms of CAM for AD include herbal medicine nents in ginkgo are flavonol glycosides (e.g. quercetin and
(e.g. Western herbal medicine, Chinese herbal medicine, kaempferol) and terpenoids (e.g. ginkgolide and bilobalide).
Ayurvedic medicine), vitamins and dietary supplements Preclinical studies demonstrate that ginkgo can decrease
(e.g. fish oil), acupuncture, aromatherapy, mind–body ther- oxygen radical discharge and pro-inflammatory functions of
apy (e.g. meditation, tai chi, yoga), physical therapies (e.g. macrophages (antioxidant and anti-inflammatory), reduce
chiropractic, reflexology, remedial massage), music therapy corticosteroid production (anti-anxiety), increase glucose
and combined and light therapy (Table 54.1). uptake and utilization and ATP production, improve blood
This chapter focuses on the scientific evidence support- flow by increasing red blood cell deformability, decrease red
ing the use of herbal medicine, vitamins and fish oil in the cell aggregation, induce nitric oxide production and inhibit
management of AD. platelet activating factor receptors (Chan et al., 2007). In

587
588 Dementia

Table 54.1 Overview of common vitamins and memory loss and mental impairment. Besides HupA’s
complementary therapies for the treatment of Alzheimer’s well-known AChE inhibiting property, other pharmaco-
disease logical effects include antioxidant activity, anti-β-amyloid
Herbal medicine Ginkgo biloba peptide fragmentation, inhibition of oxygen-glucose
deprivation and N-methyl d-aspartate (NMDA) receptor
Huperzia serrata (Huperzine A)
antagonism (Howes and Houghton, 2003) (Table 54.2).
Bacopa monnieri
Recent meta-analyses suggest that HupA can improve
Curcuma longa
cognition and behaviour in patients with AD (Yang et
Panax ginseng al., 2013; Xing et al., 2014), in particular, HupA improved
Melissa officinalis cognitive function and activities of daily living, com-
Camellia sinensis (tea) pared to placebo (Yang et al., 2013); quality of life was
Vitamins and Vitamin C and E not evaluated in these studies. While these findings are
dietary Vitamin B and folic acid positive and consistent with earlier reports (Wang et al.,
supplements Vitamin D 2009), they should be interpreted with caution due to the
Fish oil poor methodological quality, small sample sizes and short
Souvenaid® intervention periods of the included trials. More rigorous
Other Acupuncture randomized controlled trials (RCTs) are needed to con-
complementary Aromatherapy firm the clinical effectiveness of HupA.
therapies Physical therapy (chiropractic,
reflexology, remedial massage, 54.2.3 BACOPA MONNIERI
therapeutic touch)
Mind–body therapy (meditation, Bacopa monnieri (Brahmi) is an Indian Ayurvedic medi-
tai chi, yoga, qigong) cine used for the management of a range of psychological
Music therapy conditions (e.g. anxiety, attention) (Russo and Borrelli,
Combined melatonin and light 2005). Brahmi is marketed in western countries as a mem-
therapy ory enhancing agent, with effects attributable to two major
constituents: saponins, bacosides A and B. Brahmi has been
shown to have neuroprotective effects and can enhance
healthy young adults, ginkgo has been shown to improve cognition in animal dementia models (Saini et al., 2012).
speed of processing, working memory and executive func- The exact mechanisms for these actions remain unclear;
tion (Kennedy et al., 2007). however, it has been suggested that neuropharmacological/­
Despite recent evidence, the efficacy of ginkgo as nootropic actions of Brahmi are induced by antioxidant
a cognitive enhancer for dementia remains controver- effects via metal chelation at the initiation level and also
sial. This is primarily because early clinical trials dem- as a lipid peroxidation chain action breaker and modula-
onstrated inconsistent effects on memory, attention and tion of cholinergic system via reversing acetylcholine (ACh)
anxiety (for a review see Birks and Evans, 2009); however, depletion, reducing choline acetylase activity and decreas-
the use of self-report questionnaires in these early studies ing muscarinic receptor binding (Stough et al., 2008; Aguiar
raises concerns about the reliability of results (Howes and
and Borowski, 2013).
Houghton, 2003). Three recently published meta-analyses
(Gauthier and Schlaefke, 2012; Yang et al., 2014; Tan et al., Human trials suggest that Brahmi can enhance cogni-
2015) indicate that treatment with EGb761® (a standardized tion in older adults, but there is limited evidence for its
ginkgo extract preparation) stabilized or reduced the decline effectiveness in AD. Two double-blind placebo-controlled
process in cognition, function and behaviour, with limited studies examining the acute effects (320 and 640 mg) of
adverse effects in patients with cognitive impairment and Brahmi (KeenMind ® – CDRI 08) demonstrated improved
dementia, compared to placebo. Importantly, one review cognition, compared to placebo (Downey et al., 2013;
found these effects were most notable in patients with neu- Benson et al., 2014). A 12-week study on 98 older adults
ropsychiatric symptoms (Tan et al., 2015). However, a 5-year showed improved verbal learning, memory acquisition and
randomized, double-blind, placebo-controlled trial of EGb retention with 300 mg/day of Brahmi, compared to placebo
761 did not show any reduction in risk of progression to AD (Morgan and Stevens, 2010). Two systematic reviews also
for adults with memory complaints (Vellas et al., 2012).
concluded that Brahmi can improve attention and memory
(Pase et al., 2012; Kongkeaw et al., 2014); however, inconsis-
54.2.2 HUPERZIA SERRATA tent measures used by the included trials make it difficult
to ascertain whether Brahmi can enhance other aspects of
Huperzine A (HupA) is an alkaloid derived from the club cognition. For instance, another 12-week trial on 72 adults
moss, Huperzia serrata. It is a licensed anti-AD drug in (aged 35–60 years) found that Brahmi (450 mg/day) did
China (hence, most clinical trials of HupA have been not affect cognition, but did show a trend towards decreas-
conducted and published in China) and is sold over the ing anxiety, compared to placebo (Sathyanarayanan et al.,
counter as a dietary supplement in the United States for 2013). Further studies are required to ascertain the effective
Vitamins and complementary therapies used to treat Alzheimer’s disease 589

Table 54.2 Mechanisms of action, adverse effects and herb–drug interactions of commonly used herbs for Alzheimer’s
disease

Herb Possible mechanisms of action Adverse effects Herb–drug interaction


Ginkgo Decrease oxygen radical discharge Dizziness, tinnitus, Possess antiplatelet activity,
biloba and pro-inflammatory functions of headache, nausea, caution when used with
macrophages, reduce corticosteroid vomiting, diarrhoea, anticoagulant and
production, increase glucose uptake headache, weakness, antiplatelet agents (such as
and utilization and ATP production, restlessness and skin rash, warfarin)
improve blood flow by increasing etc.
red blood cell deformability,
decrease red cell aggregation,
induce nitric oxide production,
inhibit platelet activating factors
Huperzia Anti-cholinesterase, anti-oxidant, Nausea, vomiting, anorexia, May cause additive effects
serrata anti-β-amyloid peptide fragment, diarrhoea, hyperactivity, when used with other
(Huperzine A) inhibition of oxygen-glucose insomnia, indigestion and anticholinesterase agents
deprivation, muscaric receptor mild abdominal pain such as donepezil
antagonism
Bacopa Enhance nerve impulse transmission, Therapeutic doses are not Slightly sedative effect; caution
monnieri improve memory and cognition associated with major when used with other known
adverse effects sedatives, inhibits the effect
of thyroid-suppressant drugs.
Curcuma Antioxidant, anti-inflammatory, No significant side effects Possess anticoagulant or
longa cholesterol-lowering properties, reported antiplatelet activity, caution
block aggregation and fibril when used with anticoagulant
formation and antiplatelet medications
Panax Stimulating central cholinergic and Insomnia, diarrhoea, Combination of
ginseng dopaminergic receptors, stimulating headache, tremor and antidepressants and ginseng
hypothalamic–pituitary–adrenal axis skin eruptions may induce mania, caution
when used with anti-diabetic
drugs as ginseng may reduce
fasting blood glucose levels
Melissa Nicotinic and muscarinic binding No significant side effects, Caution when used together
officinalis properties, strong antioxidant effect may cause drowsiness with sedatives, anti-anxiety
drugs, narcotic pain relievers
(e.g. codeine), antihistamines
Camellia The phenolic compounds (especially No significant side effects, Caution when used with
sinensis epigallocatechin gallate [EGCG]) excessive caffeine may theophylline and ephedrine
(tea) have antioxidant effects lead to agitation, tremors
and insomnia

dosage range, the effects of long-term administration and allergy, liver disorders, anorexia, rheumatism, diabetic
potential therapeutic effects of Brahmi for AD. wounds, cough and sinusitis) and neurodegenerative dis-
orders (Goel et al., 2008). Data from animal and in vitro
54.2.4 CURCUMA LONGA studies suggest that curcuminoids possess antioxidant,
anti-inflammatory and cholesterol-lowering properties, all
The perennial herb Curcuma longa (turmeric) contains three of which are key processes associated with the pathogenesis
structurally closely-related chemical components – c­ urcumin, of AD (Ringman et al., 2005). Furthermore, curcuminoids
demethoxycurcumin and bisdemethoxycurcumin – which directly bind to small β-amyloid species to block aggrega-
together are commonly referred to as ‘curcumin’ or ‘curcumi- tion and fibril formation supporting the rationale for cur-
noids’ (Goel et al., 2008). Commercially available ‘curcumin’ cuminoids to be used therapeutically for AD (Yang et al.,
is widely promoted for its anti-inflammatory properties 2005). However, evidence from rigorous AD studies is lack-
(Jurenka, 2009). ing, with only two double-blind RCTs conducted (Baum
In Ayurvedic medicine, turmeric is a well-documented et al., 2008; Ringman et al., 2005), both of which showed
treatment for many diseases (e.g. asthma, respiratory no difference in cognition between placebo and curcumin
590 Dementia

treatment. The discrepancy between in vitro and in vivo administered to young, healthy volunteers (Kennedy et al.,
findings and human trials may be due to the low bioavail- 2003b). In mild-to-moderate AD patients, 16 weeks of treat-
ability of curcumin as a result of low absorption and rapid ment with lemon balm extract significantly improved cog-
hepatic and intestinal metabolization (Brondino et al., nitive function and reduced agitation, compared to placebo
2014). Novel formulations have attempted to remedy this, (Akhondzadeh et al., 2003). However, a more recent double-
for instance, a recent double-blind RCT using a solid lipid blind RCT that compared lemon balm oil aromatherapy
curcumin formulation (Longvida®) in healthy older adults to donepezil and placebo in 81 patients with probable or
demonstrated improved sustained attention and working possible AD found no difference between placebo and the
memory with acute administration (1 hour and 3 hours) two interventions in reducing agitation (Burns et al., 2011).
and improved working memory, mood enhancement and Interestingly, an 18%–37% improvement in agitation and
lowered total and low-density lipoprotein (LDL) cholesterol neuropsychiatric symptoms was noted in all treatment
with chronic administration (4 weeks) (Cox et al., 2014). groups suggesting that touch and interaction may be help-
Several large scale clinical trials are underway or soon to ful for treating agitation in AD patients. More experiments
be completed and other new formulas aimed at increas- are required to establish whether lemon balm is of value in
ing the bioavailability of curcumin are being developed. the management of agitation and cognitive decline in AD.
The upcoming results will help to determine if curcumin
has therapeutic value for the treatment and prevention of 54.2.7 CAMELLIA SINENSIS (TEA)
dementia.
Data from animal and epidemiological studies suggests
54.2.5 PANAX GINSENG that drinking tea (mostly green tea) may help to protect
the brain against ageing. An inverse correlation between
Panax ginseng (ginseng) root has been used to manage AD tea consumption and the incidence of AD and other neu-
in many Asian countries. The principal bioactive compo- rodegenerative diseases (e.g. Parkinson’s disease) has been
nents of ginseng are ginsenosides, which have antioxidant, reported (Sharangi, 2009). The chief bioactive components
anti-inflammatory and anti-apoptotic effects (Radad et of tea are caffeine, amino acids (e.g. l-Theanine) and poly-
al., 2006). In vitro studies have demonstrated that ginsen- phenols (antioxidants) (Sharangi, 2009). In particular, its
oside Rg3 promotes β-amyloid peptide degradation through main c­atechin polyphenol constituent, epigallocatechin
enhanced gene expression (Yang et al., 2009) and ginsen- (EGCG), has demonstrated neuroprotective/neurorescue
oside retinoblastoma 1 (Rb1) can ameliorate AD pathology properties (Mandel et al., 2008) with in vitro studies evi-
by suppressing phosphorylated tau protein expression and dencing anti-amyloidogenic properties of EGCG (Hyung
upregulating brain-derived neurotrophic factor (BDNF) et al., 2013) and in vivo work in mice dementia model
(Wang et al., 2013). showing improved cognition and less β-amyloid deposition
Most of the cognitive effects of ginseng have been stud- (Hassaan et al., 2014). A recent meta-analysis of human acute
ied in animals and healthy individuals. Some data suggest studies indicates that tea increases multisensory attentional
that ginseng modestly improves thinking and working outcomes in healthy adults (Camfield et al., 2014). Further,
memory in healthy volunteers (Kennedy and Scholey, a human cross-­sectional study assessing the effect of green
2003a; Reay et al., 2006). Two small open-label trials dem- tea on cognition in elderly Japanese participants, reported
onstrated potential therapeutic benefits of ginseng for AD, that green tea consumption of two or more cups (100 mL/
with both studies evidencing improved Alzheimer’s Disease cup/day) reduced the prevalence of cognitive impairments
Assessment Scale-cognitive subscale (ADAS-cog) scores (Kuriyama et al., 2006). Further research is required to
after 12 weeks of treatment with ginseng, compared to pla- ascertain whether tea can be used to delay AD progression
cebo (Heo et al., 2008; Lee et al., 2008). Ginseng has also and improve cognition in this cohort.
demonstrated clinical benefits when combined with ginkgo
in improving cognitive function in humans (Wesnes et al.,
2000; Kennedy et al., 2001; Yakoot et al., 2013). Large-scale,
long-term studies using standardized extracts are required 54.3 FISH OILS FOR AD
to determine the clinical efficacy of ginseng therapy in AD.
A high dietary omega-6/omega-3 polyunsaturated fatty acid
54.2.6 MELISSA OFFICINALIS (PUFA) ratio has been associated with an increased risk of
dementia (Loef and Walach, 2013). While there is a growing
Melissa officinalis (from the lemon balm plant) binds to body of evidence suggesting that omega-6 PUFAs promote
both nicotinic and muscarinic ACh receptors in the central inflammation and disease processes (Lawrence, 2013), it has
nervous system (Perry et al., 1999; Kennedy et al., 2003b) been suggested that the neuroprotective effects of omega-3
and has strong antioxidant properties (Dastmalchi et al., PUFAs are attributable to reduced cardiovascular risk fac-
2008); both of which are desirable in the management of tors (e.g. triglyceridemia) (Schilling et al., 2014), attenuation
AD. Human studies have shown that lemon balm extract of the inflammation process and reduction of amyloid pro-
can modulate mood and cognitive performance when duction and increased clearance (Fotuhi et al., 2009).
Vitamins and complementary therapies used to treat Alzheimer’s disease 591

Data from animal and cross-sectional epidemiological adverse effects (Ames and Ritchie, 2007). For example, 800
studies have demonstrated that long-chain omega-3 PUFAs, IU/day treatment with vitamin E (c.f. placebo) for 6 months
including docosahexaenoic acid (DHA) and eicosapentae- was only effective in reducing oxidative stress in some AD
noic acid (EPA), may protect against cognitive decline and patients. For those that were not responsive, a detrimen-
dementia (Conquer et al., 2000; Heude et al., 2003; Tully tal decline in cognition was observed (Lloret et al., 2009).
et al., 2003). For example, the Framingham Heart Study However, in a recent study involving 613 patients with
showed a 47% reduction in the risk of developing all-cause mild-to-moderate AD, treatment with 2000 IU/day of vita-
dementia with higher DHA intake (levels of approximately min E resulted in slower functional decline than placebo
0.18 g/day) (Schaefer et al., 2006) and that older individuals (Dysken et al., 2014). Currently, there are no published
with high (c.f. low) red blood cell DHA levels had greater clinical trials on a vitamin C intervention for dementia, but
white matter volumes and those with high DHA and EPA it has been suggested that maintaining healthy vitamin C
levels had better visual memory, executive function and levels (rather than consuming additional supplements) can
abstract thinking abilities than those with low levels (Tan protect against cognitive decline and AD (Harrison, 2012;
et al., 2012). Harrison et al., 2014).
Although omega-3 PUFAs may play a role in pre-
venting the development of dementia and even revers- 54.4.2 B VITAMINS AND FOLATE
ing aspects of cognitive decline (such as mild cognitive
impairment [MCI]), there is limited evidence on the effi- Data from retrospective case-controlled studies and pro-
cacy of omega-3s as a treatment for patients who have spective cohort studies suggest that elevated serum total
already developed dementia (Cederholm et al., 2013). For homocysteine level is a potential risk factor for cognitive
instance, a double-blind 18 month RCT of DHA on 295 decline and dementia (Clark, 2008), with high levels associ-
patients with mild-to-moderate AD did not slow down ated with increased risk of endothelial cell injury, inflam-
the rate of cognitive and functional decline compared matory processes in blood vessels, ischemia and brain
to placebo (Quinn et al., 2010). A 24-week double-blind infarcts (Morris, 2003, 2012). Homocysteine serum lev-
RCT found that 1.8 g/day of omega-3 PUFAs improved els can be lowered by vitamins B12, B6 and folate, which
ADAS-cog scores in participants with MCI but not AD, convert homocysteine to methionine and cysteine, respec-
compared to placebo (Chiu et al., 2008). Another 24-week tively. This homocysteine hypothesis of dementia has led to
intervention did not delay the rate of cognitive decline (on considerable interest in using B vitamins and folate for the
Mini-Mental State Examination [MMSE] and ADAS-cog) treatment/prevention of AD (Clark, 2008).
compared to placebo, but positive effects were observed in Evidence for this therapeutic approach from clinical
a subgroup with very mild AD (MMSE > 27) (Freund-Levi trials has not been consistent. Several systematic reviews
et al., 2006). Findings suggest that a diet rich in omega-3 (Balk et al., 2007; Vogel et al., 2009; Dangour et al., 2010;
and low in omega-6 PUFAs may help to prevent cognitive Wald et al., 2010; Ford and Almeider, 2012) have concluded
decline and dementia in older age (Martínez-Lapiscina that B vitamins cannot effectively slow down cognitive
et al., 2013). decline. However, this may be due to the low method-
ological quality and short duration of the included trials.
For example, a more recent and rigorously designed study
supplemented 900 older adults with 400 µg folate + 100 µg
54.4 VITAMINS FOR AD vitamin B12 for 24 months found improved ­immediate
and delayed memory performance compared to placebo
54.4.1 VITAMIN E AND VITAMIN C (Walker et al., 2012). Another study supplemented 818
older adults with 800 µg/day of folic acid or placebo for
A consistent body of work indicates that oxygen free radi- 3 years and found increased memory, information pro-
cals are involved in the pathological processes of AD cessing speed and sensorimotor speed with folic acid
(Ferreiro et al., 2012; Padurariu et al., 2013). This oxidative treatment, compared to placebo (Durga et al., 2007). High
stress hypothesis (Markesbery, 1997) has led to interest in quality studies on AD cohorts are also lacking at present.
the use of antioxidants, such as vitamins E and C, for the One double-blind RCT of high dose folate, vitamin B6 and
treatment of AD. B12 on 409 patients with mild-to-moderate AD found that
While lower plasma levels of vitamins C and E are treatment reduced homocysteine serum levels, but did
observed in AD (Lopes da Silva et al., 2014) and in vitro not slow cognitive decline, compared to placebo (Aisen
studies have shown that vitamin C can decrease β-amyloid et al., 2008). The same pattern of results was observed in
generation and AChE activity (Heo et al., 2013), longitu- a smaller 26-week study (Sun et al., 2007). Furthermore,
dinal epidemiological studies do not consistently show adverse effects have been noted, with a large cohort study
that habitual dietary vitamin C and E supplementa- on 1354 adults demonstrating that high serum folate lev-
tion will reduce risk of dementia (Crichton et al., 2013). els were related to impaired cognition (Moore et al., 2014).
Furthermore, evidence from clinical trials of vitamin E is These inconsistent findings, together with the potential
inconsistent (Farina et al., 2012), with some even suggesting for adverse effects indicates that, although homocysteine
592 Dementia

and B-vitamins share common biochemical pathways,


further research is required to demonstrate their efficacy 54.5 OTHER COMPLEMENTARY
in treating AD. THERAPIES FOR AD

Other complementary therapies have been used to treat


54.4.3 VITAMIN D
AD and other dementias and/or alleviate associated symp-
Vitamin D plays an important role in brain development toms. These interventions include, but are not limited to,
and function; however, despite several cohort s­tudies acupuncture, chiropractic, reflexology, remedial massage,
showing a relationship between vitamin D deficiency therapeutic touch, aromatherapy, mind–body therapy (e.g.
and cognitive decline and dementia, the mechanism meditation, qigong exercise), music therapy, Montessori-
is not well understood (McCann and Ames, 2008). A based activities, exercise programmes, combined melatonin
recent longitudinal study tracked 1658 elderly adults for and light therapy (Table 54.1). One report argued that the
approximately 5.6 years and found that vitamin D defi- most important aspect of CAM for dementia was to take an
ciency was associated with a substantially increased risk individualized approach that emphasized the therapeutic
of all-cause dementia and AD (Littlejohns et al., 2014). relationship together with the choice of CAM (Teut et al.,
A longitudinal study in women found that lower vita- 2013). For example, a trial of one-to-one Montessori-based
min D dietary intake was associated with a greater risk activities reduced anxiety in 44 dementia patients, com-
of developing AD (Annweiler et al., 2012). Even in the pared to baseline (van der Ploeg et al., 2013). Interestingly, a
absence of dementia, vitamin D insufficiency is related similar effect was observed during non-personalized social
to a decline in ­cognitive function (van der Schaft et al., contact, suggesting that activities promoting social contact
2013; Annweiler et al., 2015). Clinical trials examining the can also have positive outcomes. Some evidence exists for
efficacy of vitamin D supplementation for patients with the efficacy of other CAM therapies in dementia; for exam-
AD are currently ­lacking. Furthermore, due to the high ple, acupuncture has been shown to improve sleep quality
prevalence of ­hypovitaminosis D in older persons, those (Kwok et al., 2013) and yoga can improve physical health,
at risk should be tested for low serum 25-hydroxycholecal- depression and problem behaviours in dementia patients
ciferol (25-OH-D3) concentration and receive supplemen- (Fan and Chen, 2011). However, more rigorous scientific
tation if necessary (McCann and Ames, 2008; Annweiler research is needed to evaluate the effectiveness of these
et al., 2015). other treatments.

54.4.4 SOUVENAID® 54.6 CONCLUSIONS
Souvenaid ® is a medical nutrition drink specifically designed
to target the complex metabolic biochemical changes seen Due to the poor efficacy and tolerability of approved anti-
in AD. The formula contains precursors and cofactors that dementia pharmaceutical therapies, it is imperative that
promote synaptic formation, protection, function and opti- potentially suitable complementary medicines and thera-
mize brain metabolic activity including uridine monophos- pies be thoroughly examined and validated, if appropriate.
phate, choline, phospholipids, EPA, DHA and vitamins E, The pathogenesis and pathophysiology of AD is complex
C, B12, B6, folic acid and selenium. and diverse. Interventions need to take into account multiple
Although work on the efficacy of this nutritional factors including chronic inflammation, increased free rad-
approach as a treatment for mild-to-moderate AD is in ical production, glucose energy substrate hypometabolism
its infancy, current evidence shows modest improve- and reduced synaptic density and vascular integrity. Where
ments in cognition, most notably memory. Treatment for conventional pharmaceutical treatments typically follow a
12 weeks with Souvenaid ® compared to control, improved single-target method, complementary interventions, such
delayed verbal recall in patients with mild AD (Scheltens as herbal medicines and multivitamin supplementation can
et al., 2010) and a similar study showed improved memory contain multiple active components and have the capacity
scores and electroencephalograph (EEG) delta band func- to affect multiple targets. This makes CAMs ideal thera-
tional connectivity (Scheltens et al., 2012) over a 24-week pies for disorders (including dementia) with multifactorial/
period. Improved ADAS-cog scores were noted for AD multisystem pathophysiological components. Furthermore,
patients treated for 12 weeks relative to placebo; however, treatments that can target inflammation are of high impor-
improvement was only apparent for those with high base- tance (Lim et al., 2013).
line scores (Kamphuis et al., 2011). Furthermore, a large A large proportion of the existing evidence support-
24-week trial of Souvenaid did not slow cognitive decline ing the use of CAM for AD comes from animal and
in patients with mild-to-moderate AD compared to a con- in vitro studies and epidemiological data. Unfortunately,
trol group (Shah et al., 2013). Additional clinical trials clinical trials of these interventions frequently produce
with longer intervention periods are required to validate disappointing and inconsistent results due to poor meth-
this approach. odologies, small sample sizes and short intervention
Vitamins and complementary therapies used to treat Alzheimer’s disease 593

periods. Large scale, randomized, double-blind, placebo-­ T.C., Barnes, P.M. et al. (2015). Use of complementary
controlled trials are needed to evaluate the effectiveness health approaches among children aged 4–17 years in
and safety of these interventions. In addition, some tra- the United States: National Health Interview Survey,
ditional medical systems such as Chinese and Ayurvedic 2007–2012. National Health Statistics Reports, 78:
medicine emphasize health maintenance and disease 1–19.
prevention, which is currently not the primary focus of Brondino, N., Re, S., Boldrini, A. et al. (2014). Curcumin
western medicine. Hence, further research that assesses as a therapeutic agent in dementia: A mini systematic
the protective and preventive effects of these medicines review of human studies. The Scientific World Journal,
should also be carried out. 2014: 174282.
Burns, A., Perry, E., Holmes, C. et al. (2011). A double-
blind placebo-controlled randomized trial of Melissa
officinalis oil and donepezil for the treatment of agita-
REFERENCES
tion in Alzheimer’s disease. Dementia and Geriatric
Aguiar, S. and Borowski, T. (2013). Neuropharmacological Cognitive Disorders, 31: 158–164.
review of the nootropic herb Bacopa monnnieri. Camfield, D.A., Stough, C., Farrimond, J. et al. (2014).
Rejuvenation Research, 16 (4): 313–326. Acute effects of tea constituents L-theanine, caffeine,
Aisen, P., Scheider, L.S., Sano, M. et al. (2008). High-dose and epigallocatechin gallate on cognitive function
B vitamin supplementation and cognitive decline in and mood: A systematic review and meta-analysis.
Alzheimer disease: A randomized controlled trial. Nutrition Reviews, 72: 507–522.
JAMA, 300: 1774–1783. Cederholm, T., Salem, Jr. N. and Palmblad, J. (2013).
Akhondzadeh, S., Noroozian, M., Mohammadi, M. et al. Omega-3 fatty acids in the prevention of cogni-
(2003). Melissa officinalis extract in the treatment of tive decline in humans. Advances in Nutrition: An
patients with mild to moderate Alzheimer’s disease: International Review Journal, 4: 672–676.
A double blind, randomised, placebo controlled trial. Chan, P.-C., Xia, Q. and Fu, P.P. (2007). Ginkgo biloba
Journal of Neurology, Neurosurgery and Psychiatry, 28: leave extract: Biological, medicinal and toxicological
53–59. effects. Journal of Environmental Science and Health,
Ames, D. and Ritchie, C. (2007). Antioxidants and Part C, 25: 211–244.
Alzheimer’s disease: Time to stop feeding Vitamin E to Chiu, C.C., Su, K.P., Cheng, T.C. et al. (2008). The effects
dementia patients? International Psychogeriatrics, 19: of omega-3 fatty acids monotherapy in Alzheimer’s
1–8. disease and mild cognitive impairment: A preliminary
Annweiler, C., Dursun, E., Féron, F. et al. (2015). ‘Vitamin randomized double-blind placebo-controlled study.
D and cognition in older adults’: Updated international Progress in Neuro-Psychopharmacolgy and Biological
recommendations. Journal of Internal Medicine, 1: Psychiatry, 32: 1538–1544.
45–57. Clark, R. (2008). Plenary lecture: B-vitamins and preven-
Annweiler, C., Rolland, Y., Schott, A.M. et al. (2012) Higher tion of dementia. Proceedings of the Nutrition Society,
vitamin D dietary intake is associated with lower risk of 67: 75–81.
Alzheimer’s disease: A 7-year follow-up. The Journals Clarke, T.C., Black, L.I., Stussman, B.J. et al. (2015). Trends
of Gerontology Series A: Biological Sciences and in the use of complementary health approaches
Medical Sciences, 67: 1205–1211. among adults: United States, 2002–2012. National
Balk, E.M., Raman, G., Tatsioni, A. et al. (2007). Vitamin Health Statistics Reports, 79: 1–16.
B6, B12, and folic acid supplementation in cognitive Conquer, J.A., Tierney, M.C., Zecevic, J. et al. (2000).
function: A systematic review of randomized trials. Fatty acid analysis of blood plasma of patients with
Archives of Internal Medicine, 167: 21–30. Alzheimer’s disease, other types of dementia, and
Baum, L, Lam, C.W.K., Cheung, S.K.K. et al. (2008). cognitive impairment. Lipids, 35 (12): 1305–1312.
Six-month randomized placebo-controlled, Cox, K.H., Pipingas, A. and Scholey, A.B. (2014).
double-blind, pilot clinical trial of curcumin in Investigation of the effects of solid lipid curcumin on
patients with Alzheimer disease. Journal of Clinical cognition and mood in a healthy older population.
Psychopharmacology, 28: 110–113. Journal of Psychopharmacology, 29 (5): 642–651.
Benson, S., Downey, L.A. and Stough, C. (2014). An acute, Crichton, G.E., Bryan, J. and Murphy, K.J. (2013). Dietary
double-blind, placebo-controlled cross-over study antioxidants, cognitive function and dementia – A
of 320 mg and 640 mg doses of Bacopa monnieri systematic review. Plant Foods for Human Nutrition,
(CDRI 08) on multitasking stress reactivity and mood. 63: 279–292.
Phytotherapy Research, 28: 551–559. Dangour, A.D., Whitehouse, P.J., Rafferty, K. et al. (2010).
Birks, J. and Evans, J.G. (2009). Ginkgo biloba for cogni- B-vitamins and fatty acids in the prevention and treat-
tive impairment and dementia. Cochrane Database of ment of Alzheimer’s disease and dementia: A system-
Systematic Reviews, (1): CD003120. Black, L.I., Clarke, atic review. Journal of Alzheimer’s Disease, 22: 205–224.
594 Dementia

Dastmalchi, K., Dorman, H.J.D., Oinonen, P.P. et al. (2008). Goel, A., Kunnumakkara, A.B., Aggarwal, B.B. (2008).
Chemical composition and in vitro antioxidative activ- Curcumin as "Curecumin": From kitchen to clinic.
ity of a lemon balm (Melissa officinalis L.) extract. LWT Biochemical Pharmacology, 75: 787–809.
Food Science and Technology, 41: 391–400. Harrison, F.E. (2012). A critical review of vitamin C for
Downey, L.A., Kean, J., Nemeh, F. et al. (2013). An acute, the prevention of age-related cognitive decline and
double-blind, placebo-controlled crossover study Alzheimer’s disease. Journal of Alzheimer’s Disease,
of 320 mg and 640 mg doses of a special extract 29: 711–726.
of Bacopa monnieri (CDRI 08) on sustained cogni- Harrison, F.E., Bowman, G.L. and Polidori, M.C. (2014).
tive performance. Phytotherapy Research, 27 (9): Ascorbic acid and the brain: Rationale for the use
1407–1413. against cognitive decline. Nutrients, 6: 1752–1781.
Durga, J., van Boxtel, M.P., Schouten, E.G. et al. (2007). Hassaan, Y., Handoussa, H., El-Khatib, A.H. et al.
Effect of 3-year folic acid supplementation on (2014). Evaluation of plant phenolic metabolites as a
­cognitive function in older adults in the FACIT trial: source of Alzheimer’s drug leads. BioMed Research
A randomized, double blind, controlled trial. The International, 2014: 843263.
Lancet, 369: 208–216. Heo, J.H., Hyon-Lee, M.D. and Kyoung-Min, L. (2013). The
Dysken, M.W., Sano, M., Asthana, S. et al. (2014). Effect possible role of antioxidant vitamin C in Alzheimer’s
of vitamin E and memantine on functional decline in disease treatment and prevention. American Journal
Alzheimer disease. JAMA, 311: 33–44. of Alzheimer’s Disease and Other Dementias, 28:
Fan, J.T. and Chen, K.M. (2011). Using silver yoga exercises 120–125.
to promote physical and mental health of elders with Heo, J.H., Lee, S.T., Chu, K. et al. (2008). An open-label
dementia in long-term care facilities. International trial of Korean red ginseng as an adjuvant treatment
Psychogeriatrics, 23: 1222–1230. for cognitive impairment in patients with Alzheimer’s
Farina, N., Isaac, M.G., Clark, A.R. et al. (2012). Vitamin E disease. European Journal of Neurology, 15: 865–868.
for Alzheimer’s dementia and mild cognitive impair- Heude, B., Ducimetiere, P. and Berr, C. (2003). Cognitive
ment. Cochrane Database of Systematic Reviews, (11): decline and fatty acid composition of erythrocyte
CD002853. membranes: The EVA Study. The American Journal of
Ferreiro, E., Baldeiras, I., Ferreira, I.L. et al. (2012). Clinical Nutrition, 77: 803–808.
Mitochondrial- and endoplasamic reticulum-associated Howes, M.J.R. and Houghton, P.J. (2003). Plants used in
oxidative stress in Alzehimer’s disease: From patho- Chinese and Indian traditional medicine for improve-
genesis to biomarkers. International Journal of Cell ment of memory and cognitive function. Pharmacology
Biology, 2012: 735206. Biochemistry and Behavior, 75: 513–527.
Ford, A.H. and Almeida, O.P. (2012). Effect of homo- Hyung, S.J., DeToma, A.S., Brender, J.R. et al. (2013).
cysteine lowering treatment on cognitive function: Insights into antiamyloidogenic properties of the
A systematic review and meta-analysis of randomized green tea extract (-)-epigallocatechin-3-gallate toward
controlled trials. Journal of Alzheimer’s Disease, 29: meta-associated amyloid-β species. Proceedings of the
133–149. National Academy of Sciences USA, 110: 3743–3748.
Fotuhi, M., Mohassel, P. and Yaffe, K. (2009). Fish con- Jurenka, J.S. (2009). Anti-inflammatory properties of
sumption, long-chain omega-3 fatty acids and risk of curcumin, a major constituent of Curcuma longa:
cognitive decline or Alzheimer disease: A complex A review of preclinical and clinical research. Alternative
association. Nature Clinical Practice Neurology, 5: Medicine Review, 14: 141–153.
140–152. Kamphuis, P.J., Verhey, F.R., Olde Rikkert, M.G. et al.
Frass, M., Strassl, R.P., Friehs, H. et al. (2012). Use and (2011). Efficacy of a medical food on cognition in
acceptance of complementary and alternative medi- Alzheimer’s disease: Results from secondary analy-
cine among the general popuulation and medical ses of a randomized, controlled trial. The Journal of
personnel: A systematic review. The Oschsner Journal, Nutrition, Health and Aging, 15: 720–724.
12: 45–56. Kennedy, D.O., Jackson, P.A., Haskell, C.F. and Scholey,
Freund-Levi, Y., Eriksdotter-Jönhagen, M., Cederholm, A.M. (2007). Modulation of cognitive performance fol-
T. et al. (2006). Omega-3 fatty acid treatment in 174 lowing single doses of 120 mg Ginkgo biloba extract
patients with mild to moderate Alzheimer disease: administered to healthy young volunteers. Human
OmegAD study: A randomized double-blind trial. Psychopharmacology: Clinical and Experimental, 22:
Archives of Neurology, 63: 1402–1408. 559–66.
Gauthier, S. and Schlaefke, S. (2012). Efficacy and toler- Kennedy, D.O. and Scholey, A.B. (2003a)
ability of Ginkgo biloba extract EGb 761® in dementia: Electroencephalograph (EEG) effects of single doses
A systematic review and meta-analysis of random- of Ginkgo biloba and Panax ginseng in healthy
ized placebo-controlled trials. Clinical Interventions in young volunteers. Pharmacology, Biochemistry and
Aging, 9: 2065–2077. Behaviour, 75: 701–709.
Vitamins and complementary therapies used to treat Alzheimer’s disease 595

Kennedy, D.O., Scholey, A.B. and Wesnes, K.A. (2001). Mandel, S.A., Amit, T., Kalfon, L. et al. (2008). Targeting
Differential, dose dependent changes in cognitive multiple neurodegenerative diseases etiologies with
performance following acute administration of a multimodal-acting green tea catechins. The Journal of
Ginkgo biloba/Panax ginseng combination to healthy Nutrition, 138 (8): 1578S–1583S.
young volunteers. Nutritional Neuroscience, 4 (5): Markesbery, W.R. (1997). Oxidative stress hypothesis
399–412. in Alzheimer’s disease. Free Radical Biology and
Kennedy, D.O., Wake, G., Savelev, S. et al. (2003b). Medicine, 23: 134–137.
Modulation of mood and cognitive performance Martínez-Lapiscina, E.H., Clavero, P., Toledo, E. et al.
following acute administration of single doses of (2013). Mediterranean diet improves cognition. Journal
Melissa officinalis (Lemon Balm) with human CNS of Neurology, Neurosurgery and Psychiatry, 84:
1318–1325.
nicotinic and muscarinic receptor-binding properties.
McCann, J.C. and Ames, B.N. (2008). Is there convincing
Neuropsychopharmacology, 28: 1871–1881.
biological or behavioral evidence linking vitamin D
Kongkeaw, C., Dilothornsakul, P., Thanarangsarit, P. et
deficiency to brain dysfunction? The FASEB Journal,
al. (2014). Meta-analysis of randomized controlled
22: 1–20.
trials on cognitive effects of Bacopa monnieri extract.
Moore, E.M., Ames, D., Mander, A.G. et al. (2014).
Journal of Ethnopharmacology, 151: 528–535.
Among vitamin B12 deficient older people, high folate
Kuriyama, S., Hozawa, A., Ohmori, K. et al. (2006). Green
levels are associated with worse cognitive func-
tea consumption and cognitive function: A cross-sec- tion: Combined data from three cohorts. Journal of
tional study from the Tsurugaya Project. The American Alzheimer’s Disease, 39: 661–668.
Journal of Clinical Nutrition, 83: 355–361. Morgan, A. and Stevens, J. (2010). Does Bacopa mon-
Kwok, T., Leung, P.C., Wing, Y.K. et al. (2013). The effec- nieri improve memory performance in older per-
tiveness of acupuncture on the sleep quality of sons? Results of a randomized, placebo-controlled,
elderly with dementia: A within-subjects trial. Clinical double-blind trial. The Journal of Alternative and
Interventions in Aging, 8: 923–929. Complementary Medicine, 16: 753–759.
Lawrence, G.D. (2013). Dietary fats and health: Dietary Morris, M.S. (2003). Homocysteine and Alzheimer’s dis-
recommendations in the context of scientific evidence. ease. The Lancet Neurology, 2: 425–428.
Advances in Nutrition, 4: 293–302. Morris, M.S. (2012). The role of B vitamins in prevent-
Lee, S.T., Chu, K., Sim, J.Y. et al. (2008). Panax ginseng ing and treating cognitive impairment and decline.
enhances cognitive performance in Alzheimer disease. Advances in Nutrition: An International Review, 3:
Alzheimer Disease and Associated Disorders, 22: 801–812.
222–226. Padurariu, M., Ciobica, A., Lefter, R. et al. (2013). The
Lim, A., Krajina, K. and Marsland, A. (2013). Peripheral oxidative stress hypothesis in Alzheimer’s disease.
inflammation and cognitive aging. Modern Trends in Psychiatra Danubina, 25: 401–409.
Pharmacopsychiatry, 28: 175–187. Pase, M.P., Kean, J., Sarris, J. et al. (2012). The cognitive-
Littlejohns, T.J., Henley, W.E., Lang, I.A. et al. (2014). enhancing effects of Bacopa monnieri: A systematic
Vitamin D and the risk of dementia and Alzheimer review of randomized, controlled human clinical
disease. Neurology, 83 (10): 920–928. trials. The Journal of Alternative and Complementary
Medicine, 18: 647–652.
Lloret, A., Badía, M.C., Mora, N.J. et al. (2009). Vitamin
Perry, E.K., Pikering, A.T., Wang, W.W. et al. (1999).
E paradox in Alzheimer’s disease: It does not prevent
Medicinal plants and Alzheimer’s disease: From
loss of cognition and may even be detrimental. Journal
etnobotany to phytotherapy. Journal of Pharmacy and
of Alzheimer’s Disease, 17: 143–149.
Pharmacology, 51: 527–534.
Loef, M. and Walach, H. (2013). The Omega-6/Omega-3
Quinn, J.F., Raman, R. and Thomas, R.G. (2010).
ratio and dementia or cognitive decline: A systematic
Docosahexaenoic acid supplementation and cogni-
review on human studies and biological evidence.
tive decline in Alzheimer disease: A randomized trial.
Journal of Nutrition in Gerontology and Geriatrics, 32: JAMA, 304: 1903–1911.
1–23. Radad, K., Gille, G., Liu, L.L. and Rausch, W.D. (2006). Use
Lopes da Silva, S., Vellas, B., Elemans, S. et al. (2014). of ginseng in medicine with emphasis on neurodegen-
Plasma nutrient status of patients with Alzheimer’s dis- erative disorders. Journal of Pharmacological Science,
ease: Systematic review and meta-analysis. Alzheimer’s 100: 175–186.
and Dementia: The Journal of the Alzheimer’s Reay, J.L., Kennedy, D.O. and Scholey, A.B. (2006). Effects
Association, 10 (4): 485–502. of Panax ginseng, consumed with and without glucose,
Madhusoodanan, S. and Ting, M.B. (2014). Pharmacological on blood glucose levels and cognitive performance
management of behavioral symptoms associated with during sustained ‘mentally demanding’ tasks. Journal
dementia. World Journal of Psychiatry, 4: 72–79. of Psychopharmcology, 20: 771–781.
596 Dementia

Ringman, J.M., Frautschy, S.A., Cole, G.M. et al. (2005). A Teut, M., Bloedt, S., Baur, R. et al. (2013). Dementia:
potential role of the curry spice in Alzheimer’s disease. Treating patients and caregivers with complementary
Current Alzheimer Research, 2: 131–136. and alternative medicine – Results of a clinical expert
Russo, A. and Borrelli, F. (2005). Bacopa monniera, a conference using the World Café method. Forschende
reputed nootropic plant: An overview. Phytomedicine, Komplementärmedizin/Research in Complementary
12: 305–317. Medicine, 20: 276–280.
Saini, N., Singh, D. and Sandhir, R. (2012). Neuroprotective Tully, A.M., Roche, H.M., Doyle, R. et al. (2003). Low
effects of Bacopa monnieri in experimental model of serum cholesteryl ester-docosahexaenoic acid levels
dementia. Neurochemical Research, 37: 1928–1937. in Alzheimer’s disease: A case-control study. British
Sathyanarayanan, V., Thomas, T., Einöther, S.J. et al. Journal of Nutrition, 89: 483–489.
(2013). Brahmi for the better? New findings chal- van der Ploeg, E.S., Eppingstall, B., Camp, C.J. et al.
lenging cognition and anti-anxiety effects on (2013). A randomized crossover trial to study the
Brahmi (Bacopa monniera) in healthy adults. effect of personalized, one-to-one interaction using
Psychopharmacology (Berlin), 227: 299–306. Montessori-based activities on agitation, affect, and
Schaefer, E.J., Bongard, V., Beiser, A.S. et al. (2006). engagement in nursing home residents with dementia.
Plasma phosphatidycholine docosahexaenoic acid International Psychogeriatrics, 25: 565–575.
context and risk of dementia and Alzheimer disease. van der Schaft, J., Koek, H.L., Dijkstra, E. et al. (2013).
Archives of Neurology, 63: 1545–1550. The association between vitamin D and cognition:
Scheltens, P., Kamphuis, P.J., Verhey, F.R. et al. (2010). A systematic review. Ageing Research Reviews, 12:
Efficacy of a medical food in mild Alzheimer’s dis- 1013–1023.
ease: A randomized, controlled trial. Alzheimer’s and Vellas, B., Coley, N., Ousset, P.J., Berrut, G., Dartigues, J.F.,
Dementia: The Journal of the Alzheimer’s Association, Dubois, B. et al. (2012). Long-term use of standardised
6: 1–10. Ginkgo biloba extract for the prevention of Alzheimer’s
Scheltens, P., Twisk, J.W., Blesa, R. et al. (2012). Efficacy disease (GuidAge): A randomised placebo-controlled
of Souvenaid in mild Alzheimer’s disease: Results from trial. Lancet Neurology, 11 (10): 851-859.
a randomized, controlled trial. Journal of Alzheimer’s Vogel, T., Dali-Youcef, N., Kaltenbach, G. et al. (2009).
Disease, 31: 225–236. Homocysteine, vitamin B12, folate and cognitive
Schilling, S., Tzourio, C., Dufouil, C. et al. (2014). Plasma functions: A systematic and critical review of the
lipids and cerebral small vessel disease. Neurology, 83: literature. International Journal of Clinical Practice, 63:
1844–1852. 1061–1067.
Shah, R.C., Kamphuis, P.J., Leurgans, S. et al. (2013). Wald, D.S., Kasturiratne, A. and Simmonds, M. (2010).
The S-Connect study: Results from a randomized, Effect of folic acid, with or without other B vitamins,
controlled trial of Souvenaid in mild-to-moderate on cognitive decline: Meta-analysis of random-
Alzheimer’s disease. Alzheimer’s Research and ized trials. The American Journal of Medicine, 123:
Therapy, 5: 59. 522–527.
Sharangi, A.B. (2009). Medicinal and therapeutic poten- Walker, J.G.G., Batterham, P.J., Mackinnon, A.J. et al.
tialities of tea (Camellia sinensis L.) – A review. Food (2012). Oral folic acid and vitamin B-12 supplemen-
Research International, 42: 529–535. tation to prevent cognitive decline in community-
Stough, C., Downey, L.A., Lloyd, J. et al. (2008). Examining dwelling older adults with depressive symptoms – The
the nootropic effects of a special extract of Bacopa Beyond Ageing Project: A randomized controlled
monniera on human cognitive functioning: 90 Day trial. The American Journal of Clinical Nutrition, 95:
double-blind placebo-controlled randomized trial. 194–203.
Phytotherapy Research, 22: 1629–1634. Wang, Y., Feng, Y., Fu, Q. et al. (2013). Panax notogin-
Sun, Y., Lu, C.J., Chien, K.L. et al. (2007). Efficacy of mul- senoside Rb1 ameliorates Alzheimer’s disease by
tivitamin supplementation containing vitamins B6 and upregulating brain-derived neurotrophic factor and
B12 and folic acid as adjunctive treatment with a cho- downregulating Tau protein expression. Experimental
linesterase inhibitor in Alzheimer’s disease: A 26-week, and Therapeutic Medicine, 6: 826–830.
randomized, double-blind, placebo-controlled study Wang, B.-S., Wang, H., Wei, Z.-H. et al. (2009).
in Taiwanese patients. Clinical Therapeutics, 29: Efficacy and safety of natural acetylcholines-
2204–2214. terase inhibitor huperzine A in the treatment of
Tan, Z.S., Harris, W.S., Beiser, A.S. et al. (2012). Red blood Alzheimer’s disease: An updated meta-analysis.
cell omega-3 fatty acid levels and markers of acceler- Journal of Neural Transmission (Vienna, Austria: 1996),
ated brain aging. Neurology, 78: 658–664. 116 (4): 457–465.
Tan, M.S., Yu, J.T., Tan, C.C. et al. (2015). Efficacy and Wesnes, K.A., Ward, T., McGinty, A. and Petrini, O.
adverse effects of ginkgo biloba for cognitive impair- (2000). The memory enhancing effects of a Ginkgo
ment and dementia: A systematic review and meta- biloba/Panax ginseng combination in healthy middle-
analysis. Journal of Alzheimer’s Disease, 43: 589–603. aged volunteers. Psychopharmacology, 152: 353–361.
Vitamins and complementary therapies used to treat Alzheimer’s disease 597

Xing, S.H., Zhu, C.X., Zhang, R. et al. (2014). Huperzine and fibrils, binds plaques, and reduces amyloid
A in the treatment of Alzheimer’s disease and vas- in vivo. Journal of Biological Chemistry, 280:
cular dementia: A meta-analysis. Evidence-Based 5892–901.
Complementary and Alternative Medicine, 2014: Yang, G., Wang, Y., Tian, J. et al. (2013). Huperzine A for
363985. Alzheimer’s disease: A systematic review and meta-
Yakoot, M., Salem, A. and Helmy, S. (2013). Effect of analysis of randomized clinical trials. PLoS ONE, 8 (9):
Memo®, a natural formula combination, on Mini-Mental e74916.
State Examination scores in patients with mild cognitive Yang, M., Xu, D.D., Zhang, Y. et al. (2014). A systematic
impairment. Clinical Interventions in Aging, 8: 975–981. review on natural medicines for the prevention and
Yang, L., Hao, J., Zhang, J. et al. (2009). Ginsenoside treatment of Alzheimer’s disease with meta-analyses
Rg3 promotes beta-amyloid peptide degradation by of intervention effect of Ginkgo. American Journal of
enhancing gene expression of neprilysin. Journal of Chinese Medicine, 42 (3): 505–521.
Pharmacy and Pharmacology, 61: 375–380. Yang, Y., Zhang, Z., Li, S. et al. (2014). Synergy effects of
Yang, F., Lim, G.P., Begum, A.N. et al. (2005). Curcumin herb abstracts: Pharmacokinetics and pharmacody-
inhibits formation of amyloid beta oligomers namic basis. Fitoterapia, 92: 133–147.
55
Prevention of Alzheimer’s disease and
Alzheimer’s dementia

TOM C. RUSS, KAREN RITCHIE AND CRAIG W. RITCHIE

a description at the level of the population and one at the


55.1 INTRODUCTION level of the individual. In his classic paper ‘Sick individuals
and sick populations’, Geoffrey Rose distinguishes between
There is now consensus that a substantial proportion of factors which affect the incidence of a disease in a popu-
cases of dementia is potentially preventable. With this lation and those which affect whether an individual will
agreed, the discussion has centred on the extent to which develop the disease (Rose, 1985). For example, if a public
the risk of developing Alzheimer’s disease (ADis) could be health intervention succeeds in reducing a risk factor in the
reduced and exactly which risk factors should be the focus of population (such as shifting the population distribution of
public health attention. At this point, it is worth making the systolic blood pressure to the left – i.e. more like the Kenyan
distinction – widely accepted in the clinical and academic nomads than the London civil servants in Figure 55.1) this
community – between ADis (a pathological process) and may reduce the incidence of dementia. However, even in
Alzheimer’s dementia (ADem, the clinical dementia syn- the lower risk population as a whole, there will still be indi-
drome associated with ADis). This chapter will discuss what viduals with ‘high’ levels of the risk factor and, thus, still
prevention (or risk reduction) means in the context of both at increased risk. Indeed, an individual with an optimized
disease and dementia, which will lead us on to consider risk factor profile may still develop dementia, assuming that
the life course approach in dementia epidemiology. This this single risk factor is not, on its own, a sufficient cause.
approach hypothesizes that risk and protective factors from Alternatively, the individual may unsuccessfully attempt to
all stages of life – from before conception to late life – may be reduce this risk factor, even though the population mean of
important in affecting dementia risk. Following these theo- the risk factor and the incidence of dementia both reduce.
retical considerations, we will report the current research The individual in this case might meet talk of ‘prevention’
evidence for the prevention of ADis and ADem, summarize with consternation – they reduced their level of the risk fac-
the policy recommendations that have been made worldwide tor but still developed dementia; or they tried and failed to
and consider whether public health interventions in other diminish the risk factor, thus failing to prevent dementia.
domains, for example cardiovascular disease, may have had Does this mean they are in some way responsible for devel-
additional positive impact on dementia risk. oping the disease? It is clear that we must be very careful,
both in our thinking and our language here. Indeed, some
have called for us to talk in terms of ‘risk reduction’ rather
than ‘prevention.’ However, there is a clear and important
55.2 WHAT DO WE MEAN BY
difference between these concepts – risk reduction strategies
PREVENTION? would focus on the exposure variable thought to be associ-
ated with the outcome, whereas prevention implies a focus
Much of the scientific literature in this area uses a phrase on reducing the frequency or incidence of the outcome,
like ‘dementia prevention’ to refer to reducing the preva- which may be achieved not only by risk reduction but also
lence of dementia in a specific population. However, more specific – perhaps pharmacological – interventions. ‘Risk’ in
recently some concerns have been expressed about this use the former sense could potentially be reduced but is unlikely
of language, which highlights a frequent confusion between to be eliminated completely. Notwithstanding these caveats,

598
Prevention of Alzheimer’s disease and Alzheimer’s dementia 599

for simplicity we will continue to use the term ‘prevention’ Braak, 1997). The search for ADis decades before demen-
in this chapter. tia is expected to develop is the specific focus of projects
The distinctions between ADis and ADem as well as like PREVENT (Ritchie and Ritchie, 2012; Ritchie et al.,
between ‘risk reduction’ and ‘prevention’ outlined above are 2013). These projects hypothesize that earlier life disease
relevant when we consider the usual definitions of types of will be a more specific indicator of later dementia than it
prevention. Prevention has traditionally been divided into has been shown to be – in multiple projects – in later life.
primary, secondary and tertiary prevention and Figure 55.2 Indeed, the Religious Orders Study and the Memory and
outlines how this distinction applies to ADis and ADem. Aging Project found that a third of people who died with-
However, defining these categories closely relies on the out dementia had sufficient neuropathological changes to
natural history of the condition in question being under- be classified as ADis with intermediate certainty (Bennett
stood and, thus, a few words about this might be helpful et al., 2006). This and other evidence including a neuropatho-
here. ADis has been widely considered a condition of later logical series from the Medical Research Council Cognitive
life. However, this is not the complete picture and emerg- Function and Ageing Study (MRC CFAS) (Wharton et al.,
ing evidence points to ADis in fact having its genesis in 2011) suggests that ADis (neuropathological changes in the
midlife or even earlier, with ADem being a manifestation brain) and ADem (cognitive and functional decline) are not
of this disease typically reserved for people in later life. identical but, rather, are two related, parallel phenomena.
For example, unselected autopsy samples of adults aged Three recent Cochrane Diagnostic Test Accuracy Reviews
25 years and older showed that ADis was commonly pres- highlighted this as they demonstrated that amyloid-β lev-
ent in individuals decades before the usual age of clini- els (a core pathological lesion in ADis) whether measured
cal onset of symptoms (Braak and Braak, 1991; Braak and in cerebrospinal fluid (CSF) (Ritchie et al., 2014) or using
positron emission tomography (PET) imaging (Zhang
et al., 2014; Smailagic et al., 2015) had very low specificity
30

Kenyan nomads
London civil servants
but better sensitivity for later ADem in populations with
mild cognitive impairment (MCI). Indeed, the data sup-
20

porting the use of biomarkers for the diagnosis of dementia


in general are limited and variable in quality (Noel-Storr et
%

al., 2013). In essence, you may need Alzheimer’s pathology


10

to get ADem, but you can have Alzheimer’s pathology and


still enjoy normal cognitive health well into later life. For
this reason, it is imperative always to be clear whether we
0

60 80 100 120 140 160 180 200


are referring to ADis or ADem – an essential distinction
Systolic blood pressure (mm Hg)
that the abbreviation AD frequently obscures.
Primary prevention refers to preventing a ­ condition
Figure 55.1 Distributions of systolic blood pressure in developing in the first place in healthy people. If the neu-
middle-aged men in two populations. (Redrawn from ropathological changes of ADis begin in midlife this
Rose, G., International. Journal of Epidemiology, 1985 by means that implementing primary prevention measures
permission of Oxford University Press). would need to begin earlier than that. Therefore, it would

Primary prevention Secondary prevention Tertiary prevention


Identification: Identification: Identification:
Extent of Alzheimer’s disease pathology

Genetic risks Biomarker abnormalities Clinical symptoms


Clinical risks Clinical symptoms Functional impairment
Risk of Alzheimer’s dementia

e.g. head injury/diabetes Genetic and clinical risks


Intervention:
Intervention: Intervention: Specific pharmacology
Risk reduction Risk reduction Psychosocial
Enhanced nutritional and interventions and support
Who leads: lifestyle modifications Risk reduction
The individual Specific pharmacology Enhanced nutritional and
Public health lifestyle modifications
Education Who leads:
The individual Who leads:
Public health The individual
Education The individual’s family
Primary care Primary care
Specialists Specialists
Social services
The third sector

Life course

Figure 55.2 Primary, secondary and tertiary prevention of Alzheimer’s disease and dementia.
600 Dementia

be helpful from a methodological point of view to have a (Brookmeyer et al., 1998; Brookmeyer et al., 2007; Ritchie
reliable method to identify early brain changes of ADis et al., 2015).
in vivo, in order to identify those people in whose brains Tertiary prevention refers to attempts to modify the dis-
not even the very early stages of ADis are detectable. Many ease and its symptomatic course once it has become symp-
­middle-aged people with no symptoms of dementia are in tomatically apparent so that patients do not progress to later,
fact likely to have early ADis and considered therefore ‘at higher morbidity stages of the condition. In the context of
risk’ of developing ADem. Thus, primary prevention initia- dementia, this would mean both disease-modifying treat-
tives would have to go ‘upstream’ and begin in early adult- ments targeting individuals who have already shown cog-
hood, if not even earlier in life. nitive and/or functional decline (for simplicity, we would
Because of the long, clinically silent latent period of also include those labelled as having ‘MCI’ in this category)
ADis, rather than representing primary prevention endeav- as well as pharmacological treatments targeting symptom
ours, many midlife intervention studies would probably fall profiles and psychosocial interventions. Disappointingly,
under our definition of secondary prevention. In the con- though there have been a large number of studies targeting
text of dementia, the aim of secondary prevention initiatives disease modification over the last two decades in this popu-
is to prevent or delay the clinical onset of ADem in indi- lation, they have been predominantly negative (Figure 55.3)
viduals who already have the pathological changes of ADis (Mangialasche et al., 2010; Schneider et al., 2014). Perhaps
in their brain. There is evidence that delaying the clinical this stands to reason when we consider the patho-clinical
onset of symptoms would have substantial effects on the course being proposed for ADis, whereby people with even
number of people with dementia by compressing morbidity, mild ADem may in fact have very severe ADis, which has
therefore increasing the number of years spent disease-free been occult (using current technologies) for decades. In

Aβ production
Preclinical
Aβ aggregation
Aβ clearance (immunotherapy) ACI-24
Tau aggregation or phosphorylation Phenylthiazolylhydrazide
Cholinergic drugs
Anti-Aβ antibodies Phase 1
Others Rhodanines

RS-0406 Minocycline
Affitope AD-01*

Phase 2
GSK-933776 AL-208*
Affitope AD-02
SEN-1269 AL-108* Anti-tau
PF-04360365
MABT-5102A antibodies
ACC-001 MTC/LMT*
AN-1792* Phase 3 Nicotinamide
UB-311 IVIg
CAD-106 Valproate* N-031112
SP-233 Bapineuzumab (NP-12)* N-744
ELND-005
Solanezumab Phase 4 Lithium*
R-1450 NGF
PBT1* Tramiprosate* Donepezil† Antioxidants

Rivastigmine† SB-742457
Peptidic anti- EGCg
V-950 PBT2* Statins
aggregants Galantamine† PRX-03140*
NIC5-15 Tarenflurbil* Memantine† BDNF
Latrepirdine PUFAs*
E-2012*
Semagacestat TTP-488
Bryostatin-1 Phenserine*
Exebryl-1 Rosiglitazone* PF-04447943
MK-0752* EHT-0202*
EVP-6124
BMS-708163 Huperzine-A* Ladostigil
CHF-5074
AF-102B*
NGXsc Pioglitazone* AZD-3480*
CTS-21166* ABT-089* Talsaclidine* AF-267B

PF-3084014* M-30
GSM-1
Begacestat* Memoquin
Anti-β-secretase antibodies

BBS1

Figure 55.3 (See colour insert.) Drug development in Alzheimer’s disease: drugs being investigated for Alzheimer’s
disease therapy, reported according to the most advanced phase of study and main therapeutic properties (including data
from studies in vitro and animal models). Aβ, amyloid-β; BBS1, anti-β-site antibodies; BDNF, brain-derived neurotrophic
factor; EGCg, ­epigallocatechin-3-gallate; IVIg, intravenous immunoglobulin; LMT, leuco-methylthioninium; MTC, methyl-
thioninium chloride; NGF, nerve growth factor; NGXsc, NGX series compounds; PUFAs, polyunsaturated fatty acids; GSM,
γ-secretase modulator; RCT, randomized controlled trial. *RCTs in Alzheimer’s disease not ongoing. †Drugs approved for
the treatment of Alzheimer’s disease. (Reprinted from The Lancet Neurology, 9, Mangialasche, F. et al., Alzheimer’s dis-
ease: Clinical trials and drug development, 702–716, 2010, with permission from Elsevier.)
Prevention of Alzheimer’s disease and Alzheimer’s dementia 601

effect, people with ADem already have end-stage brain risk factor for dementia – by far – is ageing and, thus, in
failure with multiple concurrent pathological processes order to understand dementia properly (and to have any
being relevant. Therefore, a single targeted approach to one hope of preventing it) we must understand brain ageing
pathology is highly unlikely to produce substantial clinical much better (Brayne, 2007).
benefit. There has been much interest recently in findings One possible time point to intervene is before someone
presented at the 2015 Alzheimer’s Association International develops the clinical symptoms of ADem – commonly
Conference that a sample of participants in the negative ­cognitive changes (predominantly memory) and functional
phase 3 trials of solanezumab (Doody et al., 2014; Siemers decline. However, one question we have not yet addressed is
et al., 2016) with mild ADem who were allocated during what influences the timing of the clinical onset of demen-
the double-blind phase to the treatment group have subse- tia? We have already seen that a large proportion of people
quently declined more slowly than participants who were without dementia have ADis in their brains. Why do they
in the control group, despite both groups receiving the not have any symptoms of dementia? A relevant theory
treatment after the end of the trials (Liu-Seifert et al., 2015). answering this question is that of cognitive reserve – that
However, while this single positive finding is encouraging, certain people’s brains are more resilient to the pathologi-
the effect remains small and merely maintained people in cal changes of ageing and ADis (Stern, 2012). This concept
a state of dementia without recovery to an earlier phase of includes the overlapping ideas of brain reserve and cognitive
the clinical syndrome. Moreover, these findings will require reserve. The former primarily reflects structural differences,
corroboration in further ongoing trials and, practically for example numbers of neurones, whereas the latter refers to
speaking, it is unlikely that an expensive treatment admin- a more active process in which the brain adapts to compen-
istered as an infusion would become widely used in clinical sate for age- or disease-related changes by altering or opti-
practice, particularly given the small effect size. However, mizing its function. Support for the idea of cognitive reserve
if asymptomatic people showing evidence of ADis could comes from the association between lower educational and
be identified using what will come to be known as ‘partner occupational attainment and increased dementia risk, sug-
diagnostics’, the impact of disease modification for second- gesting that brain changes resulting from positive learning
ary prevention is likely to be much more meaningful and and occupational experiences could render the brain less
therefore cost-effective. susceptible to insult (Valenzuela and Sachdev, 2006). A cor-
ollary of the ability of being able to tolerate more pathology
before the clinical onset of dementia (i.e. people with greater
cognitive reserve) is that such people decline more rapidly
55.3 THE ORIGINS OF DEMENTIA after diagnosis, analogous to a dam being broken – the dam
in question being built through the person’s life through
We have already seen that ADis begins years, or even education (Stern et al., 1995, 1999). Further biological sup-
decades, before ADem. In addition to the pathological port for the concept of cognitive reserve can be found in
evidence cited above, there is also a great deal of evidence the observation that people with dementia with greater edu-
from prospective cohort studies of the association between cational attainment show decreased parietotemporal blood
midlife risk factors and later dementia. Furthermore, flow on PET scans compared to people with dementia with
findings from intervention studies in late life in people the same levels of clinical symptoms but who left school at a
with dementia – including those looking at medication younger age (Stern et al., 1992). A linked finding is that lev-
(Mangialasche et al., 2010) or risk factor modification els of neuropathology do not seem to relate well to levels of
(McGuinness et al., 2009; McGuinness et al., 2014) – have clinical symptoms. This is particularly the case for amyloid,
been uniformly disappointing. More recent studies in older though tau levels in the brain may relate better to symptoms
people who are free from dementia though have shown that (Arriagada et al., 1992).
dietary, clinical, lifestyle and brain training may reduce the The growing consensus that a longer period of an indi-
rate of brain failure (e.g. the Finnish Geriatric Intervention vidual’s life may be relevant to their dementia risk has led to
Study to Prevent Cognitive Impairment and Disability the widespread application, as with many chronic diseases,
[FINGER] [Ngandu et al., 2015] and the Mediterranean- of the life course paradigm of epidemiology to ADis (Ben-
DASH Intervention for Neurodegenerative Delay [MIND] Shlomo and Kuh, 2002; Whalley et al., 2006). This simply
[Morris et al., 2015]) optimistically pointing to the message suggests that factors from all stages of development – i.e. at
that ‘it is never too late to prevent dementia.’ However, the any point in the life course – may influence (either increase
failures in people with established dementia adds weight or decrease) an individual’s probability of developing demen-
to the idea that ADem represents the later stages of a long tia. Risk may be accumulated over time or a particular factor
process and that the logical thing to do is to intervene at may influence risk only at a particular point in time (a critical
an earlier stage and/or younger age when the brain is better period) or more at one point in time than at other times (a
preserved. Indeed, many people speculate that the secret to sensitive period). Figure 55.4 depicts hypothetical life course
therapeutic success in ADis is to intervene earlier, when the influences on brain ageing with several critical periods. An
brain is in a state more amenable to preservation of func- advantage of this life course approach is that it provides a
tion. This ties in to the observation that the most important framework to help disentangle the genetic and epigenetic
602 Dementia

ICV

Spectrum of High GM
educa
age-related Low e tion
cognitive function du cation
since birth
WM

Small birth size Education


Epigenetic influences
Intrauterine environment Socioeconomic status Lifestyle
Placental function Nutrition Cardiovascular
Maternal nutrition Education risk and disease
Genetic influences

First trimester Second trimester Third trimester 0 4 8 12 16 20 30 40 50 60 70 80 Age (years)


Prenatal period Childhood/adolescence Adulthood Old age

Figure 55.4 Hypothesized model on the origins and life course of brain ageing. Several ‘critical periods’ (prenatal period,
childhood/adolescence, adulthood and old age) are identified during which an individual is at greatest risk of damage if
exposed to putative risk factors. Normal development of intracranial volume (ICV) and brain volume (grey matter [GM] and
white matter [WM]) is presented for these critical periods, and the possible different risk factors influencing brain develop-
ment throughout these periods are described. Allegedly, genetics and epigenetic influences could alter brain structure
and function throughout life, but their impact would probably fade with age. In addition, the spectrum of age-related
cognitive ability from birth to old age is presented in this figure, with a schematic view of our findings that small birth size
is related to poor cognitive functioning only in those with lower educational levels. (From Muller, M. et al., Pediatrics, 134,
761–770, 2014. With permission.)

factors as well as complex gene–­environment interactions, including forced expiratory volume, forced vital capacity
which most probably work together to ­determine dementia and peak expiratory flow), which is influenced by smoking,
risk. It also lays the foundation for interventions – p
­ rincipally illness and socioeconomic deprivation at various stages of
at a public-health level – ­ targeting groups and focusing life. Figure 55.5 shows four different possible trajectories of
resources with modification of risks during critical or sensi- pulmonary function throughout the life course: (A) ‘­normal’
tive periods as based on the evidence. growth and decline; (B) reduced maximal lung function as
A concrete example supporting the application of the a result of impaired pre- or post-natal growth leading to a
life course paradigm in dementia is the association between greater risk of later disability and mortality from chronic
physical stature and dementia. Maximal height is attained respiratory disease in spite of a normal rate of decline; (C)
in the first two decades of life and, in addition to being normal growth but accelerated decline and, consequently,
influenced by genes, is regarded as a marker of early life premature disability and death from chronic respiratory
experience, including illness, adversity, nutrition and psy- disease (scenarios A–C could also be complicated by epi-
chosocial stress (Batty et al., 2006). Thus, height could be sodes of reversible impairments in pulmonary function
considered a proxy for these factors. The fact that shorter [D]) (Strachan and Sheikh, 2004). The association between
height is associated with an increased risk of dementia poorer lung function and increased dementia risk further
leads one to conclude that such early life adversities may supports the life course perspective in dementia and points
influence the individual’s subsequent risk of developing to the possible importance of these factors in the develop-
dementia (Russ et al., 2014). Another measure that captures ment of the disease (Russ et al., 2015b). However, the picture
relevant exposures throughout the life course is pulmonary is more complicated here. While it is difficult to imagine how
function (estimated by a variety of spirometric measures, shorter height might, in itself, affect dementia risk, poorer
Prevention of Alzheimer’s disease and Alzheimer’s dementia 603

Not to scale Not to scale


Level of lung function

Level of lung function


A A
Level below Level below B
which which
symptoms symptoms
may occur may occur

Birth 10 20 30 40 50 60 70 80 Birth 10 20 30 40 50 60 70 80

Age in years Age in years

Not to scale Not to scale


Level of lung function

Level of lung function


A A
Level below B Level below D B
which which D
symptoms C symptoms C
may occur may occur

Birth 10 20 30 40 50 60 70 80 Birth 10 20 30 40 50 60 70 80

Age in years Age in years

Figure 55.5 Schematic representation of the life course of pulmonary function measurements. (Adapted and redrawn
from Figure 10.2 in Kuh, D. et al., A Life Course Approach to Chronic Disease Epidemiology, 2004, by permission of Oxford
University Press.)

lung function could plausibly be a risk factor in its own right, Of course, in an ideal world, one would study the early
affecting the brain through hypoxia, reduced oxygenation of life origins of dementia with direct measure of the factors
brain tissues with the associated hypercapnic state leading to of interest in early life, repeated measures throughout life
a lower brain pH, which is known to drive amyloid-β aggre- and sufficiently long follow-up for a large number of people
gation (Atwood et al., 1998), as well as a risk marker because to develop ADem. Clearly, such a prospective design is cur-
of the factors mentioned above which influence it. rently unfeasible. Thus, alternative strategies must be found
We have seen that the life course paradigm suggests that for different aspects of the methodology. One possibility is a
risk and protective factors from all periods of life are poten- retrospective design (as in the AGES–Rekjavik study [Muller
tially relevant. Until recently, there has been no evidence et al., 2014]), in which individuals who are already older
directly linking birth parameters with brain ageing. An indi- (either an aged cohort or a group of people with dementia for
rect link can be inferred from the influence of birthweight whom matched controls can be found) for whom relevant
(and parental social class) on cognitive function at the age of data, by coincidence, were recorded earlier in life – either
11 years (Shenkin et al., 2001), which, in turn, is a risk fac- routinely, as is the case for birthweight, or because they were
tor for dementia, possibly more so for vascular dementia than part of an earlier study which is still accessible, for example
ADem (Whalley et al., 2000; McGurn et al., 2008). However, the Lothian and Aberdeen Birth Cohort studies, the Nun
a recent retrospective cohort study as part of the Age Gene/ study or the Aberdeen children of the 1950s cohort (Deary
Environment Susceptibility (AGES)–Reykjavik study in et al., 2009; Leon et al., 2006; Snowdon, 2011; Whalley et al.,
Iceland demonstrated that lower birthweight was associated 2011; Deary et al., 2012). An alternative solution to the same
with slower processing speed and poorer executive function problem is to measure a proxy for something that happened
at the aged of 75 years, but only in those with lower educa- earlier in life, as in the studies of height and lung function
tional attainment (Muller et al., 2014). It can be seen that mentioned above (Russ et al., 2014; Russ et al., 2015b).
there are complex interplays between cognition, education Follow-up is often limited by economic resources and one
and socioeconomic status that need to be carefully unpicked, solution to this is ‘passive’ follow-up using electronic health
but this finding that the birthweight – hypothesized to be a records, which allows a large number of people to be fol-
measure of the intrauterine environment – is related to cog- lowed up for a long time for relatively little money. There are
nitive function in later life is an exciting one which echoes understandably some concerns that this may lead to under-
other such findings in the area of ‘foetal origins of adult dis- ascertainment of dementia cases (in addition to the univer-
eases’ (Barker, 1990; Calkins and Devaskar, 2011). sal under-diagnosis in the community), but the evidence
604 Dementia

suggests that using multiple sources is likely to be adequate that the results in the real world might not be as hopeful
for epidemiological purposes, especially if the ascertainment as the report suggests. Finally, and probably most relevant
or otherwise is not related to the exposure under study and to dementia given what we know of its natural history, the
particularly in certain contexts, for example universal health- possibility that the early stages of the condition might influ-
care (Knopman et al., 2011). Indeed, this is the approach ence the risk factors and so produce spurious associations
being taken by large cohort studies such as UK Biobank (reverse causality) must be very carefully considered in all
(N = 500,000) (Sudlow et al., 2015). A final possibility is not work on dementia prevention.
waiting until people develop ADem but to identify those who In line with the life course paradigm of epidemiology
have ADis through the use of biomarkers, for example struc- ­outlined above, some of the risk factors just mentioned
tural and functional imaging, neuropsychological testing and seemed to be most important at particular stages of life:
CSF measures. This is the approach taken in the European educational attainment in early life; and hypertension
Prevention of Alzheimer’s Dementia (EPAD) project, which and obesity in midlife. Related to education, we have also
will be described in Section 55.5 (Ritchie et al., 2015). seen above that lower early life intelligence is associated
with an increased risk of dementia in later life, possibly
more so for vascular dementia than ADem (Whalley et al.,
2000; McGurn et al., 2008). These findings come from the
55.4 PRIMARY PREVENTION Aberdeen and Lothian Birth Cohort studies, which were
made possible by the discovery in the 1990s of the results
We defined primary prevention above as attempting to prevent of intelligence tests sat by all 11-year olds in Scotland in
ADis developing in the first place and suggested that many the summers of 1932 and 1947, which allowed these indi-
midlife prevention studies were, in fact, studying secondary viduals to be followed up in later life (Scottish Council for
prevention – the prevention of ADem in people who already Research in Education, 1933; Scottish Council for Research
had ADis. That said, primary prevention can be considered at in Education, 1949; Deary et al., 2009; Whalley et al., 2011;
two levels: the individual and the population. Individual-level Deary et al., 2012). A similarly serendipitous discovery led
risk factors could potentially be targeted at any stage of the to the Nun study, which linked early life linguistic ability
life course and, indeed, could be the focus of population-wide inferred from autobiographies written by the nuns shortly
public health interventions. Population-level risk factors, on before taking their religious vows with later life measures
the other hand, have been relatively understudied and in this (Snowdon, 2011). The measures of linguistic ability used
chapter we will focus on environmental risk factors and a geo- were ‘idea density’ and ‘grammatical complexity.’ Lower lev-
graphical approach through disease mapping. els of both measures – particularly the former – were associ-
ated with poorer cognition in later life and an increased risk
55.4.1 INDIVIDUAL-LEVEL RISK FACTORS of pathologically confirmed ADem (Snowdon et al., 1996;
Riley et al., 2005). These studies have caught the imagination
A group at the University of California, San Francisco sys- of the wider world and have both inspired plays – ‘Still Life
tematically reviewed the literature in relation to seven poten- Dreaming’ by the Spare Tyre Theatre Company about the
tially modifiable individual-level risk factors for ADis: in Aberdeen and Lothian Birth Cohort Studies and ‘27’ by the
early life, educational attainment; in midlife, hypertension National Theatre of Scotland and the Royal Lyceum Theatre,
and obesity; and throughout the life course, diabetes, smok- Edinburgh, about the Nun study, both first performed in
ing, depression and physical inactivity (Barnes and Yaffe, 2011 in Edinburgh.
2011). They estimated that approximately half of ADem In midlife, raised blood pressure or body mass index
cases worldwide could be attributed to these risk factors (BMI) seem to be the most important risk factors for demen-
and that a 10% reduction in each risk factors could prevent tia. However, both of these physical parameters change over
a million cases worldwide; a 25% reduction would prevent the life course, which can complicate the interpretation of
three million cases. A more recent paper took into account their association with dementia risk. Blood pressure, for
the fact that these seven risk factors are not independent: in example, decreases in the years preceding a diagnosis of
these models, eliminating all the risk factors would prevent dementia (Skoog et al., 1996). This could lead one to the erro-
28.2% cases of ADem worldwide (Norton et al., 2014). More neous conclusion that lower blood pressure in later life was
reasonable estimates of 10% or 20% reduction in risk factors a risk factor for dementia when this relationship is, in fact,
could result in the prevention 8.3% or 15.3% cases world- an example of the reverse causality mentioned above. The
wide by 2050. Findings from such modelling studies do not relationship observed between trajectories of blood pressure
necessarily translate into the real world where factors like and later dementia risk is more complex still and affected by
confounding, bias and reverse causality are likely to com- treatment with antihypertensive drugs (Figure 55.6) (Joas
plicate the association between risk factor and dementia. et al., 2012). The association between BMI and dementia
Unmeasured confounders may be the real causal risk fac- may be even more complex. An association between being
tors and so altering these measured risk factors might have underweight and an increased risk of dementia was recently
no effect on dementia risk. Bias, the consequence of selec- reported from a large study of two million patients in pri-
tive mortality or incomplete follow-up, could also mean mary care (Qizilbash et al., 2015) and this finding seems to
Prevention of Alzheimer’s disease and Alzheimer’s dementia 605

Mean age of sample* not random and varies from location to location (Whalley,
45 51 57 69 77 2012). A recent systematic review summarized the pub-
180
lished literature based on within-study comparisons (thus
removing the possibility that any differences might result
Systolic blood pressure mm Hg

160 from methodological differences between studies) and con-


cluded that there was evidence at all geographical scales for
geographical variation in dementia prevalence and inci-
140 dence (Russ et al., 2012). However, the review highlighted
the dearth of large-scale (i.e. small area) studies, which
are likely to be the most informative in terms of shedding
120 light on the possible causes for this geographical variation.
More recently, researchers using data from the Dementia in
Swedish Twins study (HARMONY) conducted a small area
100 Bayesian disease mapping study and reported two-to-three-
1968 1974 1980 1992 2000 2005
Time fold higher odds of dementia in the north of Sweden com-
No history of antihypertensive treatment: pared to the south after the removal of genetic variance and
Dementia cases the influence of factors shared within families (Figure 55.7)
Non-dementia cases – suggesting that this variation is the result of one or more,
History of antihypertensive treatment: as yet unidentified, unshared environmental risk factors
Dementia cases (Russ et al., 2015a). Indeed, the importance of the influ-
Non-dementia cases
ence of environmental differences between children from
the same family has been described as ‘[o]ne of the most
Figure 55.6 Trajectories of systolic blood pressure from
importance findings that has emerged from human behav-
1968–1969 to 2005–2006 among those with and without
dementia in 2000–2006 and stratified by antihypertensive ioral genetics’ (Plomin and Daniels, 2011). A complemen-
treatment between 1968 and 1992. The trajectories are tary disease mapping study in the United Kingdom using a
adjusted for age, education, cardiovascular disease, dia- narrow age cohort of people born in 1921 (the 1932 Scottish
betes mellitus, smoking, stroke and body mass index. The Mental Survey cohort) passively followed up over eight
predicted trajectories show the mean when the continu- decades showed no variation in dementia odds based on the
ous confounders are mean centred and the categorical county of the school participants attended aged 11 years but
confounders are set to 0. *The age axis shows the mean substantial variation, following a broadly similar pattern to
age of the study sample (N = 707). Dashed line indicates that seen in Sweden, based on residential location after the
dementia cases; solid line, non-dementia cases. (From
age of 60 years (Figure 55.8) (Russ et al., 2015a).
Joas, E. et al., Blood pressure trajectories from midlife to
late life in relation to dementia in women followed for 37 This clear geographical variation in dementia risk
years, Hypertension, 59, 796–801, 2012.) observed in two countries, showing broadly similar pat-
terns invites speculation as to the mechanism underlying
the variation. This is being actively investigated, but there
have been replicated in the Whitehall I Study (Kivimaki are several possible environmental factors that may be
et al., 2015). Since this contradicts the received opinion related. Vitamin D has been linked to dementia and cog-
that raised BMI is associated with dementia risk (Daviglus nitive decline through a variety of mechanisms including
et al., 2010a, 2010b; Prince et al., 2014), there has been a clearance of amyloid-β protein and cerebrovascular changes
great deal of speculation regarding this finding. Suggested (Balion et al., 2012; Annweiler et al., 2013; Littlejohns et al.,
explanations have included the possibility that investigators 2014; Annweiler et al., 2015). One of the factors which influ-
conflated midlife and late-life BMI measurement by includ- ence vitamin D levels is latitude, with individuals living
ing individuals aged 40–80 years at baseline in the study at higher latitudes having lower serum levels (Brouwer-
(Strand et al., 2015) and the hypothesis that the focus should Brolsma et al., 2013). Thus, vitamin D could be a plausible
be on weight loss rather than absolute BMI in relation to mechanism explaining part of the higher dementia risk seen
dementia (van der Burg et al., 2015). The latter point seems further north in both countries. Another possibility is the
to be supported by the finding that different trajectories in micronutrient selenium which has been linked to cogni-
BMI can be seen in those who go on to develop dementia tive impairment and ADem (Loef et al., 2011; Berr et al.,
compared to those who do not (Gustafson et al., 2012). 2012). Selenium levels in the soil are lower in the north of
Sweden than the south and so could plausibly explain some
55.4.2 ENVIRONMENTAL RISK FACTORS of the observed results (Parkman and Hultberg, 2002).
There is also interest in ambient air pollution, particularly
A new area of research interest is investigating risk factors particulate matter with an aerodynamic diameter of less
for dementia through a geographical approach, which has than 2.5 μm (PM2.5), which may be associated with loss of
been fruitful in other areas of medicine. It is now estab- white matter that might represent accelerated brain ageing
lished that the geographical distribution of dementia risk is (Chen et al., 2015). Clearly much more detailed research is
606 Dementia

Odds ratio of dementia


< 0.67
300
0.67–0.83 Miles
0.83–0.91
0.91–0.95
0.95–1.05
1.05–1.1
1.1–1.2
1.2–1.5
> 1.5

North

(a) (b)

Figure 55.7 (See colour insert.) Age-adjusted odds ratio of dementia in male (a) and female (b) Swedish twins with
twin-level random effects (likely to capture genetic and shared environmental variance) removed. (From Russ, T.C. et al.,
Epidemiology, 26, 263–270, 2015a. With permission.)

Odds ratio of dementia


< 0.67
0.67–0.83
0.83–0.91
0.91–0.95
0.95–1.05
1.05–1.1
1.1–1.2
1.2–1.5
>1.5

(a) (b)

100
Miles

North

(c) (d)

Figure 55.8 (See colour insert.) Age-adjusted odds ratio of dementia in the SMS1932 cohort: childhood – males (a),
females (b); adulthood – males (c), females (d). (From Russ, T.C. et al., Epidemiology, 26, 263–270, 2015a. With permission.)
Prevention of Alzheimer’s disease and Alzheimer’s dementia 607

required to clarify the mechanisms behind the observed dementia and are willing to participate in a clinical trial
geographical variation in dementia risk but it seems to be for the secondary prevention of ADem. Every partici-
a fruitful avenue to pursue alongside more well-established pant will be fully characterized biologically and clinically
efforts such as cardiovascular disease risk. (including PET-amyloid imaging, CSF- and blood-based
biomarkers in addition to a bespoke cognitive outcome
measure, the EPAD Clinical Evaluation). Thus, partici-
pants in EPAD will act as a ‘readiness cohort’ for the trial.
55.5 SECONDARY PREVENTION
The cohort will also generate a vast amount of data for
(INTERVENING IN MIDLIFE) improved disease modelling in this secondary prevention
population. For example, it will be possible to create indi-
In Figure 55.2, we outlined the concept of secondary pre- vidualized ‘probability scores’ based on the three dimen-
vention as it applies to ADis and ADem. That is, that, in sions of risk factors, biomarker expression of disease and
contrast to people with advanced, symptomatic disease, clinical expression of disease. Using these probability
people with evidence of disease but no (or minimal) scores, it will then be possible to identify subpopulations
symptoms may derive maximal benefit from disease- with evidence of the target pathology relevant to the pro-
modifying interventions or combinations of interven- posed therapy and who are most likely to decline during
tions. The benefit is likely to be maximal because of two the proposed time course for the trial. The trial is per-
facts: first, the disease processes will be at an earlier phase petual with new drugs or combinations of drugs being
and therefore more likely to be susceptible to interven- brought into the project on a regular basis and arms being
tion; second, since dementia is recognized as being the closed as they show futility or success and thereafter grad-
end point of brain failure with multiple pathological pro- uation to phase 3 confirmatory studies. The trial itself will
cesses contributing to the clinical phenotype, specific use adaptive methodologies to optimize selection of sub-
interventions lead to little, if any, net clinical benefit. It is jects to interventions showing (during the course of the
highly unlikely that all neurodegenerative disease pathol- study) the greatest likelihood of successful graduation to
ogies emerge s­imultaneously – it is much more likely phase 3 confirmatory trials. Within the same programme,
that this is a sequential process. If this sequence could be the EPAD project will aim to control numerous factors
defined empirically, it would be possible to target the most that have probably conspired to create the disappoint-
upstream, potentially triggering process, which could ments seen in recent attempts at drug development for
fundamentally affect the disease course and therefore lead disease modification. These include knowledge of every
to maximal (clinical) benefit. subject’s disease status pre-baseline to reduce screen fail-
The EPAD project has been designed to optimize the ures to a minimum, the collection of run-in data from the
proof-of-concept testing of novel interventions (predomi- longitudinal cohort study to be used analytically to mea-
nantly pharmacological) for the secondary prevention of sure the post-randomization effect of drugs and to select
ADem. It is a single trials platform that benefits from access those which appear promising, the continuous updat-
to numerous pre-existing ‘parent’ cohorts across Europe ing of disease models from all trial and cohort data and
and the development of a new EPAD longitudinal cohort finally the establishment of a dedicated network of EPAD
study of over 6000 research participants. Trial Delivery Centres to ensure the highest standards of
This work poses three major challenges: first, we need data quality to minimize measurement bias.
empirically derived models of disease in the ‘second- While EPAD focuses on single and combinational phar-
ary prevention’ population. How do you assess research macological approaches, other (non-­ pharmacological)
participants from a biological and clinical perspective multimodal approaches are already showing great prom-
to derive data which can be used to improve disease ise. One such study of people at risk of developing clini-
models – which biomarkers? How does one measure cog- cal dementia is the FINGER study (Kivipelto et al., 2013;
nition in preclinical populations in a way which captures Ngandu et al., 2014). Preliminary results from the first 2
underlying brain impairment? What other features may years of follow-up were recently published and they indi-
be relevant to optimize these models? Second, how do you cated that a multidomain intervention (focusing on diet,
take these ­so-derived probability models and apply them exercise, cognitive training and vascular risk monitoring)
to the study population to stratify probability of decline resulted in slower decline in the intervention group than
necessary in the placebo group to drive effect in the proof- in the group receiving general health advice (Ngandu
of-concept clinical trial? Third, how, in the absence of et al., 2015). This finding that a simple, non-invasive
either of these in the first instance, can you create the intervention could have an effect on the rate of cognitive
simulations necessary for adaptive trial designs to test the decline in individuals at risk of developing dementia is
interventions? These three challenges are being addressed very encouraging. Results from another trial (prevention
in the EPAD project (Ritchie et al., 2015; Vaudano et al., of dementia by intensive vascular care [PreDIVA]) inves-
2015). This Innovative Medicines Initiative funded pub- tigating whether intensive vascular care in older adults
lic–private partnership will develop a longitudinal cohort could decrease dementia incidence are awaited with inter-
study of 6000 people (at any given time) who do not have est (Richard et al., 2009).
608 Dementia

Report 2014, which focused entirely on risk reduction


55.6 POLICY CONTEXT (Prince et al., 2014). This report reviewed the evidence
for risk factors for dementia under the broad headings of
There have been a number of national and international developmental and early life factors, psychological factors,
statements on dementia prevention in recent years and it lifestyle-related factors and cardiovascular disease risk fac-
would be useful to summarize their conclusions to provide tors. They concluded from their systematic review of the lit-
context for the foregoing consideration. In 2010, the US erature that there was strong evidence for potentially causal
National Institutes of Health held a State-of-the-Science associations between low educational attainment, midlife
conference on ‘Preventing Alzheimer’s disease and cogni- hypertension and smoking and diabetes (both at any stage
tive decline’ (Daviglus et al., 2010a, 2010b). Their conclu- of life) with dementia. There was also evidence for associa-
sions were not optimistic: ‘Currently, no evidence of even tions between physical and cognitive activity and dementia
moderate scientific quality exists to support the associa- risk, but this was less secure and the possibility of reverse
tion of any modifiable factor … with reduced risk for ADis’ causality still remains. They concluded that two mecha-
(Daviglus et al., 2010b). However, they did make a number nisms underlying dementia development could be cognitive
of recommendations for future research, including robust (or brain) reserve and vascular pathology.
definitions of the outcome used in dementia studies, involv- A more recent systematic review and Delphi consensus
ing caregivers in estimating daily function and institut- study came to similar conclusions to the ADI report, finding
ing robust new p ­ opulation-based cohort studies as well as support for midlife hypertension and obesity, depression,
using existing cohort studies, for example of cardiovascular physical inactivity, diabetes, hyperlipidaemia and smoking
disease risk factors, participants in which are now reach- (Deckers et al., 2015). Finally, the Alzheimer’s Association
ing later life. A few years later, following the G8 dementia report to the World Dementia Council concluded that there
summit in London, a group of international experts issued was strong evidence from population-based studies for reg-
a more optimistic statement entitled simply ‘Dementia ular physical activity and cardiovascular disease risk factors
(Including Alzheimer’s ­disease) can be prevented’. They (diabetes, obesity, smoking and hypertension) influencing
concluded that more work was needed to discover modifi- dementia risk (Baumgart et al., 2015).
able risk factors for dementia but, importantly, that ‘there
is already sufficient evidence to justify immediate action …
[i.e.] tell people that adopting a healthy lifestyle may help
55.7 DECREASING RATES OF
to ward off dementia as it does for other diseases’ (Smith
and Yaffe, 2014). The authors estimated that approximately
DEMENTIA
half of ADem cases could be attributed to known risk fac-
tors and that instituting public health measures including As alluded to above, there is growing evidence for declining
smoking cessation, adopting the Mediterranean diet, pre- prevalence or incidence of dementia with time (Larson et al.,
venting obesity and diabetes, avoiding excessive alcohol 2013). The MRC CFAS-II showed a decrease in dementia
consumption and treating hypertension could prevent one prevalence in England from 8.3% to 6.5% over two decades
in five predicted new cases over the next decade. The opti- (Matthews et al., 2013). There is a great deal of interest in
mism of the Blackfriars Consensus on brain health and these and similar findings from Europe (Schrijvers et al.,
dementia (Lincoln et al., 2014; Public Health England, UK 2012; Qiu et al., 2013) and the United States (Manton et al.,
Health Forum, 2014), which came out around the same time 2005; Langa et al., 2008). Hypothesized mechanisms include
was fuelled by reports of decreasing prevalence of dementia decreased rates of cardiovascular disease (a public health suc-
over time (but approximately stable absolute numbers due cess story) and increased levels of educational attainment in
to demographic change), which will be discussed in more more recent generations – paralleling the mechanisms of vas-
detail in Section 55.7. The focus of this report was the overlap cular risk and cognitive reserve highlighted in the ADI report
in aetiology between vascular risk and dementia and con- above (Prince et al., 2014). However, even with reductions in
cluded that ‘the scientific evidence is evolving rapidly and dementia prevalence of approximately 20%, the projected
sufficient to justify considered action and further research demographic trends of an ageing population worldwide will
on dementia risk reduction, both by reducing the modifi- dwarf these reductions and result in substantial increases in
able risk factors and improving the recognized protective the absolute number of people with dementia, particularly in
factors” (Public Health England, UK Health Forum, 2014). low-to-middle income countries (Prince et al., 2013).
They too called for more population-based cohort studies
(both new and existing cohorts) and highlighted the MRC-
funded Dementias Platform UK (DPUK) and the European
Union Joint Programme on Neurodegenerative Disorders 55.8 CONCLUSIONS
(JPND) in this regard.
Possibly the most important document in this area is There is evidence that a substantial proportion of cases of
Alzheimer’s Disease International’s (ADI) World Alzheimer ADis and ADem may be preventable and, indeed, lower levels
Prevention of Alzheimer’s disease and Alzheimer’s dementia 609

of cardiovascular disease risk factors and greater educational Bennett, D., Schneider, J., Arvanitakis, Z. et al. (2006).
attainment may already be having an effect to reduce demen- Neuropathology of older persons without cogni-
tia rates, even if absolute numbers are likely to continue to tive impairment from two community-based studies.
increase. If total prevention is impossible, even delaying the Neurology, 66: 1837–1844.
onset of dementia would reduce the number of cases and have Ben-Shlomo, Y. and Kuh, D. (2002). A life course approach
substantial public health impact. The question then becomes to chronic disease epidemiology: Conceptual mod-
one of which risk factors to target and, importantly, at what els, empirical challenges and interdisciplinary per-
point in the life course to intervene. The most important spectives. International Journal of Epidemiology, 31:
risk factors seem to be low educational attainment, midlife 285–293.
hypertension, smoking and diabetes. We have seen that it is Berr, C., Arnaud, J. and Akbaraly, T.N. (2012). Selenium
likely that risk and protective factors are probably at work and cognitive impairment: A brief-review based on
from birth, if not earlier, and that a comprehensive dementia results from the EVA study. Biofactors, 38: 139–144.
prevention strategy would take this into account and send Braak, H. and Braak, E. (1991). Neuropathological
out the message that it is never too early to start thinking stageing of Alzheimer-related changes. Acta
about reducing one’s risk of developing dementia. Neuropathologica, 82: 239–259.
Braak, H. and Braak, E. (1997). Frequency of stages of
Alzheimer-related lesions in different age categories.
REFERENCES Neurobiology of Aging, 18: 351–357.
Brayne, C. (2007). The elephant in the room – Healthy
Annweiler, C., Dursun, E., Feron, F. et al. (2015). ‘Vitamin brains in later life, epidemiology and public health.
D and cognition in older adults': Updated international Nature Reviews Neuroscience, 8: 233–239.
recommendations. Journal of Internal Medicine, 277: Brookmeyer, R., Gray, S. and Kawas, C. (1998). Projections
45–57. of Alzheimer’s disease in the United States and the
Annweiler, C., Llewellyn, D.J. and Beauchet, O. public health impact of delaying disease onset.
(2013). Low serum vitamin D concentrations American Journal of Public Health, 88: 1337–1342.
in Alzheimer’s disease: A systematic review and Brookmeyer, R., Johnson, E., Ziegler-Graham, K. and
­meta-analysis. Journal of Alzheimer’s Disease, Arrighi, H.M. (2007). Forecasting the global burden
33: 659–674. of Alzheimer’s disease. Alzheimer’s & Dementia: The
Arriagada, P.V., Growdon, J.H., Hedley-Whyte, E.T. Journal of the Alzheimer’s Association, 3: 186–191.
and Hyman, B.T. (1992). Neurofibrillary tangles but Brouwer-Brolsma, E.M., Bischoff-Ferrari, H.A., Bouillon, R.
not senile plaques parallel duration and severity of et al. (2013). Vitamin D: Do we get enough? A discus-
Alzheimer’s disease. Neurology, 42: 631. sion between vitamin D experts in order to make a
Atwood, C.S., Moir, R.D., Huang, X. et al. (1998). step towards the harmonisation of dietary reference
Dramatic aggregation of Alzheimer Aβ by Cu(II) is intakes for vitamin D across Europe. Osteoporosis
induced by conditions representing physiological International, 24: 1567–1577.
acidosis. The Journal of Biological Chemistry, 273: Calkins, K. and Devaskar, S.U. (2011). Fetal origins of adult
12817–12826. disease. Current Problems in Pediatric and Adolescent
Balion, C., Griffith, L.E., Strifler, L. et al. (2012). Vitamin Health Care, 41: 158–176.
D, cognition, and dementia: A systematic review and Chen, J.C., Wang, X., Wellenius, G.A. et al. (2015).
meta-analysis. Neurology, 79: 1397–1405. Ambient air pollution and neurotoxicity on brain struc-
Barker, D.J. (1990). The fetal and infant origins of adult ture: Evidence from women’s health initiative memory
disease. British Medical Journal, 301: 1111. study. Annals of Neurology, 78 (3): 466–476.
Barnes, D.E. and Yaffe, K. (2011). The projected Daviglus, M.L., Bell, C.C., Berrettini, W. et al. (2010a). NIH
effect of risk factor reduction on Alzheimer’s state-of-the-science conference statement: Preventing
­disease ­prevalence. The Lancet Neurology, 10: Alzheimer’s disease and cognitive decline. NIH
819–828. Consensus and State-of-the-Science Statements, 27:
Batty, G.D., Shipley, M.J., Langenberg, C. et al. (2006). 1–30.
Adult height in relation to mortality from 14 can- Daviglus, M.L., Bell, C.C., Berrettini, W. et al. (2010b).
cer sites in men in London (UK): Evidence from the National Institutes of Health State-of-the-Science
­original Whitehall study. Annals of Oncology, 17: Conference Statement: Preventing Alzheimer disease
157–166. and cognitive decline. Annals of Internal Medicine,
Baumgart, M., Snyder, H.M., Carrillo, M.C. et al. (2015). 153: 176–181.
Summary of the evidence on modifiable risk factors for Deary, I.J., Gow, A.J., Pattie, A. and Starr, J.M. (2012).
cognitive decline and dementia: A population-based Cohort profile: The Lothian Birth Cohorts of 1921
perspective. Alzheimer’s & Dementia: The Journal of and 1936. International Journal of Epidemiology, 41:
the Alzheimer’s Association, 11: 718–726. 1576–1584.
610 Dementia

Deary, I.J., Whalley, L.J. and Starr, J.M. (2009). A Lifetime Liu-Seifert, H., Siemers, E., Holdridge, K.C. et al. (2015).
of Intelligence: The Scottish Mental Surveys of 1932 Delayed-start analysis: Mild Alzheimer’s disease
and 1947. Washington, DC: American Psychological patients in solanezumab trials, 3.5 years. Alzheimer’s
Association. and Dementia: Translational Research and Clinical
Deckers, K., van Boxtel, M.P.J., Schiepers, O.J.G. Interventions, 1 (2): 111–121.
et al. (2015). Target risk factors for dementia pre- Loef, M., Schrauzer, G.N. and Walach, H. (2011). Selenium
vention: A systematic review and Delphi consensus and Alzheimer’s disease: A systematic review. Journal
study on the evidence from observational stud- of Alzheimer’s Disease, 26: 81–104.
ies. International Journal of Geriatric Psychiatry, 30: Mangialasche, F., Solomon, A., Winblad, B. et al. (2010).
234–246. Alzheimer’s disease: Clinical trials and drug develop-
Doody, R.S., Thomas, R.G., Farlow, M. et al. (2014). ment. The Lancet Neurology, 9: 702–716.
Phase 3 trials of solanezumab for mild-to-moder- Manton, K.C., Gu, X.L. and Ukraintseva, S.V.
ate Alzheimer’s disease. New England Journal of (2005). Declining prevalence of dementia in the U.S.
Medicine, 370: 311–321. elderly population. Advances in Gerontology, 16:
Gustafson, D.R., Backman, K., Joas, E. et al. (2012). 37 30–37.
Years of body mass index and dementia: Observations Matthews, F.E., Arthur, A., Barnes, L.E. et al. (2013).
from the prospective population study of women in A two-decade comparison of prevalence of dementia
Gothenburg, Sweden. Journal of Alzheimer’s Disease, in individuals aged 65 years and older from three geo-
28: 163–171. graphical areas of England: Results of the Cognitive
Joas, E., Backman. K., Gustafson, D. et al. (2012). Blood Function and Ageing Study I and II. Lancet, 382:
pressure trajectories from midlife to late life in rela- 1405–1412.
tion to dementia in women followed for 37 years. McGuinness, B., Craig, D., Bullock, R. et al. (2014). Statins
Hypertension, 59: 796–801. for the treatment of dementia. Cochrane Database of
Kivimaki, M., Singh-Manoux, A., Shipley, M.J. and Systematic Reviews, (7): CD007514.
Elbaz, A. (2015). Does midlife obesity really McGuinness, B., Todd, S., Passmore, P. and Bullock,
lower dementia risk? The Lancet Diabetes and R. (2009). Blood pressure lowering in patients
Endocrinology, 3: 498. ­without prior cerebrovascular disease for pre-
Kivipelto, M., Solomon, A., Ahtiluoto, S. et al. vention of cognitive impairment and dementia.
(2013). The Finnish Geriatric Intervention Study Cochrane Database of Systematic Reviews, (4):
to Prevent Cognitive Impairment and Disability CD004034.
(FINGER): Study design and progress. Alzheimer’s & McGurn, B., Deary, I.J. and Starr, J.M. (2008).
Dementia: The Journal of the Alzheimer’s Association, Childhood cognitive ability and risk of late-onset
9: 657–665. Alzheimer and vascular dementia. Neurology, 71:
Knopman, D.S., Petersen, R.C., Rocca, W.A. et al. (2011). 1051–1056.
Passive case-finding for Alzheimer’s disease and Morris, M.C., Tangney, C.C, Wang, Y. et al. (2015). MIND
dementia in two U.S. communities. Alzheimer’s & diet slows cognitive decline with aging. Alzheimer’s &
Dementia: The Journal of the Alzheimer’s Association, Dementia: The Journal of the Alzheimer’s Association,
7: 53–60. 11 (9): 1015–1022.
Langa, K.M., Larson, E.B., Karlawish, J.H. et al. (2008). Muller, M., Sigurdsson, S., Kjartansson, O. et al. (2014).
Trends in the prevalence and mortality of cognitive Birth size and brain function 75 years later. Pediatrics,
impairment in the United States: Is there evidence of 134: 761–770.
a compression of cognitive morbidity? Alzheimer’s & Ngandu, T., Lehtisalo, J., Levalahti, E. et al. (2014).
Dementia: The Journal of the Alzheimer’s Association, Recruitment and baseline characteristics of par-
4: 134–144. ticipants in the Finnish Geriatric Intervention Study
Larson, E.B., Yaffe, K. and Langa, K.M. (2013). New to Prevent Cognitive Impairment and Disability
insights into the dementia epidemic. New England (FINGER) – A randomized controlled lifestyle trial.
Journal of Medicine, 369: 2275–2277. International Journal of Environmental Research and
Leon, D.A., Lawlor, D.A., Clark, H. and Macintyre, S. Public Health, 11: 9345–9360.
(2006). Cohort profile: The Aberdeen children of the Ngandu, T., Lehtisalo, J., Solomon, A. et al. (2015).
1950s study. International Journal of Epidemiology, A 2 year multidomain intervention of diet, exercise,
35: 549–552. cognitive training, and vascular risk monitoring versus
Lincoln, P., Fenton, K., Alessi, C. et al. (2014). The control to prevent cognitive decline in at-risk elderly
Blackfriars Consensus on brain health and dementia. people (FINGER): A randomised controlled trial.
Lancet, 383: 1805–1806. Lancet, 385: 2255–2263.
Littlejohns, T.J., Henley, W.E., Lang, I.A. et al. (2014). Noel-Storr, A.H., Flicker, L., Ritchie, C.W. et al.
Vitamin D and the risk of dementia and Alzheimer (2013). Systematic review of the body of evidence
disease. Neurology, 83: 920–928. for the use of biomarkers in the diagnosis of
Prevention of Alzheimer’s disease and Alzheimer’s dementia 611

dementia. Alzheimer’s & Dementia: The Journal of the biomarkers of late-onset Alzheimer’s disease. BMJ
Alzheimer’s Association, 9: e96–e105. Open, 2 (6): e001893.
Norton, S., Matthews, F.E., Barnes, D.E. et al. (2014). Ritchie, C.W., Wells, K. and Ritchie, K. (2013). The
Potential for primary prevention of Alzheimer’s PREVENT research programme – A novel research
disease: An analysis of population-based data. The programme to identify and manage midlife risk for
Lancet Neurology, 13: 788–794. dementia: The conceptual framework. International
Parkman, H. and Hultberg, H. (2002). Occurrence and Review of Psychiatry, 25: 748–754.
Effects of Selenium in the Environment – A Literature Rose, G. (1985). Sick individuals and sick populations.
Review. Göteborg, Sweden: IVL Swedish International Journal of Epidemiology, 14: 31–38.
Environmental Research Institute. Russ, T.C., Batty, G.D., Hearnshaw, G.F. et al. (2012).
Plomin, R. and Daniels, D. (2011). Why are children in Geographical variation in dementia: Systematic
the same family so different from one another? review with meta-analysis. International Journal of
International Journal of Epidemiology, 40: 563–582. Epidemiology, 41: 1012–1032.
Prince, M., Albanese, E., Guerchet, M. and Prina, M. Russ, T.C., Gatz, M., Pedersen, N.L. et al. (2015a).
(2014). World Alzheimer Report 2014. Dementia Geographical variation in dementia: Examining the
and Risk Reduction: An Analysis of Protective and role of environmental factors in Sweden and Scotland.
Modifiable Factors. London, United Kingdom: Epidemiology, 26: 263–270.
Alzheimer’s Disease International. Russ, T.C., Kivimäki, M., Starr, J.M. et al. (2014). Height
Prince, M., Guerchet, M. and Prina, M. (2013). The Global in relation to dementia death: Individual participant
Impact of Dementia 2013–2050. London, United meta-analysis of 18 UK prospective cohort studies.
Kingdom: Alzheimer’s Disease International. The British Journal of Psychiatry: The Journal of Mental
Public Health England, UK Health Forum. (2014). Science, 205: 348–354.
Blackfriars Consensus on Promoting Brain Health: Russ, T.C., Starr, J.M., Stamatakis, E. et al. (2015b).
Reducing Risks for Dementia in the Population. Pulmonary function as a risk factor for ­dementia death:
London, United Kingdom: UK Health Forum. An individual participant meta-­analysis of six UK general
Qiu, C., von Strauss, E., Backman, L. et al. (2013). Twenty- population cohort studies. Journal of Epidemiology and
year changes in dementia occurrence suggest Community Health , 69: 550–556.
decreasing incidence in central Stockholm, Sweden. Stern Y. (2012). Cognitive reserve in ageing and
Neurology, 80: 1888–1894. Alzheimer’s disease. The Lancet Neurology, 11:
Qizilbash, N., Gregson, J., Johnson, M.E. et al. (2015). BMI 1006–1012.
and risk of dementia in two million people over two Stern, Y., Albert, S., Tang, M.X. and Tsai, W.Y. (1999).
decades: A retrospective cohort study. The Lancet Rate of memory decline in AD is related to education
Diabetes and Endocrinology, 3: 431–436. and occupation: Cognitive reserve? Neurology, 53:
Richard, E., Van den Heuvel, E., Moll van Charante, 1942–1947.
E.P. et al. (2009). Prevention of dementia by inten- Stern, Y., Alexander, G.E., Prohovnik, I. and Mayeux, R.
sive vascular care (PreDIVA): A cluster-randomized (1992). Inverse relationship between education and
trial in progress. Alzheimer Disease and Associated parietotemporal perfusion deficit in Alzheimer’s dis-
Disorders, 23: 198–204. ease. Annals of Neurology, 32: 371–375.
Riley, K.P., Snowdon, D.A., Desrosiers, M.F. and Stern, Y., Tang, M.X., Denaro, J. and Mayeux, R. (1995).
Markesbery, W.R. (2005). Early life linguistic ability, Increased risk of mortality in Alzheimer’s disease
late-life cognitive function, and neuropathology: patients with more advanced educational and occupa-
Findings from the Nun Study. Neurobiology of Aging, tional attainment. Annals of Neurology, 37: 590–595.
26: 341–347. Schneider, L.S., Mangialasche, F., Andreasen, N. et al.
Ritchie, C.W., Smailagic, N., Noel-Storr, A.H. et al. (2014). (2014). Clinical trials and late-stage drug development
Plasma and cerebrospinal fluid amyloid beta for the for Alzheimer’s disease: An appraisal from 1984 to
diagnosis of Alzheimer’s disease dementia and other 2014. Journal of Internal Medicine, 275: 251–283.
dementias in people with mild cognitive impairment Schrijvers, E.M., Verhaaren, B.F., Koudstaal, P.J. et al.
(MCI). Cochrane Database of Systematic Reviews, (6): (2012). Is dementia incidence declining? Trends in
CD008782. dementia incidence since 1990 in the Rotterdam
Ritchie, C.W., Molinuevo, J.L., Satlin, A. et al. (2015). The Study. Neurology, 78: 1456–1463.
European Prevention of Alzheimer’s Dementia (EPAD) Scottish Council for Research in Education (1933). The
Programme: An innovative approach to the develop- Intelligence of Scottish Children: A National Survey of
ment of interventions for the secondary prevention an Age-Group. London, United Kingdom: University of
of Alzheimer’s dementia. Lancet Psychiatry, 3 (2): London Press.
179–186. Scottish Council for Research in Education. (1949).
Ritchie, C.W. and Ritchie, K. (2012). The PREVENT study: The Trend of Scottish Intelligence: A Comparison
A prospective cohort study to identify mid-life of the 1947 and 1932 Surveys of the Intelligence of
612 Dementia

Eleven-Year-Old Pupils. London, United Kingdom: Sudlow, C., Gallacher, J., Allen, N. et al. (2015). UK
University of London Press. ­biobank: An open access resource for identifying
Shenkin, S.D., Starr, J.M., Pattie, A. et al. (2001). Birth the causes of a wide range of complex diseases of
weight and cognitive function at age 11 years: The middle and old age. PLoS Medicine, 12: e1001779.
Scottish Mental Survey 1932. Archives of Disease in Valenzuela, M.J. and Sachdev, P. (2006). Brain reserve
Childhood, 85: 189–196. and dementia: A systematic review. Psychological
Siemers, E.R., Sundell, K.L., Carlson, C. et al. (2016). Phase Medicine, 36: 441–454.
3 solanezumab trials: Secondary outcomes in mild van der Burg, J.M., Pijl, H., Campman, Y.J. et al. (2015).
Alzheimer’s disease patients. Alzheimer’s & Dementia: Does midlife obesity really lower dementia risk? Lancet
The Journal of the Alzheimer’s Association, 12 (2): Diabetes and Endocrinology, 3: 499–500.
110–120. Vaudano, E., Vannieuwenhuyse, B., Van Der Geyten,
Skoog, I., Lernfelt, B., Landahl, S. et al. (1996). 15-Year S. et al. (2015). Boosting translational research
longitudinal study of blood pressure and dementia. on Alzheimer’s disease in Europe: The Innovative
Lancet, 347: 1141–1145. Medicine Initiative AD research platform. Alzheimer’s
Smailagic, N., Vacante, M., Hyde, C. et al. (2015). (18) & dementia: The Journal of the Alzheimer’s
F-FDG PET for the early diagnosis of Alzheimer’s Association, 11 (9): 1121–1122.
­disease dementia and other dementias in people with Whalley, L.J. (2012). Spatial distribution and secu-
mild cognitive impairment (MCI). Cochrane Database lar trends in the epidemiology of Alzheimer’s
of Systematic Reviews, (1): CD010632. disease. Neuroimaging Clinics of North America,
Smith, A.D. and Yaffe, K. (2014). Dementia (including 22: 1–10.
Alzheimer’s disease) can be prevented: Statement sup- Whalley, L.J., Dick, F.D. and McNeill, G. (2006). A life-
ported by international experts. Journal of Alzheimer’s course approach to the aetiology of late-onset demen-
Disease, 38: 699–703. tias. The Lancet Neurology, 5: 87–96.
Snowdon, D.A. (2011). Aging with Grace: The Nun Study Whalley, L.J., Murray, A.D., Staff, R.T. et al. (2011). How the
and the Science of Old Age. How We Can all Live 1932 and 1947 mental surveys of Aberdeen schoolchil-
Longer, Healthier and more Vital Lives. London, United dren provide a framework to explore the childhood
Kingdom: Fourth Estate. origins of late onset disease and disability. Maturitas,
Snowdon, D.A., Kemper, S.J., Mortimer, J.A. et al. (1996). 69: 365–372.
Linguistic ability in early life and cognitive function and Whalley, L.J., Starr, J.M., Athawes, R. et al. (2000).
Alzheimer’s disease in late-life. Findings from the Nun Childhood mental ability and dementia. Neurology,
Study. JAMA, 275: 528–532. 55: 1455–1459.
Strachan, D.P. and Sheikh, A. (2004). A life course Wharton, S.B., Brayne, C., Savva, G.M. et al. (2011).
approach to respiratory and allergic diseases. In Epidemiological neuropathology: The MRC Cognitive
Kuh, D., Ben-Shlomo, Y., Ezra, S. (eds.), A Life Course Function and Aging Study experience. Journal of
Approach to Chronic Disease Epidemiology, (Second Alzheimer’s Disease, 25: 359–372.
Edition), pp. 240–259. Oxford, United Kingdom: Zhang, S., Smailagic, N., Hyde, C. et al. (2014). (11)C-PIB-
Oxford University Press. PET for the early diagnosis of Alzheimer’s disease
Strand, B.H., Langballe, E.M., Rosness, T.A. et al. (2015). dementia and other dementias in people with mild
Does midlife obesity really lower dementia risk? Lancet cognitive impairment (MCI). Cochrane Database of
Diabetes and Endocrinology, 3: 498–499. Systematic Reviews, (7): CD010386.
56
Trial designs

SERGE GAUTHIER

56.1 INTRODUCTION treatment, with considerable face validity and impact on


care (Galasko et al., 1995). For example, studies in mild
cognitive impairment (MCI) of the amnestic type may
Any therapy, whether pharmacological or not, requires
demonstrate that the diagnosis of dementia (predomi-
proof of safety and efficacy. This chapter outlines the vari-
nately caused by AD) is delayed by 6 months or longer.
ous trial designs that have been used for the treatment of
Delaying loss of autonomy for self-care and even death
Alzheimer’s disease (AD). The experience gained so far
in moderate-to-severe stages of AD using αtocopherol in
has been predominately in the symptomatic treatment
only one study of the AD Cooperative Study group (Sano
of AD, using parallel group designs over 3–12 months.
et al., 1997) has influenced clinical practice to use vita-
A number of randomized clinical trials (RCT) attempt-
min E in all stages of AD, at least in the United States (see
ing to modify disease progression have been completed
Chapters 54 and 55). Delaying the loss of autonomy for
or are in the process of progression using parallel group
instrumental activities of daily living (ADLs) or the need
designs over 12–18 months. New adaptive and combina-
for nursing home care would reduce the need for support-
tion designs are being proposed to accelerate the approval
ive care. Delaying emergence of some of the behavioural
of new treatments.
and psychological symptoms of dementia (BPSD) would
The natural history of AD is described first (see also
reduce caregiver burden and later patient’s need for nurs-
Chapter 45), introducing the concepts of disease stages,
ing home placement.
disease milestones, symptomatic domains that fluctuate
Symptomatic domains in dementia include cogni-
in intensity as the disease runs its course and biomark-
tion, ADLs and behaviour. One can even add a domain
ers. Lessons from symptomatic studies using cholinester-
of changes in motricity, since patents with AD will mani-
ase inhibitors (ChEIs) and memantine are summarized.
fest some features of parkinsonism late in the disease. In
Current disease modification study designs are then
many patients early changes in mood and anxiety precede
outlined.
the formal diagnosis of AD, sometimes with spontaneous
improvement as insight into the disease is lost. Cognitive
and functional ADL decline are relatively linear over time,
whereas BPSD fluctuations through the course of the dis-
56.2 NATURAL HISTORY OF AD ease resolves spontaneously through the severe stage as
motricity becomes impaired (Gauthier et al., 2001). These
The natural history of AD can be broadly considered as a natural fluctuations in the intensity of individual symp-
presymptomatic stage during which a number of pathologi- tomatic domains through the stages of AD have an impact
cal events take place, an early symptomatic or prodromal into trial design and outcomes (Table 56.2). It should be
stage with affective and/or cognitive manifestations, symp- noted that studies can be of shorter duration and/or of
tomatic mild-to-moderate and severe stages. Each of these smaller numbers of subjects in moderate stage compared
stages could be targeted for specific treatments, requiring to mild stages of AD because of the faster rate of decline
different trial designs and outcomes (Table 56.1). in the former, which may be related to the sensitivity of
Disease milestones have been defined for AD in measurement scales or to the progression of AD in the
Box 56.1. Some of these milestones can be targets for particular patient.

613
614 Dementia

Table 56.1 Examples of trial design and outcomes for each stage of AD

Stage Target population Trial design Primary outcome


Presymptomatic Cognitively normal Survival over 5–7 years Incident dementia
Prodromal Mild cognitive impairment Survival over 3 years Progression to dementia
Mild to moderate AD in the community 6 Months parallel groups Cognition and global
impression of change
Moderate to severe AD in the community or in 6 Months parallel groups ADL and global impression
assisted living of change
Severe AD in institution 6 Months parallel groups Behaviour
Abbreviations: AD, Alzheimer’s disease; ADL, activity of daily living.

BOX 56.1: Clinical milestones in Alzheimer’s disease

●●  mergence of cognitive symptoms


E
●● Progression from amnestic mild cognitive impairment to diagnosable dementia
●● Loss of instrumental activities of daily living (ADLs)
●● Emergence of behavioural and psychological symptoms of dementia
●● Nursing home placement
●● Loss of self-care in ADL
●● Death

Table 56.2 Example of impact of symptoms through stages of AD on trial design and outcomes

Stage Prominent features Types of outcomes Examples


Mild Depression may be present, few BPSD, Cognition, instrumental ADL ADAS-Cog, CDR-SB,
cognitive decline slow but predominant ADCS-ADL, DAD
feature; some instrumental ADL losses
Moderate Cognitive decline more rapid, functional Cognition; instrumental and ADAS-Cog, DAD, ADCS-
decline more rapid; BPSD emerging basic ADL, behaviour ADL, NPI
Severe Cognitive losses harder to measure (floor Cognition, self-care ADL, SIB, ADCS-ADL, NPI, UPDRS
effect); few self-care ADL remaining, behaviour parkinsonism
BPSD abating, parkinsonism emerging
Notes:           BPSD, behavioural and psychological symptoms of dementia; ADAS-Cog, Alzheimer Disease Assessment Scale – Cognitive subscale
(Rosen et al., 1984); CDR-SB, Clinical Dementia Rating Sum of Boxes (Cedarbaum et al., 2013); ADCS-ADL, Alzheimer Disease
Cooperative Study – Activities of Daily Living scale (Galasko et al., 1997); DAD, Disability Assessment for Dementia (Gélinas et al.,
1999); NPI, Neuropsychiatric Inventory (Cummings et al., 1994); SIB, Severe Impairment Battery (Panisset et al., 1994); UPDRS,
Unified Parkinson’s Disease Rating Score (Fahn and Elton, 1987).

of change became the primary outcomes in mild-to-mod-


56.3 SYMPTOMATIC CLINICAL TRIALS erate AD, usually defined operationally as scores between
USING CHOLINESTERASE 10 and 26 on the Mini-Mental State Examination (MMSE).
INHIBITORS AND MEMANTINE Unfortunately, as these FDA guidelines cautioned against the
‘pseudospecificity’ of measurable benefits on neuropsychiat-
The modern treatment of AD was initiated by the report that ric manifestations in AD, they delayed research in this symp-
tacrine improved some aspects of cognition and daily life. tomatic domain. In recent years, discussions and publications
The follow-up confirmatory studies used crossover and par- from the FDA and other regulatory agencies have been more
allel group designs. The US Food and Drug Administration accepting of ADLs and behaviour as important outcomes of
(FDA) published guidelines (Leber, 1990) greatly influenced the former. Guidance is also available for developing drugs
the choice of outcomes for proof of efficacy of drugs improv- for the treatment of early stage disease (US Department of
ing symptoms in AD: a cognitive performance-based scale Health and Human Services, FDA, CDER, 2013).
such as the Alzheimer Disease Assessment Scale – Cognitive The following study designs have been used in the proof
subscale (ADAS-Cog) and an interview-based impression of efficacy for ChEIs: parallel groups over 3–12 months and
Trial designs 615

survival to a predefined clinical end point over 1 year or paving the way for a number of studies, where novel drugs
longer. or placebo are added to ‘standard treatment’.
Parallel groups offer the possibility of short-term (mini-
mum 3 months) studies comparing the efficacy of differ-
ent doses of the drug versus placebo. The primary analysis
56.4 DISEASE MODIFICATION
is done on outcomes at the end of the study using the last
observation carried forward (LOCF) or intent-to-treat (ITT)
STRATEGIES
analyses, which compensate for missing values in case of
dropouts. Although LOCF/ITT has been favoured by the Many studies have been made and others are ongoing
FDA, investigators and other regulatory agencies are now with the hope of arresting or slowing significant disease
suggesting that for studies of 12 months or longer duration, progression. So far they have used parallel group designs
the primary analysis should be on observed cases (OCs), i.e. over 12–18 months in mild-to-moderate AD, with the
completers. For practical purpose, both types of analysis will new drug or a placebo added to standard treatment with a
usually be performed. Although ‘cognitive enhancement’ was ChEI and/or memantine. The primary efficacy outcomes
the main hope for ChEIs as a therapeutic class, the reality has are usually the ADAS-Cog and the Clinical Dementia
emerged from 6-month studies with open-label extensions Rating Sum of Boxes (Hugues et al., 1982; Cedarbaum
and the 1-year placebo-controlled Nordic study (Winblad et et al., 2013) and secondary outcomes include volumetric
al., 2001) is that, although there is a small but statistically sig- brain measurement using magnetic resonance imaging at
nificant improvement in cognition reaching its peak point at the beginning and end of the study, looking for a reduced
3 months with the ChEIs, the most clinically relevant finding rate of brain (whole brain or hippocampus) atrophy in the
has been the stabilization of cognitive decline with ‘return actively treated group compared to placebo. Although,
to baseline’ at 9–12 months for the actively treated groups at this design appears promising, there are uncertainties
the higher therapeutics doses, compared with placebo treated and limitations. For instance, the difference in the rate of
groups who decline steadily. It should be noted that this natu- brain atrophy may be absent or opposite to expectations,
ral decline varies greatly between studies in AD and is even with accelerated atrophy in the actively treated group.
less evident in AD with cerebrovascular disease or in vascu- There may be a discrepancy between a reduced rate of
lar dementia (VaD), where control of vascular risk factors atrophy but no measurable clinical benefit, as was seen in
appear to modify p ­ rogression, at least in studies of 6 months’ some immunotherapy trials. Other biomarkers are being
duration (Schneider, 2003). incorporated in ongoing RCT, some reflecting disease-
Survival studies have primarily looked at loss of ADLs specific pathology, such as amyloid-positron emission
and have successfully demonstrated a delay in the loss of tomography (PET) ligands and cerebrospinal fluid (CSF)
autonomy for patients on ChEIs compared with placebo. aß42 levels, others reflecting neurodegeneration, such as
Parallel group 6-month studies in patients ranging from PET-fluorodeoxyglucose (PET-FDG) and CSF tau levels
mild-to-moderately severe AD (MMSE 5-26) have also (Langbaum et al., 2013). The usefulness of biomarkers for
established that ADLs are more stable during treatment purpose of enrichment of study populations or as second-
compared to placebo, but show no reversibility for lost ary efficacy outcomes is periodically reviewed by interna-
instrumental ADLs. tional work groups (Carrillo et al., 2013).
The most difficult domain to study, although very signifi- One difficult issue in disease modification strategies is
cant clinically, has been behaviour. Despite the availability of the decision of the stage of disease, where the proposed
general BPSD scales such as the Neuropsychiatric Inventory drug is most likely to work. On the ‘proof-of-concept’
(NPI), as well as specific scales such as Cohen-Mansfield Phase II/III efficacy and safety study hinges on the entire
Agitation Inventory (Cohen-Mansfield et al., 1989), there is future of the drug. For example, numerous attempts at
yet little proof of drug efficacy against BPSD. New methods treating patients with AD in mild-to-moderate stages
of analysis of behaviour have been proposed (Gauthier et al., using non-steroidal anti-inflammatory drugs (NSAIDs)
2008; Geda et al., 2013) and will likely be more successful have failed, despite the weight of evidence from epide-
in defining categories of BPSD symptoms most responsive miological research and the biological plausibility of an
to ChEIs (anxiety, hallucinations), to memantine (agitation) inflammatory response to β amyloid deposition. It may
and possibly to other drugs. be that treatment in the late presymptomatic or in the
Memantine has been found to be effective in a range of prodromal stages would be the most appropriate time to
studies using parallel groups, in moderate-to-severe AD. initiate treatment. On the other hand, studies in these
Scales appropriate for this stage of disease, such as SIB, the stages of AD would require 3–5 years. Alternative subjects
Alzheimer Disease Cooperative Study – Activities of Daily are being studied for ‘proof-of-concept’ using high risk
Living scale (ADCS-ADL) and the NPI have been used and groups, such as presenilin or amyloid precursor protein
accepted by the FDA and other regulatory agencies. The mutation carriers (see Chapter 50), subjects homozygous
trial design of adding memantine or placebo to a stable with apolipoprotein E ε4 or asymptomatic but amyloid
dose of ChEI has been used successfully (Tariot et al., 2004), positive on PET scan (Langbaum et al., 2013).
616 Dementia

Cohen-Mansfield, J., Marx, M.S. and Rosenthal, A.S.


56.5 FUTURE STRATEGIES TO DELAY (1989). A description of agitation in a nursing home.
EMERGENCE OF AD Journal of Gerontology, 44 (3): M77–M84.
Cummings, J.L., Mega, M. and Gray, K. et al. (1994).
As hypotheses on the pathophysiology of AD emerges Neuropsychiatric Inventory: Comprehensive assess-
from epidemiological research in human populations, ment of psychopathology in dementia. Neurology, 44:
post-mortem and biomarker studies in patients and animal 2308–2314.
models, we will need to establish whether new therapies DeKosky, S.T., Williamson, J.D., Fitzpatrick, A.L. et al.
can delay the onset of symptoms in asymptomatic people (2008). Ginkgo biloba for prevention of dementia.
at varying degrees of risk of AD. The prototype of trial A randomized controlled trial. JAMA, 300: 2253–2262.
designs to establish the safety and efficacy of such thera- Fahn, S. and Elton, R. (1987). United Parkinson’s disease
pies is the 5- to 7-year studies comparing Ginkgo biloba rating scale. In S. Fahn, S., Marsden, C., Golstein, M.
to placebo in elderly subjects with or without memory et al. (eds.), Recent Development in Parkinson’s Disease.
complaints but no measurable impairment with incident Florham Park, NJ: Macmillan Health Care, pp. 153–163.
dementia as primary end point was noticed (Vellas et al., Galasko, D., Bennett, D., Sano, M. et al. (1997). An i­nventory
2006; DeKosky et al., 2008). Variations of this design are to assess activities of daily living for ­clinical trials in
possible, by enriching the study population with different Alzheimer’s disease. Alzheimer Disease and Associated
levels of risk, such as a positive family history of AD and/ Disorders, 11 (Suppl. 2): S33–S39.
or selected gene or biological markers, although enrich- Galasko, D., Edland, S.D., Morris, J.C. et al. (1995). The
ment of a study population will limit the applicability of Consortium to Establish a Registry for Alzheimer’s
findings to the population as a whole. There are also ethi- Disease (CERAD). Part IX. Clinical milestones in
cal considerations in using potentially harmful treatments patients with Alzheimer’s disease followed over 3
on persons who may never progress to dementia in their years. Neurology, 45: 1451–1455.
lifetime (Peters et al., 2013). Gauthier, S., Loft, H. and Cummings, J. (2008).
Simultaneous multiple interventions may be required Improvement in behavioral symptoms in patients with
to obtain benefit in AD: this is being studied using non- moderate to severe Alzheimer’s disease by meman-
pharmacological intervention such as cognitive training, tine: A pooled data analysis. International Journal of
physical exercise and diet enrichment (Kivipelto et al., 2013; Geriatric Psychiatry, 23: 537–545.
Vellas et al., 2014), cognitive training and psychosocial inter- Gauthier, S., Thal, L.J. and Rossor, M.N. (2001). The future
ventions (Bier et al., 2014). The combination of pathological diagnosis and management of Alzheimer’s disease. In
factors in AD (amyloid toxicity, tau pathology, inflamma- Gauthier, S. (ed.), Clinical Diagnosis and Management
tory responses) may require combinations of drugs in a way of Alzheimer’s Disease. London, United Kingdom:
similar to cancer chemotherapy. This will need to be care- Martin Dunitz, pp. 369–378.
fully thought through with participation of clinical trialists, Geda, Y.E., Schneider, L.S., Gitlin, L.N. et al. (2013).
regulators and patients’ representatives. Neuropsychiatric symptoms in Alzheimer’s dis-
ease: Past progress and anticipation of the future.
Alzheimer’s and Dementia: The Journal of the
REFERENCES Alzheimer’s Association, 9: 602–608.
Gélinas, I., Gauthier, L., McIntyre, M. and Gauthier, S.
Bier, N., Grenier, S., Brodeur, C. et al. (2014). Measuring (1999). Development of a functional measure for
the impact of cognitive and psychosocial interven- persons with Alzheimer’s disease: The Disability
tions in persons with MCI with a randomized single- Assessment for Dementia. American Journal of­
blind controlled trial: Rationale and design of the Occupational Therapy, 53: 471–481.
MEMO+ study. International Psychogeriatrics, 27 Hugues, C.P., Berg, L., Danziger, W.L. et al. (1982). A
(03): 511–525. new clinical scale for the staging of dementia. British
Carrillo, M.C., Rowe, C.C., Szoeke, C. et al. (2013). Journal of Psychiatry, 140: 566–572.
Research and standardization in Alzheimer’s trials: Kivipelto, M., Solomon, A., Ahtiluoto, S. et al. (2013).
Reaching international consensus. Alzheimer’s and The Finnish Geriatric Intervention Study to
Dementia: The Journal of the Alzheimer’s Association, Prevent Cognitive Impairment and Disability (FINGER):
9 (2): 160–168. Study design and progress. Alzheimer’s and Dementia:
Cedarbaum, J.M., Jaros, M., Hernandes, C. et al. (2013). The Journal of the Alzheimer’s Association, 9 (6):
Rationale for the use of the Clinical Dementia Rating 657–665.
Scale sum of boxes as a primary outcome measure Langbaum, J.B., Fleisher, A.S., Chen, K. et al. (2013).
for Alzheimer’s disease clinical trials. Alzheimer’s and Ushering in the study and treatment of preclinical
Dementia: The Journal of the Alzheimer’s Association, Alzheimer’s disease. Nature Reviews, Neurology, 9 (7):
9: S45–S55. 371–381.
Trial designs 617

Leber, P. (1990). Guidelines for Clinical Evaluation of Tariot, P.N., Farlow, M.R., Grossberg, G.T. et al. (2004).
Antidementia Drugs. Washington, DC: US Food and Memantine treatment in patients with moderate to
Drug Administration. severe Alzheimer disease already receiving donepezil:
Panisset, M., Roudier, M., Sexton, J. et al. (1994). Severe A randomized controlled trial. JAMA, 291 (3): 317–324.
Impairment Battery: A neuropsychological test for US Department of Health and Human Services, FDA,
severely demented patients. Archives of Neurology, CDER (2013). Guidance for Industry Alzheimer’s
51: 41–45. Disease: Developing Drugs for the Treatment of Early
Peters, K.R., Beattie, B.L., Feldman, H.H. and Illes, J. Stage Disease. Silver Spring, MD: U.S. Department of
(2013). A conceptual framework and ethics analysis for Health and Human Services, FDA, CDER.
prevention trials of Alzheimer’s disease. Progress in Vellas, B., Andrieu, S., Ousset, P.J. et al. (2006). The
Neurobiology, 110: 114–123. GuiAge study. Methodological issues. A 5-year dou-
Rosen, W.G., Mohs, R.C. and Davis, K.L. (1984). A new ble-blind randomized trial of the efficacy of EGb 761
rating scale for Alzheimer’s disease. The American for prevention of Alzheimer disease in patients over 70
Journal of Psychiatry, 141: 1356–1364. with a memory complaint. Neurology, 67: S6–S11.
Sano, M., Ernesto, C., Thomas, R.G. et al. (1997). A con- Vellas, B., Carrie, I., Gillette-Guyonnet, S. et al. (2014).
trolled trial of selegiline, alpha-tocopherol, or both as MAPT study: A multidomain approach for preventing
treatment for Alzheimer’s disease. The Alzheimer’s Alzheimer’s disease: Design and baseline data. The
Disease Cooperative Society. The New England Journal of Prevention of Alzheimer’s Disease, 1: 1–12.
Journal of Medicine, 326 (17): 1216–1222. Winblad, B., Engedal, K., Soininen, H. et al. (2001). A
Schneider, L.S. (2003). Cholinesterase inhibitors 1-year, randomized, placebo-controlled study of done-
for ­v ascular dementia? The Lancet Neurology, 2: pezil in patients with mild to moderate AD. Neurology,
658 – 659. 57 (3): 489–495.
4
Part    

The Overlap and Interaction


between Alzheimer’s Disease and
Cerebrovascular Disease

57 Vascular factors and Alzheimer’s disease 619


Robert Stewart
57
    

Vascular factors and Alzheimer’s disease

ROBERT STEWART

rather than an assumed sole underlying cause (vascular


57.1 VASCULAR FACTORS IN dementia or ‘vascular neurocognitive disorder’). Depressive
ALZHEIMER’S DISEASE disorder after bereavement may have different features from
depression occurring in other circumstances, but this does
The increasing acceptance of vascular factors in Alzheimer’s not mean that ‘bereavement depression’ is necessarily help-
disease (AD) has challenged traditional dementia sub-­ ful as a diagnosis.
classification. In particular, the assumption that AD and A more fundamental challenge to understanding the
vascular dementia are always distinguishable clinical disor- role of vascular factors in AD is the limited interface
ders in late-onset dementia has become untenable because between risk factor outcome research and research into
of the large pathological overlap and poor correlations underlying causal pathways. AD can be considered as, in
between diagnosis and pathology. The sub-classification effect, two disorders – first, a process of nerve cell death
of dementia according to presumed Alzheimer or vascular with particular appearances at a cellular level, which can
aetiology emerged in the 1960s when dementia was occur- only be observed at autopsy, and the second, a clinical dis-
ring earlier in life, when treatment of vascular risk factors order characterized by its symptoms and clinical course.
was limited and when florid cerebrovascular disease was fre- AD the pathological disorder is likely to be have been
quently present at post-mortem – i.e. could be assumed to be present for a decade or two before AD the clinical disorder
a single underlying cause. Dementia cases today, particularly becomes apparent. However, it is important to bear in mind
in high-income nations, are arising much more frequently that sizeable numbers of people have significant Alzheimer
in later old age (e.g. ninth and tenth decades) where mixed pathology with no clinical evidence of dementia and/or
preserved cognitive function (Neuropathology Group of
pathology is the norm. Developments in neuroimaging have
the Medical Research Council Cognitive Function and
been a mixed blessing: the ability to detect more minor levels
Ageing Study, 2001; Jack et al., 2014). Furthermore, these
of vascular disturbance is undoubtedly useful in investigat-
pathological changes more poorly predict dementia in
ing causal pathways in a research context; however, detect-
later old age despite similar associations with cerebral
able ‘abnormalities’ are now more likely to be coincidental
atrophy (Savva et al., 2009), suggesting that links between
than causal, particularly in older age groups, which needs to
‘traditional’ pathologies and cognitive decline are more
be borne in mind in clinical decision making.
complex than originally envisaged.
‘Vascular dementia’ as a diagnosis implies a single cause
and a discrete outcome. However, this has become hard
to sustain as a useful entity because of evidence that vas-
cular factors frequently influence dementia occurrence in
combination with other pathology (principally AD) – i.e.
57.2 VASCULAR FACTORS AND
because most dementia is ‘mixed’. The fact that people can CLINICAL AD
be identified with ‘pure’ AD or vascular dementia does not
support separate diagnoses if most cases sit somewhere in There is a substantial body of prospective epidemiological
the middle with combinations of underlying pathologies. research supporting a role for vascular risk factors in the
Cerebrovascular disease might influence the course of aetiology of AD. Identified risk factors include large vessel
dementia or its manifestations but would be better described atherosclerosis as estimated by carotid ultrasound or the
as a comorbidity (dementia with cerebrovascular disease) ankle:brachial blood pressure index (Hofman et al., 1997),
619
620 Dementia

conventional risk factors for cerebrovascular disease such as hypotension. Of relevance, relative weight loss also occurs
hypertension (Skoog et al., 1996b; Kivipelto et al., 2001), type prior to the onset of dementia over a similar period (Stewart
2 diabetes (Launer, 2009) and atrial fibrillation (Ott et al., et al., 2005), which may reflect similar underlying processes.
1997). ‘Lifestyle’-related risk factors remain a little more con- However, it is also possible that late-life hypotension might
tentious, with reasonable evidence for raised cholesterol lev- be a consequence of earlier sustained hypertension since
els (Notkola et al., 1998; Kivipelto et al., 2001; Anstey et al., dementia-associated decline was modified by antihyperten-
2008; Solomon et al., 2009) and smoking (Anstey et al., 2007; sive treatment in one study (Stewart et al., 2009). This has
Beydoun et al., 2014; Zhong et al., 2015), but more equivocal been supported by findings from low- and middle-income
evidence for obesity (Gustafson et al., 2003; Hassing et al., countries, where hypertension is substantially less common
2009; Albanese et al., 2015; Qizilbash et al., 2015). They also and where associations between dementia and low blood
include risk factors such as homocysteine in some studies pressure were, if present at all, much weaker than those
(Clarke et al., 1998; Seshadri et al., 2002; Beydoun et al., 2014), reported in higher-income settings (Albanese et al., 2013a),
insulin resistance (Kuusisto et al., 1997) and the metabolic despite comparable associations with markers of weight loss
syndrome (Razay et al., 2007) and protective factors such as (Albanese et al., 2013b).
moderate alcohol intake (Ruitenberg et al., 2002), increased Associations between dementia and prior cholesterol
physical activity (Laurin et al., 2001; Scarmeas et al., 2009; levels appear to show a similar pattern to those with blood
Beydoun et al., 2014; Blondell et al., 2014) and Mediterranean pressure – i.e. with relatively high midlife levels but rela-
diet (Scarmeas et al., 2009) where other causal pathways may tively low or normal contemporaneous levels (Notkola
be involved, but whose associations with AD may have a vas- et al., 1998) and declining levels as a predictor of dementia
cular component. (Solomon et al., 2007). One study found that this decline
occurred approximately 10–15 years prior to the clinical
onset, and then followed a parallel course (Stewart et al.,
2007), suggesting that a fall in cholesterol levels might be
57.3 CHANGES IN VASCULAR RISK a marker of another event (e.g. inflammatory or infective)
FACTORS PRIOR TO DEMENTIA associated with very early AD pathology, although this find-
ing requires replication and further investigation. Changes
There is now also substantial evidence that associations in folate, B12 and homocysteine levels have also been
between vascular factors and dementia change over time, described around the clinical onset of dementia (Kim et al.,
even reversing in some instances. This has been described 2008), so the general picture emerges of a range of physi-
in most detail for blood pressure. Raised blood pressure in cal changes occurring around this time that do need to be
midlife is associated with later cognitive impairment and borne in mind in clinical settings when diagnoses are being
dementia (Elias et al., 1993; Launer et al., 1995). However, assigned and pre-existing risk factors evaluated (because
this has tended to be only apparent in studies with follow-up these earlier risk factors may now no longer be apparent).
periods of 10 years or more. Studies with shorter follow-up From a research perspective there are two key implications:
often find no association (Posner et al., 2002), and cross- (1) a need for further investigation of ‘frailty’ and the extent
sectional studies frequently show associations between to which dementia is linked with wider accelerated ageing
dementia and low rather than high blood pressure (Qiu processes and (2) caution about inferring causality from
et al., 2005). This relative hypotension also seems to become short-term predictive relationships.
more marked at more advanced stages of dementia (Guo
et al., 1996). Although a younger age at which blood pres-
sure is measured has been suggested as increasing the likeli-
57.4 INTERACTIONS WITH APOE
hood of a positive association with later dementia (Qiu et al.,
2005), an analysis of 15-year follow-up data on people aged
GENOTYPE
70 at baseline also found that those with AD at the end of
the period had raised systolic blood pressure at the start of The apolipoprotein E (APOE) e4 allele was first recognized
the study but lower blood pressure by the time dementia had for its associations with cholesterol metabolism and in some
developed (Skoog et al., 1996b). The interval between mea- studies has been associated with stroke (Margaglione et al.,
surement of blood pressure and ascertainment of demen- 1998; McCarron et al., 1999). However, there has been no
tia therefore seems to be more important. Longitudinal evidence that cholesterol levels or other vascular factors
studies suggest that blood pressure declines over at least explain the association between e4 and AD (Slooter et al.,
3–6 years prior to the clinical onset of dementia (Qiu et al., 1999; Prince et al., 2000). The focus instead has shifted to
2004; Stewart et al., 2009), as well as an exaggerated mid- investigating interactions between vascular factors and e4
to late-life increase in blood pressure preceding this decline as risk factors, although findings have been conflicting.
(Stewart et al., 2009). One community study found that atherosclerosis was more
One possible explanation for this late decline in blood strongly associated with AD in the presence of APOE e4
pressure is that early neurodegenerative disease affects (Hofman et al., 1997), but later examinations in the same
regulating centres (Burke et al., 1994) resulting in relative study did not support this (Slooter et al., 1999), although
Vascular factors and Alzheimer’s disease 621

other studies have reported similar interactions for cerebro- ischaemia due to small-vessel occlusion or hypoperfusion
vascular events (Johnston et al., 2000) and atherogenic diet may underlie the demyelination and neuronal loss also
(Laitinen et al., 2006). On the other hand, insulin resistance observed. Whatever the pathology, WMHs clearly repre-
and smoking have been found to be more strongly associ- sent subclinical cerebrovascular disease in many cases, as
ated with AD in people without e4 (Kuusisto et al., 1997; Ott they are associated with other risk factors for cerebrovas-
et al., 1998; Zhong et al., 2015), and because interactions are cular disease such as hypertension (Breteler et al., 1994b;
frequently not reported when absent, it is difficult to exclude Liao et al., 1997; Dufouil et al., 2001), smoking (Liao et al.,
publication bias. 1997), hypercholesterolaemia (Breteler et al., 1994b) and
atherosclerosis (Bots et al., 1993). In community samples,
they are associated with worse cognitive function (Breteler
57.5 POTENTIAL UNDERLYING et al., 1994a; Kuller et al., 1998) in people both with and
MECHANISMS – THE ROLE OF without dementia (Skoog et al., 1996a). However, although
WMHs on post-mortem imaging were found to improve
STROKE AND CEREBROVASCULAR
the prediction of dementia in life (Fernando et al., 2004),
DISEASE they are common findings in older populations and may
be present at an apparently severe degree without detect-
To what extent are the associations between vascular risk able evidence of cognitive impairment (Fein et al., 1990).
factors and dementia accounted for by stroke or subclinical WMHs, therefore, appear to be insufficient in isolation to
cerebrovascular disease? Most studies of AD as an outcome give rise to dementia and are likely to require interactions
have divided these cases into those with and without clini- with other pathology.
cal evidence of co-occurring cerebrovascular disease, and
most have found that associations with vascular risk factors
remain in the less mixed group. An exception may be atrial
57.6 POTENTIAL UNDERLYING
fibrillation, which appears to be particularly linked with
MECHANISMS – VASCULAR
stroke-associated dementia (Ott et al., 1997; Kwok et al.,
2011). Furthermore, studies of clinical stroke and cogni-
PATHOLOGY IN AD
tive impairment or dementia have tended to support mixed
pathology. Although dementia is unusual following a sin- Epidemiological research, as described, supports a role for
gle stroke (Srikanth et al., 2004; Pendlebury and Rothwell, conventional vascular risk factors in the aetiology of clin-
2009), post-stroke dementia frequently follows an AD-like ical AD. Parallel neurobiological research has suggested
clinical course (Kokmen et al., 1996) and post-stroke cogni- that vascular processes may be an important feature of
tive decline is more strongly predicted by medial temporal AD at a pathological level. Traditionally, research into the
atrophy than the level of cerebrovascular disease (Firbank role of amyloid in AD has focused on its deposition in
et al., 2012). Furthermore, a history of pre-stroke cognitive parenchymal neuritic plaques. However, other pathology
decline is present in many cases of post-stroke dementia associated with the disorder has long been recognized.
(Hénon et al., 2001); cognitive impairment is a risk factor for Cerebral amyloid angiopathy (CAA) describes the depo-
later first onset of stroke (Ferucci et al., 1996). Post-stroke sition of β-amyloid, among other proteins, in the walls
dementia in turn predicts an increased risk of stroke recur- (vascular smooth muscle cell layer) of cortical vessels.
rence (Moroney et al., 1997). These findings suggest that CAA increases with age and is associated with dementia
the stroke episode itself, although undoubtedly having an in population-based studies (Keage et al., 2009). It is also
impact on cognitive function, may frequently occur in the recognized to be a risk factor for cerebrovascular disorders
context of more insidious and gradual decline, suggesting such as small and large vessel haemorrhage, leukoenceph-
co-occurring AD in a substantial number of cases. This pic- alopathy and infarction and is a feature of some familial
ture may change over the period following a stroke, with early-onset dementia syndromes. CAA is therefore a fea-
early cases of dementia following an AD-like picture and ture of AD and was also associated with cognitive impair-
later cases showing a more typical vascular dementia syn- ment in a community-based autopsy series (Pfeiffer et al.,
drome (Altieri et al., 2004). 2002). One possible mechanism underlying its role in AD
There is substantial evidence that vascular and is that impaired amyloid clearance across the blood–brain
Alzheimer pathologies frequently co-occur. A ­community- barrier (potentially influenced by APOE genotype) may
based neuropathological study found that vascular pathol- give rise to CAA, which in turn compromises vessel wall
ogy was frequently present in dementia and that ‘pure’ AD integrity and gives rise to cerebrovascular disorders, fur-
occurred only in approximately 20% of cases (Fernando ther exacerbating blood–brain barrier function and AD
et al., 2004). In life, white matter hyperintensities (WMHs) pathology (Bell and Zlokovic, 2009; Burgmans et al.,
on magnetic resonance imaging (MRI) are frequently 2013). APOE e4 has also been proposed as underlying at
found in association with AD, although this is by no means least some of the observed co-occurrence of small vessel
a characteristic feature. Arteriosclerotic changes are often disease and vascular amyloid deposition, in combination
seen in association with white matter alterations, and with blood–brain barrier disturbance (Grinberg and Thal,
622 Dementia

2010), and microbleeds have been suggested as a common AGEs have been found to be associated with cognitive
consequence (Hommet et al., 2011). Vascular abnormali- impairment and decline, supporting a relationship
ties in AD are not restricted to arterial and arteriolar amy- (Yaffe et al., 2011).
loid deposition, but are also seen in cerebral microvessels
and capillaries. Microangiopathic changes include base-
ment membrane thickening and collagen accumulation
(Farkas et al., 2000), as well as smooth muscle cell irregu- 57.8 POTENTIAL UNDERLYING
larities, endothelial degeneration and other abnormalities MECHANISMS – EXACERBATION
(Kalaria and Skoog, 2002). Underlying causal pathways OF COGNITIVE IMPAIRMENT AND
might include toxic effects of accumulating amyloid ACCELERATED AGE OF ONSET
(Smith and Greenberg, 2009) possibly exacerbated by the
microangiopathy itself (de la Torre and Mussivand, 1993; The mechanisms above concern direct influences of
Preston et al., 2003). vascular factors on the pathological processes that are
believed to underlie AD. However, clinical AD might still
be ‘caused’ without any direct interaction at a pathologi-
57.7 POTENTIAL UNDERLYING cal level and instead by interaction in clinical manifesta-
MECHANISMS – PATHWAYS FOR tions. Alzheimer pathology develops slowly: possibly over
VASCULAR ‘INDUCTION’ OF AD 10–20 years or more. The chance of someone developing
dementia therefore depends a great deal on the extent to
which a given level of pathology affects cognition and
The neuropathological findings summarized above suggest on the extent to which a person can compensate for this.
that vascular abnormalities are a feature of AD pathology Alzheimer pathology begins in the hippocampus and
and may be important in its clinical expression. Several manifests in its early stages as declining memory func-
pathways have been proposed, which could potentially tion. Mild levels of cerebrovascular disease on the other
explain an association between vascular risk factors and AD hand would be expected to exert most damage on deep
and which concern direct causal links at the level of patho- white matter and on fronto-­subcortical connections. The
logical processes: consequent disruption of executive functions such as
selective attention and mental flexibility may be enough
Ischaemic injury and amyloid deposition: β-amyloid to ‘unmask’ early AD, which would not otherwise have
production appears to be a non-specific response to become apparent for several years. Vascular factors may
cerebral trauma but has been described in particular therefore accelerate the age of onset of clinical AD and
in the hippocampi of rodents after severe but tran- precipitate a dementia syndrome at relatively early patho-
sient ischaemia (Kogure and Kato, 1993), providing a logical stages.
potential link between ischaemia/infarction and AD
pathology (Pluta et al., 2013).
Inflammation: Inflammatory processes are important
in mediating cerebral damage following stroke 57.9 POTENTIAL UNDERLYING
(Emsley and Tyrrell, 2002). Inflammation is also MECHANISMS – EFFECTS ON
an important feature of AD pathology (Halliday et
‘FUNCTIONAL RESERVE’
al., 2000) and might underlie links between the two
processes.
The blood–brain barrier: Increased permeability of the The ‘onset’ of dementia is defined, at least in research set-
blood–brain barrier has been found in association tings, as the point where declining cognitive function
with transient ischaemia, and this has been suggested starts to interfere with daily functioning. This is an unreli-
to underlie both perivascular amyloid deposition and able definition of ‘incidence’ since it involves considerable
white matter changes (Grinberg and Thal, 2010). subjective judgement on the part of the participant, their
Abnormal protein glycation: Advanced glycation end family and the research worker. It also assumes a process
products (AGEs) are metabolic oxidation products that can be isolated from all the other factors influencing
associated with diabetes and hyperglycaemia (Vlassara daily functioning in older people. A given level of cognitive
et al., 1992). Their associations with ­neurofibrillary impairment may be more likely to be classified as problem-
tangles and neuritic plaques provide a potential atic if daily function is already impaired – e.g. because of
connection between diabetes and AD (Stewart and motor system impairment due to cerebrovascular disease.
Liolitsa, 1999), although more recent attention has Or, from the opposite perspective, a person in good physical
also focused on the potential role of the AGE ­receptor, health may be able to sustain a greater degree of cognitive
which also transports amyloid-β from the blood to decline before this can be said to be problematic. Clearly,
the brain, and its potential regulation in hypo- and cerebrovascular disease may have an influence on this com-
hyperglycaemic states (Prasad et al., 2014). Peripheral ponent of ‘reserve’.
Vascular factors and Alzheimer’s disease 623

57.10 COMMON UNDERLYING 57.11 DIRECTION OF CAUSE AND


FACTORS EFFECT

Associations between vascular factors and AD need not Since vascular risk factors emerge in midlife and a consid-
imply direct causal links between the two but might, at erable period before AD incidence begins to increase, it is
least in part, arise from common underlying processes. generally assumed that any causal processes are directed
APOE genotype has been considered in this respect, from the vascular risk factor to AD. However, this does not
although, as discussed earlier, it has not been found to exclude additional adverse effects of AD on the vascula-
explain substantially the co-occurrence of cerebrovas- ture at a later stage. Links between vascular and Alzheimer
cular disease and AD. Both AD and cerebrovascular pathology have been described above and may be complex
disease are ‘disorders of ageing’ – that is, they become and bidirectional. Vascular factors might, therefore, be
more common with increased age. However, ‘ageing’ as both a cause and a consequence of AD. At a clinical level,
a process is not synonymous with increased chronologi- it is likely that worsening cognitive function due to early
cal age and there is substantial individual variation in the AD may have adverse effects on risk for vascular disease,
particularly through difficulties with treatment adherence
physiological changes that occur in the transition from
because of declining cognitive function, creating a vicious
mid- to late life. ‘Ageing’ is likely to be influenced by a
cycle of decline. This has been suggested for diabetes self-
variety of factors, both genetic and acquired. Many of
management (Tran et al., 2014), with some supportive evi-
these are unknown – e.g. genetic factors common to dis-
dence cited below, but remains under-investigated for other
eases of ageing – and others may be unquantifiable, such
risk factors.
as lifetime exposure to oxidative stress. Dementia is con-
ventionally emphasized as a disorder that is distinct from
‘normal ageing’. However, it would be surprising if there
were not processes that mediate vulnerability to ageing
processes whether these are experienced in the brain
57.12 IMPLICATIONS FOR PREVENTING
parenchyma or in the vasculature. Finally, both vascular
DEMENTIA
disorders and AD can be said to have common origins
in social adversity: increased risk of AD being associated This chapter has focused on evidence for the involvement of
with lower education and ‘cognitive reserve’; cardiovas- vascular factors in AD, and on processes that might underlie
cular risk factors known to track with social disadvan- these associations. Clearly, there are potentially important
tage across the life course. These processes are difficult to implications for both prevention and treatment of dementia.
evaluate directly; however, they do predict that societies A reduction in vascular risk across a population would in the-
with improving socio-economic status will experience ory reduce the risk of later dementia, whether vascular risk
dementia with a later onset, and lower age-specific preva- factors were acting on AD pathology or reducing ­cognitive/
lence, for which there is at least some emerging evidence functional reserve. However, it is doubtful whether the
(Matthews et al., 2013). effectiveness of this approach can ever be demonstrated in a
A more specific factor that might link both vascular dis- standard randomized controlled trial design given the very
long period over which the risk factors may exert their effect.
ease and AD is insulin resistance, recognized to underlie the
This is likely to be particularly the case for hypertension and
clustering of vascular disorders such as hypertension, type 2
may well account for the lack of evidence concluded by the
diabetes mellitus and hypercholesterolaemia (Reaven, 1988),
most recent Cochrane review of antihypertensive treatment
all of which have been found to be associated with AD, as
to prevent cognitive impairment and dementia for people
mentioned. Insulin levels themselves could also have direct
without cerebrovascular disease (McGuinness et al., 2009b).
effects on cognitive function, given that insulin receptors The same challenge could explain similarly negative find-
are dense in the hippocampus (Marks et al., 1991), although ings for dementia prevention trials of statins (McGuinness
the role of insulin in brain function remains to be elucidated et al., 2009a). Of preventative interventions for cognitive
(Gray et al., 2014). Several epidemiological studies have inves- decline, increased physical activity currently has the most
tigated markers of insulin resistance in association with support from randomized controlled trials (Kramer et al.,
dementia/AD risk and findings include associations with 1999; Lautenschlager et al., 2008).
hyperinsulinaemia (Kuusisto et al., 1997; Luchsinger et al., Prevention of cognitive decline in diabetes is a more read-
2004; Muller et al., 2007), metabolic syndrome (Razay et al., ily assessable objective, although evidence is still lacking.
2007) and impaired midlife insulin secretion (Rönnemaa et Cognitive impairment has been found to be associated with
al., 2008). One potential causal link is in competition between earlier age of onset, longer duration, insulin treatment and
insulin and amyloid beta for insulin degrading enzyme (Craft presence of complications (Roberts et al., 2008), and higher
and Watson, 2004; Qiu and Folstein, 2006), but empirical evi- HbA1c has been associated with cognitive impairment in
dence for mediating pathways remains lacking. people with diabetes (Cukierman-Yaffe et al., 2009; Yaffe
624 Dementia

et al., 2012). In contrast, potential risks of hypoglycaemia close attention. In terms of formal and informal social care,
have been highlighted (Dominquez et al., 2010), and severe people with AD will often also have clinically apparent cere-
hypoglycaemic episodes have been found to be a risk factor brovascular disease that may already be affecting non-cog-
for dementia incidence (Whitmer et al., 2009). More specifi- nitive areas of functioning, increasing the level of support
cally, a two-way relationship was found in the Health ABC required. Vascular factors may have an important impact
Study (Yaffe et al., 2013) with hypoglycaemic episodes both on quality of life for people with AD and their caregivers,
predicting and being predicted by dementia. In the Action regardless of whether relationships with AD reflect cause,
to Control Cardiovascular Risk in Diabetes–Memory IN effect or coincidence. This aspect of the interface continues
Diabetes (ACCORD-MIND) trial of intensive glycaemic to receive scant attention but is clearly an important consid-
control in type 2 diabetes mellitus, no difference between eration in day-to-day clinical practice.
randomization groups was found in the primary cognitive
outcome (Launer et al., 2011), although total brain volume
was higher in the intervention group, and further analysis REFERENCES
indicated intervention-associated deceleration in a particular
(frontal and temporal) distribution of volume loss that was Albanese, E., Davis, B., Jonsson, P.V. et al. (2015).
also associated with longer duration of diabetes, but not with Overweight and obesity in midlife and brain structure
HbA1c (Erus et al., 2015). Cognitive decline in the full cohort and dementia 26 years later: The AGES-Reykjavik Study.
was predicted by depression (Sullivan et al., 2013) and reti- American Journal of Epidemiology, 181: 672–679.
nopathy (Hugenschmidt et al., 2014), but baseline cognitive Albanese, E., Lombardo, F.L., Prince, M.J., and Stewart, R.
impairment also predicted worse diabetes outcomes: specifi- (2013a). Dementia and lower blood pressure in Latin
cally, higher risk of severe hypoglycaemia (Punthakee et al., America, India and China – A 10/66 cross-cohort study.
2012). The picture in diabetes, therefore, appears to be one Neurology, 81: 228–235.
of bidirectional, potentially mutually exacerbating effects of Albanese, E., Taylor, C., Siervo, M. et al. (2013b). Dementia
poor control leading to cognitive impairment leading to fur- severity and weight loss: A comparison across eight
ther worsening of control. cohorts. The 10/66 study. Alzheimer’s & Dementia:
Journal of the Alzheimer’s Association, 9: 649–656.
Altieri, M., Di Piero, V., Pasquini, M. et al. (2004). Delayed
poststroke dementia: A 4-year follow-up study.
57.13 IMPLICATIONS FOR TREATING Neurology, 62: 2193–2197.
DEMENTIA Anstey, K.J., Lipnicki, D.M. and Low, L.F. (2008).
Cholesterol as a risk factor for dementia and cognitive
Implications for treating AD (i.e. reducing cognitive/­ decline: A systematic review of prospective stud-
functional decline after diagnosis) through modifying vas- ies with meta-analysis. American Journal of Geriatric
cular risk depend on whether there are direct effects on Psychiatry, 16: 343–354.
neurodegenerative processes (i.e. whether vascular risk fac- Anstey, K.J., von Sanden, C., Salim, A. and O’Kearney, R.
tors continue to influence cognitive decline rather than sim- (2007). Smoking as a risk factor for dementia and cog-
ply advancing the age of ‘dementia’ onset). The presence of nitive decline: A meta-analysis of prospective studies.
cerebrovascular disease has been associated with slower pro- American Journal of Epidemiology, 166: 367–378.
gression of dementia (Bruandet et al., 2009), as has comorbid Bell, R.D. and Zlokovic, B.V. (2009). Neurovascular mecha-
diabetes (Mielke et al., 2007; Sanz et al., 2009), which might nisms and blood-brain barrier disorder in Alzheimer’s
indicate that these factors have precipitated the clinical disease. Acta Neuropathologica, 118: 103–113.
manifestations of AD at a relatively early stage of pathology. Beydoun, M.A., Beydoun, H.A., Gamaldo, A.A. et al.
However, other vascular risk factors such as atrial fibrilla- (2014). Epidemiologic studies of modifiable fac-
tion, hypertension and angina have been found to be associ- tors associated with cognition and dementia:
ated with faster dementia progression (Mielke et al., 2007) Systematic review and meta-analysis. BMC Public
and the picture remains unclear and under-­investigated. Health, 14: 643.
Furthermore, a recent review concluded that randomized Blondell, S.J., Hammersley-Mather, R. and Veerman, J.L.
controlled trial evidence is currently insufficient, principally (2014). Does physical activity prevent cognitive decline
on grounds of methodology, to support treatment of vascular and dementia? A systematic review and meta-analysis
risk factors as an intervention for slowing dementia progres- of longitudinal studies. BMC Public Health, 14: 510.
sion (Valenti et al., 2014). However, a commonly neglected Bots, M.L., van Swieten, J.C., Breteler, M.M.B. et al. (1993).
implication concerns the care of people with dementia, who Cerebral white matter lesions and atherosclerosis in
will commonly have disorders such as hypertension or dia- the Rotterdam Study. Lancet, 341: 1232–1237.
betes that require pharmacotherapy and medical review. Breteler, M.M.B., van Amerongen, N.M., van Swieten,
Neurodegenerative processes themselves may have effects on J. C. et al. (1994a). Cognitive correlates of ventricular
factors such as blood pressure and body composition, alter- enlargement and cerebral white matter lesions on
ing susceptibility to drug effects and side effects, and require magnetic resonance imaging. Stroke, 25: 1109–1115.
Vascular factors and Alzheimer’s disease 625

Breteler, M.M.B., van Swieten, J.C., Bots, M.L. et al. Fein, G., Van Dyke, C., Davenport, L. et al. (1990).
(1994b). Cerebral white matter lesions, vascular Preservation of normal cognitive functioning in elderly
risk factors, and cognitive function in a population- subjects with extensive white-matter lesions of long
based study: The Rotterdam Study. Neurology, 44: duration. Archives of General Psychiatry, 47: 220–223.
1246–1252. Fernando, M.S., Ince, P.G. and MRC Cognitive Function
Bruandet, A., Richard, F., Bombois, S. et al. (2009). and Ageing Neuropathology Study Group (2004).
Alzheimer disease with cerebrovascular disease and Vascular pathologies and cognition in a population-
vascular dementia: Clinical features and course com- based cohort of elderly people. Journal of the
pared with Alzheimer disease. Journal of Neurology, Neurological Sciences, 226: 13–17.
Neurosurgery and Psychiatry, 80: 133–139. Ferucci, L., Guralnik, J.M., Salive, M.E. et al. (1996).
Burgmans, S., van de Haar, H.J., Verhey, F.R. and Backes, Cognitive impairment and risk of stroke in the older
W.H. (2013). Amyloid-β interacts with blood-brain bar- population. Journal of the American Geriatrics Society,
rier function in dementia: A systematic review. Journal 44: 237–241.
of Alzheimer’s Disease, 35: 859–873. Firbank, M.J., Allan, L.M., Burton, E.J. et al. (2012).
Burke, W.J., Coronado, P.G., Schmitt, C.A. et al. (1994). Neuroimaging predictors of death and dementia in a
Blood pressure regulation in Alzheimer’s disease. cohort of older stroke survivors. Journal of Neurology,
Journal of the Autonomic Nervous System, 48: 65–71. Neurosurgery and Psychiatry, 83: 263–267.
Clarke, R., Smith, D., Jobst, K. et al. (1998). Folate, vitamin Gray, S.M., Meijer, R.I. and Barrett, E.J. (2014). Insulin
B12, and serum total homocysteine levels in con- regulates brain function, but how does it get there?
firmed Alzheimer disease. Archives of Neurology, 55: Diabetes, 63: 3992–3997.
1449–1455. Grinberg, L.T. and Thal, D.R. (2010). Vascular pathology in
Craft, S. and Watson, G.S. (2004). Insulin and neurode- the aged human brain. Acta Neuropathologica, 119:
generative disease: Shared and specific mechanisms. 277–290.
Lancet Neurology, 3: 169–178. Guo, Z., Viitanen, M., Fratiglioni, L. and Winblad, B. (1996).
Cukierman-Yaffe, T., Gerstein, H.C., Williamson, J.D. et al. Low blood pressure and dementia in elderly people:
(2009). Relationship between baseline glycemic control The Kungsholmen project. British Medical Journal,
and cognitive function in individuals with type 2 dia- 312: 805–808.
betes and other cardiovascular risk factors: The action Gustafson, D., Rothenberg, E., Blennow, K. et al. (2003).
to control cardiovascular risk in diabetes-memory in An 18-year follow-up of overweight and risk of
diabetes (ACCORD-MIND) trial. Diabetes Care, 32: Alzheimer disease. Archives of Internal Medicine, 163:
221–226. 1524–1528.
de la Torre, J.C. and Mussivand, T. (1993). Can disturbed Halliday, G., Robinson, S.R., Shepherd, C. and Kril, J.
brain microcirculation cause Alzheimer’s disease? (2000). Alzheimer’s disease and inflammation: A review
Neurological Research, 15: 146–153. of cellular and therapeutic mechanisms. Clinical and
Dominquez, L.J., Paolisso, G. and Barbaquallo, M. (2010). Experimental Pharmacology and Physiology, 27: 1–8.
Glucose control in the older patient: From intensive, Hassing, L.B., Dahl, A.K., Thorvaldsson, V. et al. (2009).
to effective and safe. Aging Clinical and Experimental Overweight in midlife and risk of dementia: A 40-year
Research, 22: 274–280. follow-up study. International Journal of Obesity, 33:
Dufouil, C., de Kersaint-Gilly, A., Besançon, V. et al. (2001). 893–898.
Longitudinal study of blood pressure and white matter Hénon, H., Durieu, I., Guerouaou, D. et al. (2001).
hyperintensities. Neurology, 56: 921–926. Poststroke dementia: Incidence and relationship to
Elias, M.F., Wolf, P.A., D’Agostino, R.B. et al. (1993). prestroke cognitive decline. Neurology, 57: 1216–1222.
Untreated blood pressure level is inversely related Hofman, A., Ott, A., Breteler, M.M.B. et al. (1997).
to cognitive functioning: The Framingham study. Atherosclerosis, apolipoprotein E, and prevalence of
American Journal of Epidemiology, 138: 353–364. dementia and Alzheimer’s disease in the Rotterdam
Emsley, H.C.A. and Tyrrell, P.J. (2002). Inflammation and Study. Lancet, 349: 151–154.
infection in clinical stroke. Journal of Cerebral Blood Hommet, C., Mondon, K., Constans, T. et al. (2011).
Flow and Metabolism, 22: 1399–1419. Review of cerebral microangiopathy and Alzheimer’s
Erus, G., Battapady, H., Zhang, T. et al. (2015). Spatial disease: Relation between white matter hyperin-
patterns of structural brain changes in type 2 dia- tensities and microbleeds. Dementia and Geriatric
betic patients and their longitudinal progression with Cognitive Disorders, 32: 367–378.
intensive control of blood glucose. Diabetes Care, 38: Hugenschmidt, C.E., Lovato, J.F., Ambrosius, W.T. et al.
97–104. (2014). The cross-sectional and longitudinal associa-
Farkas, E., De Jong, G.I., De Vos, R.A. et al. (2000). tions of diabetic retinopathy with cognitive func-
Pathological features of cerebral cortical capillaries tion and brain MRI findings: The Action to Control
are doubled in Alzheimer’s disease and Parkinson’s Cardiovascular Risk in Diabetes (ACCORD) trial.
disease. Acta Neuropathologica, 100: 395–402. Diabetes Care, 37: 3244–3252.
626 Dementia

Jack, C.R.J., Wiste, H.J., Weigand, S.D. et al. (2014). Age- Launer, L.J., Miller, M.E., Williamson, J.D. et al. (2011).
specific population frequencies of cerebral β-amyloidosis Effects of intensive glucose lowering on brain struc-
and neurodegeneration among people with normal ture and function in people with type 2 diabetes
cognitive function aged 50–89 years: A cross-sectional (ACCORD MIND): A randomised open-label substudy.
study. Lancet Neurology, 13: 997–1005. Lancet Neurology, 10: 969–977.
Johnston, J.M., Nazar-Stewart, V., Kelsey, S.F. et al. (2000). Laurin, D., Verreault, R., Lindsay, J. et al. (2001). Physical
Relationships between cerebrovascular events, APOE activity and risk of cognitive impairment and dementia
polymorphism and Alzheimer’s disease in a community in elderly persons. Archives of Neurology, 58: 498–504.
sample. Neuroepidemiology, 19: 320–326. Lautenschlager, N.T., Cox, K.L., Flicker, L. et al.
Kalaria, R.N. and Skoog, I. (2002). Pathophysiology: (2008). Effect of physical activity on cognitive func-
Overlap with Alzheimer’s disease. In Erkinjuntti, T. tion in older adults at risk for Alzheimer disease.
and Gauthier, S. (eds.), Vascular Cognitive Impairment. Journal of the American Medical Association, 300:
London, United Kingdom: Martin Dunitz. 1027–1037.
Keage, H.A.D., Carare, R.O., Friedland, R.P. et al. (2009). Liao, D., Cooper, L., Cai, J. et al. (1997). The prevalence
Population studies of sporadic cerebral amyloid and severity of white matter lesions, their relationship
angiopathy and dementia: A systematic review. BMC with age, ethnicity, gender, and cardiovascular disease
Neurology, 9: 3. risk factors: The ARIC study. Neuroepidemiology, 16:
Kim, J.-M., Stewart, R., Kim, S.-W. et al. (2008). Changes 149–162.
in folate, vitamin B12, and homocysteine associ- Luchsinger, J.A., Tang, M.-X., Shea, S. and Mayeux, R.
ated with incident dementia. Journal of Neurology, (2004). Hyperinsulinemia and risk of Alzheimer disease.
Neurosurgery and Psychiatry, 79: 1079–1081. Neurology, 63: 1187–1192.
Kivipelto, M., Helkala, E.-L., Laakso, M. P. et al. (2001). Margaglione, M., Seripa, D., Gravina, C. et al. (1998).
Midlife vascular risk factors and Alzheimer’s disease in Prevalence of apolipoprotein E alleles in healthy
later life: Longitudinal, population based study. British subjects and survivors of ischemic stroke. Stroke, 29:
Medical Journal, 322: 1447–1451. 399–403.
Kogure, K. and Kato, H. (1993). Altered gene expression in Marks, J.L., King, M.G. and Baskin, D.G. (1991).
cerebral ischemia. Stroke, 24: 2121–2127. Localisation of insulin and type 1 IGF receptors in rat
Kokmen, E., Whistman, J.P., O’Fallon, W.M. et al. (1996). brain by in vitro autoradiography and in situ hybridisa-
Dementia after ischemic stroke: A population-based tion. Advances in Experimental Medical Biology, 293:
study in Rochester, Minnesota (1960–1984). Neurology, 459–470.
19: 154–159. Matthews, F.E., Arthur, A., Barnes, L.E. et al. (2013). A two-
Kramer, A.F., Hahn, S., Cohen, N.J. et al. (1999). Ageing, fit- decade comparison of prevalence of dementia in indi-
ness and neurocognitive function. Nature, 400: 418–419. viduals aged 65 years and older from three geographical
Kuller, L.H., Shemanski, L., Manolio, T. et al. (1998). areas of England: Results of the Cognitive Function and
Relationship between ApoE, MRI findings, and cogni- Ageing Study I and II. Lancet, 382: 1405–1412.
tive function in the Cardiovascular Health Study. McCarron, M.O., Delong, D. and Alberts, M.J. (1999).
Stroke, 29: 388–398. APOE genotype as a risk factor for ischemic cerebro-
Kuusisto, J., Koivisto, K., Mykkänen, L. et al. (1997). vascular disease. Neurology, 53: 1308–1311.
Association between features of the insulin resistance McGuinness, B., Craig, D., Bullock, R. and Passmore,
syndrome and Alzheimer’s disease independently P. (2009a). Statins for the prevention of demen-
of apolipoprotein E4 phenotype: Cross sectional tia. Cochrane Database of Systematic Reviews, (2):
population based study. British Medical Journal, 315: CD003160.
1045–1049. McGuinness, B., Todd, S., Passmore, P. and Bullock, R.
Kwok, C.S., Loke, Y.K., Hale, R. et al. (2011). Atrial fibrilla- (2009b). Blood pressure lowering in patients without
tion and incidence of dementia: A systematic review prior cerebrovascular disease for prevention of cogni-
and meta-analysis. Neurology, 76: 914–922. tive impairment and dementia. Cochrane Database of
Laitinen, M.H., Ngandu, T., Rovio, S. et al. (2006). Fat Systematic Reviews, (4): CD004034.
intake at midlife and risk of dementia and Alzheimer’s Mielke, M.M., Rosenberg, P.B., Tschanz, J. et al. (2007).
disease: A population-based study. Dementia and Vascular factors predict rate of progression in
Geriatric Cognitive Disorders, 22: 99–107. Alzheimer disease. Neurology, 69: 1850–1858.
Launer, L.J. (2009). Diabetes: Vascular or neurodegen- Moroney, J.T., Bagiella, E., Tatemichi, T.K. et al. (1997).
erative: An epidemiologic perspective. Stroke, 40: Dementia after stroke increases the risk of long-term
S53–S55. stroke recurrence. Neurology, 48: 1317–1325.
Launer, L.J., Masaki, K., Petrovitch, H. et al. (1995). The Muller, M., Tang, M.X., Schupf, N. et al. (2007). Metabolic
association between midlife blood pressure levels and syndrome and dementia risk in a multiethnic elderly
late-life cognitive function. Journal of the American cohort. Dementia and Geriatric Cognitive Disorders,
Medical Association, 274: 1846–1851. 24: 185–192.
Vascular factors and Alzheimer’s disease 627

Neuropathology Group of the Medical Research Qiu, C., Winblad, B. and Fratiglioni, L. (2005). The age-
Council Cognitive Function and Ageing Study (2001). dependent relation of blood pressure to cognitive func-
Pathological correlates of late-onset dementia in a tion and dementia. Lancet Neurology, 4: 487–499.
multicentre, community-based population in England Qizilbash, N., Gregson, J., Johnson, M.E. et al. (2015).
and Wales. Lancet, 357: 169–175. BMI and risk of dementia in two million people over
Notkola, I.-L., Sulkava, R., Pekkanen, J. et al. (1998). two decades: A retrospective cohort study. Lancet
Serum total cholesterol, apolipoprotein E 4 allele, and Diabetes and Endocrinology, 3 (6): 431–436.
Alzheimer’s disease. Neuroepidemiology, 17: 14–20. Razay, G., Vreugdenhill, A. and Wilcock, G. (2007). The
Ott, A., Breteler, M.M.B., de Bruyne, M.C. et al. (1997). metabolic syndrome and Alzheimer disease. Archives
Atrial fibrillation and dementia in a population-based of Neurology, 64: 93–96.
study. Stroke, 28: 316–321. Reaven, G.M. (1988). Role of insulin resistance in human
Ott, A., Slooter, A.J.C., Hofman, A. et al. (1998). Smoking disease. Diabetes, 37: 1595–1607.
and the risk of dementia and Alzheimer’s disease in a Roberts, R.O., Geda, Y.E., Knopman, D.S. et al. (2008).
population-based cohort study: The Rotterdam Study. Association of duration and severity of diabetes
Lancet, 351: 1840–1843. mellitus with mild cognitive impairment. Archives of
Pendlebury, S.T. and Rothwell, P.M. (2009). Prevalence, Neurology, 65: 1066–1073.
incidence, and factors associated with pre-stroke and Rönnemaa, E., Zethelius, B., Sundelöf, J. et al. (2008).
post-stroke dementia: A systematic review and meta- Impaired insulin secretion increases the risk of
analysis. Lancet Neurology, 8: 1006–1018. Alzheimer disease. Neurology, 71: 1065–1071.
Pfeiffer, L.A., White, L.R., Ross, G.W. et al. (2002). Ruitenberg, A., van Swieten, J.C., Witteman, J.C.M. et al.
Cerebral amyloid angiopathy and cognitive function: (2002). Alcohol consumption and risk of dementia: The
The HAAS autopsy study. Neurology, 58: 1587–1588. Rotterdam Study. Lancet, 359: 281–286.
Pluta, R., Jablonski, M., Ulamek-Koziol, M. et al. (2013). Sanz, C., Andrieu, S., Sinclair, A. et al. and REAL.FR Study
Sporadic Alzheimer’s disease begins as episodes Group (2009). Diabetes is associated with a slower rate
of brain ischemia and ischemically dysregulated of cognitive decline in Alzheimer’s disease. Neurology,
Alzheimer’s disease genes. Molecular Neurobiology, 73: 1359–1366.
48: 500–515. Savva, G.M., Wharton, S.B., Ince, P.G. et al. (2009). Age,
Posner, H.B., Tang, M.-X., Luchsinger, J. et al. (2002). neuropathology, and dementia. New England Journal
The relationship of hypertension in the elderly to AD, of Medicine, 360: 2302–2309.
vascular dementia, and cognitive function. Neurology, Scarmeas, N., Luchsinger, J.A., Schupf, N. et al. (2009).
58: 1175–1181. Physical activity, diet, and risk of Alzheimer disease.
Prasad, S., Sajja, R.K., Naik, P. and Cucullo, L. (2014). Journal of the American Medical Association, 302:
Diabetes mellitus and blood-brain barrier dysfunction: 627–637.
An overview. Journal of Pharmacovigilance, 2: 125. Seshadri, S., Beiser, A., Selhub, J. et al. (2002). Plasma
Preston, S.D., Steart, P.V., Wilkinson, A. et al. (2003). homocysteine as a risk factor for dementia and
Capillary and arterial cerebral amyloid angiopathy in Alzheimer’s disease. New England Journal of
Alzheimer’s disease: Defining the perivascular route Medicine, 346: 476–483.
for the elimination of amyloid beta from the human Skoog, I., Berg, S., Johansson, B. et al. (1996a). The influ-
brain. Neuropathology and Applied Neurobiology, 29: ence of white matter lesions on neuropsychological
106–117. functioning in demented and non-demented 85-year-
Prince, M., Lovestone, S., Cervilla, J. et al. (2000). The olds. Acta Neurologica Scandanavica, 93: 142–148.
association between APOE and dementia is medi- Skoog, I., Lernfelt, B., Landahl, S. et al. (1996b). 15-Year
ated neither by vascular disease nor its risk factors longitudinal study of blood pressure and dementia.
in an aged cohort of survivors with hypertension. Lancet, 347: 1141–1145.
Neurology, 54: 397–402. Slooter, A.J.C., Cruts, M., Ott, A. et al. (1999). The
Punthakee, Z., Miller, M.E., Launer, L.J. et al. (2012). Poor effect of APOE on dementia is not through atheroscle-
cognitive function and risk of severe hypoglycemia in rosis: The Rotterdam Study. Neurology, 53: 1593–1595.
type 2 diabetes: Post hoc epidemiological analysis of Smith, E.E. and Greenberg, S.M. (2009). β-Amyloid, blood
the ACCORD trial. Diabetes Care, 35: 787–793. vessels, and brain function. Stroke, 40: 2601–2606.
Qiu, W.Q. and Folstein, M.F. (2006). Insulin, insulin- Solomon, A., Kareholt, I., Ngandu, T. et al. (2007). Serum
degrading enzyme and amyloid-beta peptide cholesterol changes after midlife and late-life cogni-
in Alzheimer’s disease: Review and hypothesis. tion: Twenty-one-year follow-up study. Neurology, 68:
Neurobiology of Aging, 27: 190–198. 751–756.
Qiu, C., von Strauss, E., Winblad, B. and Fratiglioni, L. Solomon, A., Kivipelto, M., Wolozin, B. et al. (2009). Midlife
(2004). Decline in blood pressure over time and risk of serum cholesterol and increased risk of Alzheimer’s and
dementia: A longitudinal study from the Kungsholmen vascular dementia three decades later. Dementia and
Project. Stroke, 35: 1810–1815. Geriatric Cognitive Disorders, 28: 75–80.
628 Dementia

Srikanth, V.K., Anderson, J.F.I., Donnan, G.A. et al. Vlassara, H., Fuh, H., Makita, Z. et al. (1992).
(2004). Progressive dementia after first-ever stroke: Exogenous advanced glycosylation end products
A ­community-based follow-up study. Neurology, 63: induce complex vascular dysfunction in normal ani-
785–792. mals: A model for diabetic and aging complications.
Stewart, R. and Liolitsa, D. (1999). Type 2 diabetes mel- Proceedings of the National Academy of Science, 89:
litus, cognitive impairment and dementia. Diabetic 12043–12047.
Medicine, 16: 93–112. Whitmer, R.A., Karter, A.J., Yaffe, K. et al. (2009).
Stewart, R., Masaki, K., Xue, Q.-L. et al. (2005). A 32-year Hypoglycemic episodes and risk of dementia
prospective study of change in body weight and in older patients with type 2 diabetes mellitus.
incident dementia: The Honolulu-Asia Aging Study. Journal of the American Medical Association,
Archives of Neurology, 62: 55–60. 301: 1565–1572.
Stewart, R., Xue, Q.-L., Masaki, K. et al. (2009). Change Yaffe, K., Falvey, C., Hamilton, N. et al. (2012). Diabetes,
in blood pressure and incident dementia: A 32-year glucose control, and 9-year cognitive decline among
prospective study. Hypertension, 54: 233–240. older adults without dementia. Archives of Neurology,
Stewart, R., Xue, Q.-L., White, L.R. and Launer, L.J. (2007). 69 (9): 1170–1175.
26-Year change in total cholesterol levels and incident Yaffe, K., Falvey, C.M., Hamilton, N. et al. (2013).
dementia. The Honolulu-Asia Aging Study. Archives of Association between hypoglycemia and dementia in a
Neurology, 62: 55–60. biracial cohort of older adults with diabetes mellitus.
Sullivan, M.D., Katon, W.J., Lovato, L.C. et al. (2013). JAMA Internal Medicine, 173: 1300–1306.
Association of depression with accelerated cognitive Yaffe, K., Lindquist, K., Schwartz, A.V. et al. (2011).
decline among patients with type 2 diabetes in the Advanced glycation end product level, diabetes,
ACCORD-MIND trial. JAMA Psychiatry, 70: 1041–1047. and accelerated cognitive aging. Neurology, 77:
Tran, D., Baxter, J., Hamman, R.F. and Grigsby, J. (2014). 1351–1356.
Impairment of executive cognitive control in type 2 Zhong, G., Wang, Y., Zhang, Y. et al. (2015). Smoking is
diabetes, and its effects on health-related behavior associated with an increased risk of dementia: A meta-
and use of health services. Journal of Behavioural analysis of prospective cohort studies with investi-
Medicine, 37: 414–422. gation of potential effect modifiers. PLoS ONE, 10:
Valenti, R., Pantoni, L. and Markus, H.S. (2014). Treatment e0118333.
of vascular risk factors in patients with a diagnosis
of Alzheimer’s disease: A systematic review. BMC
Medicine, 12: 160.
5
Part    

Cerebrovascular Disease and Cognitive


Impairment

58 What is vascular cognitive impairment? 630


Timo Erkinjuntti, Leonardo Pantoni and Susanna Melkas
59 The neuropathology of vascular dementia 643
Raj N. Kalaria
60 Therapeutic strategies for vascular cognitive disorders 660
Gustavo C. Román
58
What is vascular cognitive impairment?

TIMO ERKINJUNTTI, LEONARDO PANTONI AND SUSANNA MELKAS

the diagnosis of dementia in general. The criteria included


58.1 VASCULAR BURDEN OF THE early and prominent memory loss, progressive cognitive
BRAIN – BACKGROUND impairment, evidence of irreversibility and presence of
cognitive impairment sufficient to affect normal activities
As early as 1896, ‘arteriosclerotic dementia’ (referring to vas- of daily living (ADLs) (McKhann et al., 1984; Erkinjuntti
cular dementia [VaD]) was separated from ‘senile dementia’ et al., 1997). The characteristic episodic memory impair-
(referring to Alzheimer’s disease [AD]). Alois Alzheimer ment apparent in AD is attributed to atrophy of the medial
together with Otto Binswanger recognized the hetero- temporal lobe. In contrast, CVD lesions do not have the
geneity of VaD by describing four clinical–­ pathological same regional predilection. The emphasis of the current
­subtypes of VaD (Roman, 2001), as well as vascular lesions Alzheimer type dementia criteria limited to the episodic
in AD. Nevertheless, until the 1970s cerebral atherosclero- memory impairment underestimates the vascular burden
sis causing chronic decrease of blood supply to the brain on cognition. The conventional dementia syndrome con-
was thought to be the commonest cause of dementia, and cept recognizes the vascular burden of the brain too late,
AD was regarded as a rare cause affecting only younger when often opportunities to prevent and treat are lost.
patients. Tomlinson et al. (1970) reinvented AD as the most Accordingly, it was suggested that the ‘Alzheimerized’
frequent cause of dementia. Hachinski et al. (1974) used the dementia concept should be abandoned in the setting of
term multi-infarct dementia (MID) to describe the mecha- CVD, and indeed this was one of the motives behind the
nism by which they considered VaD was produced. As the development of the broader category of vascular cogni-
pendulum swung in the direction of AD, vascular forms of tive impairment (VCI) (Hachinski, 1990; Bowler and
dementia became relegated to a position of relative obscu- Hachinski, 1995; Pasquier and Leys, 1997; O’Brien et al.,
rity (Brust, 1988). 2003; Roman et al., 2004; Moorhouse and Rockwood,
During the 1980s and the early 1990s almost all cere- 2008), sometimes also referred as vascular cognitive disor-
brovascular disease (CVD) injury leading to dementia was der (Roman et al., 2004).
ascribed to large cortical and subcortical infarcts (so-called
MID) (Erkinjuntti and Hachinski, 1993). The concept of
VaD was introduced to refine further the description of 58.2 THE MODERN CONCEPT OF VCI
dementias caused by infarcts of varying sizes, including the
smaller lacunar and microinfarcts, as well as white matter Now VaD has come full circle with the resurgence of inter-
lesions (WMLs) (Roman et al., 1993). VaD appropriately est in the whole spectrum of vascular causes of cognitive
defined a group of heterogeneous dementia syndromes of impairment and dementia (Hachinski, 1990; O’Brien et al.,
CVD origin of which the cortical and subcortical vascular 2003). The new term VCI recognizes the broad spectrum of
disease were considered as important subtypes (Erkinjuntti cognitive and behavioural changes associated with vascu-
and Hachinski, 1993; Roman et al., 1993, 2002). Although lar pathology (Bowler and Hachinski 1995; O’Brien, et al.,
this was an important step forward, it was not adequate 2003; Moorhouse and Rockwood 2008).
to fully describe the vascular causes of early cognitive Evidence of complex interactions between vascular aetiolo-
impairments. gies, changes in the brain, host factors and cognition prompted
The recognition of AD as the commonest cause of formulation of the new concepts (Chui, 1989; Tatemichi,
dementia led to the development of operational criteria for 1990; Erkinjuntti and Hachinski, 1993; Desmond, 1996;

630
What is vascular cognitive impairment? 631

Pasquier and Leys, 1997), as well as underlying the heteroge- (vMCI) in a similar way to that of amnestic mild cognitive
neity of VaDs and recognition of the small vessel ischaemic impairment (aMCI) for AD (Petersen et al., 2002). Early
subcortical vascular disease and dementia (SIVD) as a con- clinical stages of VCI starting from pre-dementia may be
cept applicable in clinical studies (Roman, 1985; Erkinjuntti, recognized only in a part of patients with VaD, typically in
1987; Erkinjuntti et al., 2000; Erkinjuntti and Pantoni, 2000; those with small vessel disease dementia. In other patients,
Roman et al., 2002). The facts that vascular factors relate to a single step from the brain-at-risk stage to dementia may
and coexist with AD (Kalaria and Ballard, 1999; Skoog et al., occur (Pantoni et al., 2009) (Figure 58.1).
1999; Kivipelto et al., 2006) were important too. VCI refers to all aetiologies of CVD including vascular
Accordingly, the modern concept of VCI aims to cover the risk factors that can result in brain damage leading to cog-
spectrum of cognitive impairments, as well as the spectrum nitive impairment. VCI may include cases with cognitive
of the aetiologies of vascular burden of the brain related to impairment related to hypertension, diabetes or athero-
cognitive impairment (O’Brien et al., 2003; Bowler, 2007; sclerosis, transient ischaemic attacks, cortico-subcortical
Hachinski, 2008; Moorhouse and Rockwood, 2008). The infarcts, silent infarcts, strategic infarcts, small vessel dis-
modern concept of VCI relates to important shifts in think- ease with WMLs and lacunae, as well as AD pathology with
ing (Hachinski, 2008): coexisting CVD. VCI can also encompass those patients
who survive intracerebral and other intracranial haemor-
1. Shift from arbitrary thresholds, such as the AD rhages but are left with residual cognitive impairment.
­dementia threshold, to a continuum of cognitive
impairment
2. Shift from the late stages to the early stages of disease 58.3 VCI PATHOPHYSIOLOGY
3. Shift from the effects to the causes
The pathophysiology incorporates interactions between vas-
The evolution of the new VCI concept is similar to the cular aetiologies (CVDs and vascular risk factors), changes
more recent evolution in degenerative causes of memory in the brain (infarcts, WMLs, atrophy, AD pathology) and
diseases, i.e. the new research criteria for the amnestic pro- host factors (age, education) (Chui, 1989; Tatemichi, 1990;
drome of AD (Dubois et al., 2007). Erkinjuntti and Hachinski, 1993; Desmond, 1996; Pasquier
The cognitive syndrome of VCI encompasses all levels and Leys, 1997; O’Brien et al., 2003) (Figure 58.2). Aetiologies
of cognitive decline, from the earliest deficits to a severe of VCI include both CVD and risk factors (Table 58.1).
and broad dementia-like cognitive syndrome (Bowler et al.,
1999; O’Brien et al., 2003). VCI cases that do not meet the 58.3.1 CEREBROVASCULAR DISEASES
criteria for dementia can also be labelled as VCI with no
dementia or vascular cognitive impairment no demen- CVDs include small vessel disease, large vessel disease and
tia (CIND) (Rockwood et al., 2000). These patients have strategic infarcts that may be either cortical or subcortical.
also been labelled as vascular mild cognitive impairment CVDs also include haemorrhage, vascular malformations,

Post-stroke dementia Small vessel dementia

VaD

VCI - no dementia

Perisymptomatic
stage

Brain at risk
(vascular risk factors)

Figure 58.1 The pyramid of vascular cognitive impairment (VCI). (From Pantoni et al., Cerebrovascular Disease, 27, 191–
196, 2009.)
632 Dementia

Brain AD Host factors


pathology e.g. age
education
genetics
Vasular
aetiologies Infarcts, Cognitive
Cerebrovascular white impairment
disorders matter
Vascular risk factors lesions Dementia

Brain
atrophy

Figure 58.2 Interactions between vascular causes, brain changes and host factors in vascular cognitive impairment.

Table 58.1 Aetiologies of vascular dementia conditions due to hypoperfusion and hereditary condi-
tions (e.g. cerebral autosomal dominant arteriopathy with
CVDs
subcortical strokes and leukoencephalopathy [CADASIL])
Large vessel disease
(Erkinjuntti, 2007; Pantoni et al., 2009).
  Artery-to-artery embolism
   Occlusion of an extra- or intracranial artery
Cardiac embolic events 58.3.2 RISK FACTORS
Small vessel disease
Risk factors associated with VCI include risks for stroke
  Lacunar infarcts
and ischaemic WMLs (Basile et al., 2006). Clinically
  Ischaemic WMLs
symptomatic infarcts, clinically silent infarcts and
Haemodynamic mechanisms WMLs relate to higher dementia risk (Verdelho et al.,
Specific arteriopathies 2010). Similarly to AD, the risks for VCI may be consid-
Haemorrhages ered under demographic, vascular, genetic and ischaemic
  Intracerebral haemorrhage lesions related ­variables (Skoog, 1994; Gorelick, 1997;
   Subarachnoidal haemorrhage Skoog, 1998; Kivipelto et al., 2006). Hypoxic ischaemic
   Chronic subdural haemorrhage events giving rise to global cerebrovascular insufficiency
  Microbleeds are also important risk factors for incident dementia
Haematological factors in patients with stroke (Sulkava and Erkinjuntti, 1987;
Venous diseases Moroney et al., 1996, 1997a). Furthermore, the tradi-
Hereditary entities tional vascular risk factors and stroke are also indepen-
Risk factors dent factors for the clinical presentation of MCI and AD
Vascular (Snowdon et al., 1997; Skoog et al., 1999). The important
  Arterial hypertension independent midlife risk factors of clinical AD include
  Atrial fibrillation arterial hypertension, high cholesterol, diabetes, obesity
  Myocardial infarction and reduced physical activity among others (Skoog et al.,
   Coronary heart disease 1999; Kivipelto et al., 2006).
  Diabetes
  Generalized atherosclerosis 58.3.3 BRAIN CHANGES
  Lipid abnormalities
  Smoking Brain changes related to VCI include both ischaemic and
Demographic factors non-ischaemic factors (Tatemichi, 1990; Chui et al., 1992;
  High age Brun, 1994; Erkinjuntti et al., 1999b) (Table 58.2). Static
  Low education ischaemic lesions include arterial territorial infarct, distal
Genetic factors field (watershed) infarct, lacunar infarct, ischaemic WMLs
  Family history and incomplete ischaemic injury. Incomplete ischaemic
   Specific genetic factors injury includes focal gliosis and incomplete white mat-
Stroke-related factors ter infarctions in areas of selective vulnerability (Englund
   Type of CVD et al., 1988; Pantoni and Garcia, 1997). Functional isch-
   Site and size of infarcts aemic changes include both focal (around the ischaemic
Abbreviations: CVD, cerebrovascular diseases; WMLs, white matter
lesion) and remote (disconnection, diaschisis) effects (Shim
lesions. et al., 2006). Ischaemia relates also to atrophy without AD
What is vascular cognitive impairment? 633

Table 58.2 Brain changes in vascular dementia ●● It is doubtful that only a single small lesion could sup-
Static lesions port a diagnosis of VaD. However, rare cases with pure
strategic infarcts may be exceptions.
Arterial territorial infarct
●● Absence of CVD lesions on CT or MRI is against a
Distal field (watershed) infarct
diagnosis of VaD.
Lacunar infarct
Ischaemic WMLs Examples of MRI findings in the main subtypes of VADs
Incomplete ischaemic injury are shown in Figure 58.3.
Microbleeds With newer MRI techniques, such as diffusion tension
Atrophy imaging (DTI), susceptibility weighted imaging (SWI) and
Functional ischaemic changes functional MRI, microangiopathic markers of SVD and
Focal (around the ischaemic lesion) white matter tract integrity can be detected and followed
Remote (disconnection, diaschisis) more accurately than before. Diffusion changes in brain
tissue that appears normal in conventional MRI associate
with cognitive decline and gradual loss of independence
pathology (Vinters et al., 2000), and brain atrophy accel- (Jokinen et al., 2013).
erates cognitive decline in cerebral small vessel disease
(Jokinen et al., 2012).
58.4 VCI – THE SIZE OF THE PROBLEM
58.3.4 BRAIN MECHANISMS
Brain mechanisms proposed to be related to VCI include The size of the VCI problem can be studied by reviewing
volume of brain infarcts, number of infarcts (additive, data on (1) VCI and CIND, (2) post-stroke dementia (PSD),
synergistic), site of infarcts, WMLs, other ischaemic fac- (3) post-stroke cognitive impairment (PSCI), (4) VaD and
tors (incomplete ischaemic injury, delayed neuronal death, (5) AD with CVD.
functional changes), atrophy and additive effects of other
pathologies (e.g. AD) (Erkinjuntti et al., 1988; Chui, 1989; 58.4.1 VCI
Tatemichi, 1990; Desmond, 1996; Erkinjuntti, 1999; O’Brien
et al., 2003; Kalaria et al., 2004). The relationship between Estimates of the population distribution of VCI and its
vascular factors and cognition is challenging: do the identi- outcomes are influenced by the variety of definitions used
fied vascular factors cause, compound or only coexist with (Erkinjuntti et al., 1997; Pohjasvaara et al., 1997; Lobo et al.,
the VCI syndrome? Small vessel disease with subsequent 2000; Pohjasvaara et al., 2000b). For example, if AD with
brain atrophy may well be the cardinal origin of the main CVD or the previously defined VaD with AD pathology is
subtypes of VCI (Erkinjuntti, 1987; Roman, 1987; Pantoni included, then VCI would most certainly be the most com-
and Garcia, 1995; Roman et al., 2002) (see Chapters 59 and mon cause of chronic progressive cognitive impairment in
60). Another important question is whether they contribute elderly people (Rockwood et al., 2000). In a Canadian study,
to the risk and clinical picture of AD (Snowdon et al., 1997). the prevalence of VCI (including CIND) was estimated at 5%
in people over age 65 years (90). The prevalence of vascular
58.3.5 ANATOMICAL BRAIN IMAGING CIND was 2.4%, that of AD with CVD was 0.9% and of VaD
alone was 1.5%. By comparison, the prevalence of AD with-
Computed tomography (CT) and magnetic resonance imag- out a vascular component, at all ages up to age 85 years, was
ing (MRI) studies on VaD indicate that bilateral ischaemic 5.1% and was less common than VCI (Rockwood et al., 2000).
lesions are of importance (Erkinjuntti and Hachinski, 1993;
DeCarli and Scheltens 2002). A summary of the interaction 58.4.2 POST-STROKE DEMENTIA (PSD)
between lesion and cognition in VaD includes (Erkinjuntti,
1999; Erkinjuntti et al., 1999; DeCarli and Scheltens, 2002; The frequency of PSD varies from 12% to 32% within 3
Roman et al., 2002; O’Brien et al., 2003): months to 1 year after stroke (Leys et al. 2005). In the
Helsinki study, the frequency was 25% 3 months after the
●● No single feature, but a combination of infarct features, incident stroke and the frequency increased with increasing
extent and type of WMLs, degree and site of atrophy, and age: 19% among those aged 55–64 years and 32% in those
host factors characteristics are correlates of VCI/VaD. aged 75–85 years (Pohjasvaara et al., 1997). The incidence
●● Infarct features favouring VaD include bilaterality, mul- of PSD increases with a longer follow-up time from 10% at
tiplicity (>2), location in the dominant hemisphere and 1 year to 32% after 5 years (Leys et al., 2005). A history of
location in the fronto-subcortical and limbic structures. stroke increases the risk of subsequent dementia by a factor
●● WML features favouring VaD are extensive WMLs of 5 (Linden et al., 2004; Leys et al., 2005).
(extending periventricular WMLs and confluent to Determinants of PSD include high age, low education,
extending WMLs in the deep WM). pre-stroke dependency and cognitive impairment including
634 Dementia

population and the clinical methods used (Lobo et al., 2000).


The prevalence of VaD ranges from 1.2% to 4.2% in persons
(a) (b) aged 65 years and older (Hebert and Brayne, 1995). Using
population-based identification of persons aged 65 years and
older, the European collaborative study reported that the age-
standardized prevalence of dementia was 6.4% (all causes),
4.4% for AD and 1.6% for VaD (Lobo et al., 2000). In this
study, 15.8% of all cases had VaD and 53.7% AD. However,
(c)
these studies did not estimate the size of AD with CVD
(d)
population in more detail. The incidence of VaD increases
with age, without any substantial difference between men
and women (Fratiglioni et al., 2000). The reported incidence
estimates of VaD range from 6 to 12 cases per year in 1000
persons aged 70 years and older (Hebert and Brayne, 1995).

58.4.5 AD WITH CVD


Figure 58.3 Magnetic resonance imaging scans in For too long the focus has been the two ‘pure’ ends of the
patients with (a) cortical vascular dementia, multi-infarct dementia spectrum: probable AD (McKhann et al., 1984)
dementia (bilateral infarcts), (b) subcortical ischaemic vas- and probable VaD (Roman et al., 1993). One factor has been
cular dementia (SIVD) with multiple lacunes, (c) SIVD with
the need for homogenous patient populations and high ante-­
confluent white matter lesions and (d) bilateral strategic
thalamic infarcts.
mortem diagnostic specificity for randomized clinical trials. At
the same time, combined cases, those with AD and CVD, were
neglected. Alois Alzheimer already recognized the issue of AD
pre-stroke dementia (Leys et al., 2005). Risk factors of inci- with CVD, which was reinvented by Tomlinson and colleagues
dent PSD include epileptic seizures, sepsis, cardiac arrhyth- (Tomlinson et al., 1970). In the seminal Newcastle series,
mias and congestive heart failure (Moroney et al., 1996; 8%–18% of the patients had AD with CVD (Tomlinson et al.,
Leys et al., 2005). In one large cohort study, the independent 1970). Accordingly, Roth concluded that ‘There is evidence that
clinical correlates of PSD included dysphasia, major domi- the two types of pathological change augment one another to
nant stroke syndrome, history of prior CVD and low educa- a statistically significant degree in the production of dementia’
tion (Pohjasvaara et al., 1998). Brain lesion correlates of PSD (Roth, 1971). Today, based on unselected neuropathological
include a combination of infarcts features (volume, site), series of patients, we acknowledge that AD with CVD may be
presence of WMLs (extent, location) as well as brain atro- the most frequent single cause leading to cognitive impair-
phy (Pohjasvaara et al., 2000a; Leys et al., 2005). Important ment: in the brains of cognitively impaired elderly at least 50%
critical locations include dominant hemisphere and lesions show combined AD and CVD changes (Roth, 1971; Snowdon
affecting the prefrontal–subcortical circuit. Lesions mediat- et al., 1997; Neuropathology Group, Medical Research Council
ing executive dysfunction are critical (Vataja et al., 2003). Cognitive Function and Aging Study, 2001). In addition, vas-
cular brain changes relate to a much earlier clinical presenta-
58.4.3 POST-STROKE COGNITIVE tion of the AD syndrome (Snowdon et al., 1997), and vascular
IMPAIRMENT (PSCI) risk-factors are independent factors related to clinical expres-
sion of the AD syndrome (Kivipelto et al., 2006). Importantly,
PSCI is a frequent, though neglected, consequence of stroke. AD and CVD can be seen as secret partners in cognitive and
An example of a detailed clinical study is the Helsinki Stroke brain health risks; one in every two females aged over 65 years
Ageing study (Pohjasvaara et al., 1997). Cognitive impair- will have in their coming years of life either CVD, AD or AD
ment 3 months after ischaemic stroke was present in one with CVD (Seshadri and Wolf, 2007).
domain in 62% and in two domains in 35% of patients aged
55–85 years. The cognitive domains affected included short-
term memory (31%), long-term memory (23%), constructive
and visuospatial functions (37%), executive functions (25%) 58.5 WML BURDEN
and aphasia (14%) (Pohjasvaara et al., 1997).
WMLs, frequently detected on neuroimaging, are associ-
58.4.4 VaD ated with cognitive, mood, motor and urinary disorders, all
contributing towards disability (Inzitari et al., 2007, 2009).
VaD, defined as the subset of VCI patients who fulfil tradi- In particular, confluent and extensive WMLs relate to cog-
tional AD type dementia criteria, is considered the second nitive decline, faster progression of disability and death
most common cause of dementia accounting for 10%–50% (Inzitari et al., 2009). In addition, confluent WMLs relate
cases, but this depends on the geographic location, patient to other important adverse outcomes including depression,
What is vascular cognitive impairment? 635

impaired gait and stability and urinary problems. Further, (Roman et al., 1993). In addition, research criteria for SIVD
extensive WMLs relate to the dysexecutive syndrome have also been proposed (Erkinjuntti et al., 2000).
(Jokinen et al., 2005). WMLs are seen as the surrogate of
small vessel disease and they relate to the SIVD syndrome, 58.6.1 LARGE VESSEL DISEASE
the main subtype of progressive VCI (Erkinjuntti et al., 2000;
Roman et al., 2002; Jokinen et al., 2009a). Importantly, the Large vessel disease associated VCI (cortical VaD, MID,
executive small vessel brain network is the largest human post-stroke VaD) relates predominantly to atherosclerotic
endothelial organ (Roman, 2008). disease in large vessels and cardiac embolic events. It is a
syndrome related to strokes, not a disease entity, and rarely
fulfils current criteria modelled on AD type dementia. It
is characterized by predominantly cortical and cortico-
58.6 MAIN SUBTYPES OF VCI subcortical arterial territorial and distal field (watershed)
infarcts. It is traditionally characterized by a relative abrupt
onset (days to weeks), a stepwise deterioration (some recov-
VCI encompasses many clinical features, which themselves ery after worsening) and a fluctuating course of cognitive
reflect a variety of vascular mechanisms and changes in functions (Erkinjuntti, 1987; Erkinjuntti et al., 1988, 1993;
the brain, with different causes and neurological outcomes Chui et al., 1992; Roman et al., 1993). The early cognitive syn-
(Figure 58.4). The pathophysiology is attributed to interac- drome of large vessel ­disease includes some memory impair-
tions between vascular aetiologies (CVD and vascular risk ment, which may be mild, and some heteromodal cortical
factors), changes in the brain (infarcts, WMLs, atrophy) and symptom(s) such as aphasia, apraxia, agnosia and visuospa-
host factors (age, education). tial or constructional difficulty. In addition, most patients
The main subtypes of VaD included in current classifica- have some degree of dysexecutive syndrome (Mahler and
tions are small vessel disease, large vessel disease and strate- Cummings, 1991). Due to the multiple cortico-subcortical
gic infarcts that may be either cortical or subcortical (Chui infarcts, patients with large vessel disease associated VCI
et al., 1992; Roman et al., 1993; World Health Organisation, often have additional neurological impairments (visual field
1993b; American Psychiatric Association, 1994; Erkinjuntti deficits, lower facial weakness, lateralized sensorimotor
et al., 1997; Chui et al., 2000) (Table 58.3). Hypoperfusion changes and/or gait impairment) (Erkinjuntti, 1987).
dementia resulting from global cerebrovascular insuffi-
ciency is also included. Further, subtypes include haemor- 58.6.2 SMALL VESSEL DISEASE
rhagic dementia, hereditary VaD (e.g. CADASIL) and AD
with CVD. The most widely used clinical diagnostic criteria Small vessel disease associated VCI (SIVD) incorporates
for VaD are the National Institute of Neurological Disorders two entities: ‘the lacunar state’ and ‘Binswanger’s disease’
and Stroke–Association Internationale pour la Recherche et (Erkinjuntti et al., 2000; Chui, 2001; Roman et al., 2002)
l’Enseignement en Neurosciences (NINDS-AIREN) criteria (Figure 58.5). Small vessel disease is characterized by lacunar

Vascular risk factors


Arterial hypertension, diabetes, genetics, high cholesterol, heart disease.

Damage to cerebral vasculature

Multiple distinct pathologies

Large vessel disease Small vessel disease Haemorrhage Hypoperfusion


Strategic single infarcts Multiple Lacunae Chronic SDH Global (e.g. cardiac arrest)
White matter lesions SAH Hypotension
Multiple infarcts ICH
CADASIL
Final common
pathway
Damage to critical cortical Damage/interruption of
and subcortical structures Cholinergic subcortical circuits and
transmission projections

VCI

Figure 58.4 VCI: a heterogeneous vascular disorder; SDH, subdural haemorrhage; SAH, subarachnoidal haemorrhage;
ICH, intracerebral haemorrhage; VCI, vascular cognitive impairment.
636 Dementia

Table 58.3 Vascular mechanisms, changes in the brain and clinical heterogeneity in subtypes of traditional VaD (VCI)

Cortical VaD/MID Strategic infarct VaD Subcortical ischaemic VaD/SIVD


Vascular mechanisms
Large vessel disease + + −
Cardiac embolic events + + −
Hypoperfusion + + +
Small vessel disease − + +
Changes in the brain
Arterial territorial infarct + + −
Distal field (watershed) infarct + + −
Lacunar infarct − + +
Focal, diffuse WMLs − + +
Incomplete ischaemic injury − − +
Clinical heterogeneity ++ +++ +
Abbreviations: VaD, vascular dementia; VCI, vascular cognitive impairment; MID, multi-infarct dementia; SIVD, subcortical ischaemic vascular
dementia.

Risk factors and set maintenance, as well as in abstraction (Cummings,


Small vessel change 1993, 1994; Jokinen et al., 2006, 2009a, 2009b). The memory
Critical stenosis
deficit in small vessel disease is usually milder than in AD
Occlusion Hypoperfusion and is characterized by impaired recall, relative intact rec-
Complete infarct Incomplete infarct
ognition, less severe forgetting and better benefit from cues.
Behavioural and psychological symptoms include depression,
Lacunae White matter lesions
personality change, emotional lability and incontinence, as
‘Lacunar state’ ‘Binswanger syndrome’ well as inertia, emotional bluntness and psychomotor retar-
dation. Neurological symptoms and signs in the early phases
Subcortical VCI / SIVD of small vessel disease may include subtle episodes of mild
upper motor neurone signs (drift, reflex asymmetry, incoor-
Figure 58.5 Two main pathways associated with SIVD.
dination), gait disorder (apractic–atactic or small stepped),
imbalance and falls, urinary frequency and incontinence,
dysarthria, dysphagia as well as extrapyramidal signs such
infarcts, focal and diffuse ischaemic WMLs and incom- as hypokinesia and rigidity (Cummings, 1994; Pohjasvaara
plete ischaemic injury (Erkinjuntti, 1987; Erkinjuntti et al., et al., 2003).
2000; Roman et al., 2002; Pantoni, 2010). The MRI corre- Although stroke due to small vessel disease seems to have
lates of small vessel disease are multiple lacunes, extensive a favourable short-term outcome, patients with this condi-
WMLs, perivascular spaces, microhaemorrhages and atro- tion seem to have a more malignant long-term outcome
phy (Erkinjuntti et al., 2000; Wardlaw et al., 2013). The onset compared with patients with other stroke subtypes (Melkas
is variable: as reported by Babikian and Ropper (1987), 60% et al., 2011). This probably relates on the one hand to the
of the patients had a slow onset and only 30% had an acute damage in the fronto-subcortical pathways that are espe-
onset of cognitive symptoms; the course was gradual without cially vulnerable in small vessel disease and on the other
(40%) and with (40%) acute deficits and fluctuating in only hand to the systemic nature of small vessel disease.
20%. There is often a clinical history of ‘prolonged transient
ischaemic attacks (TIAs)’ or ‘multiple TIAs’, which mostly
are small strokes without residual symptoms, with only mild 58.6.3 STRATEGIC INFARCT DEMENTIA
focal findings. Clinically, small vessel dementia is character-
ized by a subcortical cognitive syndrome plus pure motor Depending on the precise location, the time course and clini-
hemiparesis, bulbar signs and dysarthria, gait disorder, cal features of strategic infarct dementia are highly variable.
variable depressive illness, emotional lability and deficits in Strategic infarct dementia is characterized by focal, often
executive functioning (Pohjasvaara et al., 2003; Jokinen et al., small, ischaemic lesions involving specific sites critical for
2006). The early cognitive syndrome of small vessel disease is higher cortical functions. The cortical sites include the hip-
characterized by a dysexecutive syndrome with slowed infor- pocampal formation, angular gyrus and cingulate gyrus.
mation processing, usually mild memory deficit and behav- The subcortical sites leading to impairment are the thala-
ioural symptoms. The dysexecutive syndrome in small vessel mus, fornix, basal forebrain, caudate, globus pallidus and
disease includes impairment in goal formulation, initiation, the genu or anterior limb of the internal capsule (Tatemichi,
planning, organizing, sequencing, executing, set shifting 1990; Erkinjuntti and Hachinski, 1993).
What is vascular cognitive impairment? 637

58.6.4 AD WITH CVD DSM-5 was launched in 2013, introducing the concepts
of mild and severe neurocognitive disorders, the latter cor-
Accumulating evidence shows that different vascular fac- responding to dementia (American Psychiatric Association,
tors, including hypertension and stroke, increase the risk 2013). One of the subtypes of neurocognitive disorder is vas-
of AD and frequently CVD coexists with AD (Kalaria and cular neurocognitive disorder; among other subtypes there
Ballard, 1999; Skoog et al., 1999; DeCarli, 2004; de la Torre, are AD and frontotemporal disorder. The International
2004). This overlap is increasingly important in the oldest Society for Vascular Behavioral and Cognitive Disorders
old (>85 years). Clinical recognition of patients with AD (VASCOG) has launched its own definition of vascular
and CVD is problematic as evident from recent neuropatho- cognitive disorder based on DSM-5 criteria (Sachdev et al.,
logical series (Snowdon et al., 1997; Neuropathology Group, 2014), but a compact definition with capacity to standardize
Medical Research Council Cognitive Function and Aging research and clinical practice still remains to be formulated.
Study, 2001; Schneider et al., 2007). These patients exhibit The two cardinal elements implemented in the clinical
a history of vascular risk and signs of CVD, providing a criteria for VaD are the definition of the cognitive syndrome
clinical picture that is close to VaD. However, fluctuating of dementia (Erkinjuntti et al., 1997) and the definition of
course and history of strokes were the only items differenti- the vascular cause of the dementia (Wetterling et al., 1996;
ating AD from AD with CVD (Moroney et al., 1997b). AD Pohjasvaara et al., 2000b). Variation in defining these two
with CVD patients may present clinically either as AD with critical elements has caused different definitions in use to
evidence of CVD lesions on brain imaging or with features give different point prevalence estimates, identify different
of both AD and VCI (Rockwood et al., 1999). It remains a groups of subjects and consequently also identify different
major clinical undertaking to distinguish dementia due to types and distribution of brain lesions. Further, this hetero-
AD from that arising from CVD in view of the considerable geneity may have been a factor in negative results in prior
overlap (Langa et al., 2004). Both result in cognitive, func- clinical trials on VaD (Inzitari et al., 2000). All the clini-
tional and behavioural impairments. There are also shared cal criteria used are consensus criteria, which are neither
pathophysiological mechanisms (e.g. WMLs, delayed neu- derived from prospective community-based studies on vas-
ronal death and apoptosis) (Pantoni and Garcia, 1995; cular factors affecting the cognition nor based on detailed
Snowdon et al., 1997; Skoog et al., 1999), associated risk fac- natural histories. All the cited criteria are mainly based on
tors (e.g. age, education, arterial hypertension) (Skoog et al., the ischaemic infarct concept and designed to have high
1999; DeCarli, 2004) and neurochemical deficits, includ- specificity, although they have been poorly implemented
ing cholinergic neuronal dysfunction (Wallin et al., 2002; and validated.
Roman and Kalaria, 2006). Based on the findings from the The general NINDS-AIREN criteria include (1) empha-
Nun Study, it has been further suggested that CVD may play size heterogeneity of VaD syndromes and pathologic sub-
an important role in determining the presence and severity types including not only ischaemic stroke but also other
of clinical symptoms of AD (Snowdon et al., 1997). A better causes of CVD including cerebral hypoxic–ischaemic
solution to recognizing patients with AD plus CVD would events, WMLs and haemorrhagic strokes; (2) recognize
be to discover reliable biological markers of clinical AD. the variability in clinical course, which may be static,
Potential markers include early prominent episodic mem- remitting or progressive; (3) highlight the question of the
ory impairment, early and significant medial temporal lobe location of ischaemic lesions and the need to establish a
atrophy on MRI, bilateral parietal hypoperfusion on single causal relations between vascular brain lesions and cogni-
photon emission CT or positron emission tomography, tion; (4) recognize the need to establish a temporal rela-
findings in brain amyloid imaging or low concentrations tionship between stroke and dementia onset; (5) include
of cerebral spinal fluid amyloid-β42 (CSF Aβ42) peptide specific findings early in the course that support a vascular
(Dubois et al., 2007). rather than a degenerative cause; (6) emphasize the impor-
tance of brain imaging to support clinical findings and
(7) give value of neuropsychological testing to document
impairments in multiple cognitive domains. The NINDS-
58.7 CLINICAL CRITERIA FOR VCI
AIREN criteria handle VaD as a syndrome with different
aetiologies and different clinical manifestations and not
Since the 1970s, several general clinical criteria for VaD as a single entity and list possible subtypes to be used in
have been used (Pohjasvaara et al., 1997). Widely used cri- research studies. The focus is on consequences of CVD
teria for VaD include the Diagnostic and Statistical Manual but the criteria also take into account different aetiolo-
of Mental Disorders, 4th Edition (DSM-IV) (World Health gies. These criteria incorporate different levels of certainty
Organisation, 1993a; American Psychiatric Association, of the clinical diagnosis (probable, possible and definite).
1994), International Statistical Classification of Diseases, However, in randomized clinical trials using the NINDS-
10th Revision (ICD-10) (World Health Organisation, AIREN criteria, all potential subtypes have been lumped
1993b), the Alzheimer’s Disease Diagnostic and Treatment together as ‘general VaD’. The SIVD criteria of Erkinjuntti
Center (ADDTC) criteria (Chui et al., 1992) and the NINDS- et al. (2000) represent an attempt to define a more homog-
AIREN criteria (Roman et al., 1993). enous subtype.
638 Dementia

In a neuropathological series, the sensitivity of the a recommendation on minimum, common, clinical and
NINDS-AIREN criteria for probable and possible VaD was research standards for the description and study of VCI.
58% and specificity was 80% (Gold et al., 1997). The crite-
ria successfully excluded AD in 91% of cases; the propor-
tion of combined cases misclassified as probable VaD was
29%. Compared to the other modern criteria (the ADDTC 58.9 CONCLUSIONS
criteria), the NINDS-AIREN criteria were more specific
and better at excluding combined cases (54% vs. 29%). In VCI is the modern term related to vascular burden of the
another series, the sensitivity of the NINDS-AIREN criteria brain, reflecting all the encompassing effects of CVD on cog-
for probable VaD was 20% and specificity 93%; the corre- nition. VCI includes all levels of cognitive decline from mild
sponding figures for probable ADDTC were 25% and 91% deficits in one or more cognitive domain to a broad dementia-
(Gold et al., 2002). The inter-rater reliability of the NINDS- like syndrome. VCI incorporates the complex interactions
AIREN criteria is moderate to substantial (κ 0.46–0.72) between vascular risk factors, CVD aetiologies and cellular
(Lopez et al., 1994). changes within the brain and cognition. Vascular risk factors
towards VCI include arterial hypertension, high cholesterol
and diabetes. VCI includes the common PSD and VaD. The
main subtype of VCI is caused by cerebral small vessel disease
and other subtypes are caused by large vessel disease, strategic
58.8 CLINICAL CRITERIA FOR VCI infarcts or a combination of AD and CVD. Traditional vas-
cular risk factors and stroke are also independent factors for
The concept and definition of VCI or vascular CIND are the clinical presentation of AD. In addition to these vascular
still evolving but it seems clear that the diagnosis should factors, CVD/strokes, infarcts and WMLs may trigger and
not be confined to a single aetiology comparable to the tra- modify the progression of AD. While CVD is preventable and
ditional ‘pure AD’ concept. The three main factors to be treatable it clearly is a major factor in the prevalence of cog-
defined in VCI are the severity of cognitive impairment, nitive impairment worldwide. The new VCI harmonization
the pattern of affected cognitive domains and the assumed standards are an important step towards international coop-
aetiologies and brain changes. It is important to avoid new eration and the new imaging methods that enable assessment
arbitrary threshold criteria, recognize a continuum of cog- of small vessels pave way for innovative intervention studies.
nitive impairment, focus on causes instead of consequences
and acknowledge complex interactions of coexisting differ-
ent aetiologies and brain changes. General VCI criteria are REFERENCES
probably not needed. It is better to avoid arbitrary clinical
threshold criteria such as the old AD type dementia crite- American Psychiatric Association (1994). Diagnostic and
ria. General common VCI criteria may hamper discovery of Statistical Manual of Mental Disorders, Fifth Edition.
interactive processes related to the continuum of individual Washington, DC: American Psychiatric Association.
cognitive changes. Separate criteria, designed according to American Psychiatric Association (2013). Diagnostic and
the target group, the assumed biological surrogate and the Statistical Manual of Mental Disorders, Fifth Edition.
assumed clinical outcomes would be better. Washington, DC: American Psychiatric Association.
The target groups may be Babikian, V. and Ropper, A.H. (1987). Binswanger’s dis-
ease: A review. Stroke, 18: 2–12.
1. CVD patients at risk of cognitive impairment Basile, A.M., Pantoni, L., Pracucci, G. et al. (2006). Age,
2. VCI patients at risk of further cognitive and functional hypertension, and lacunar stroke are the major deter-
decline minants of the severity of age-related white matter
3. Non-demented, non-disabled patients with extensive changes. The LADIS (Leukoaraiosis and Disability in the
WMLs at risk of functional decline Elderly) study. Cerebrovascular Diseases, 21: 315–322.
4. AD patients with WMLs at risk of cognitive and func- Bowler, J.V. (2007). Modern concept of vascular cognitive
tional decline impairment. British Medical Bulletin, 83: 291–305.
5. Elderly with gait instability Bowler, J.V. and Hachinski, V. (1995). Vascular cognitive
6. Depressed elderly with dysexecutive syndrome impairment: A new approach to vascular dementia.
Baillieres Clinical Neurology, 4: 357–376.
Potential surrogates include white matter burden, lacu- Bowler, J.V., Steenhuis, R. and Hachinski, V. (1999).
nar infarct burden as well as frontal cortical atrophy. The Conceptual background of vascular cognitive impair-
clinical outcomes may relate to cognitive functions, behav- ment. Alzheimer Disease and Associated Disorders, 13:
ioural symptoms, ADLs or gait and stability. S30–S37.
The vascular cognitive impairment harmonization stan- Brun, A. (1994). Pathology and pathophysiology of cere-
dards (Hachinski et al., 2006) are an important step to brovascular dementia: Pure subgroups of obstructive
guide and harmonize VCI studies wordwide. They include and hypoperfusive etiology. Dementia, 5: 145–147.
What is vascular cognitive impairment? 639

Brust, J.C. (1988). Vascular dementia is overdiagnosed. Erkinjuntti, T., Haltia, M., Palo, J. et al. (1988). Accuracy of
Archives of Neurology, 45: 799–801. the clinical diagnosis of vascular dementia: A prospec-
Chui, H.C. (1989). Dementia: A review emphasizing clini- tive clinical and post-mortem neuropathological study.
copathologic correlation and brain–behavior relation- Journal of Neurology Neurosurgery and Psychiatry, 51:
ships. Archives of Neurology, 46: 806–814. 1037–1044.
Chui, H.C. (2001). Vascular dementia, a new beginning. Erkinjuntti, T., Inzitari, D., Pantoni, L. et al. (1999b).
Shifting focus from clinical phenotype to ischemic Limitations of clinical critera for the diagnois of vascu-
brain injury. Neurologic Clinics, 18: 951–977. lar dementia in clinical trials: Is a focus on subcortical
Chui, H.C., Mack, W., Jackson, J.E. et al. (2000). Clinical vascular dementia a solution? Annals of the New York
criteria for the diagnosis of vascular dementia. Academy of Sciences, 903 (1): 262–272.
Archives of Neurology, 57: 191–196. Erkinjuntti, T., Inzitari, D., Pantoni, L. et al. (2000).
Chui, H.C., Victoroff, J.I., Margolin, D. et al. (1992). Criteria Research criteria for subcortical vascular dementia
for the diagnosis of ischemic vascular dementia pro- in clinical trials. Journal of Neural Transmission, 59
posed by the State of California Alzheimer’s Disease (Suppl. 2): 23–30.
Diagnostic and Treatment Centers. Neurology, 42: Erkinjuntti, T., Ostbye, T., Steenhuis, R. and Hachinski, V.
473–480. (1997). The effect of different diagnostic criteria on
Cummings, J.L. (1993). Fronto-subcortical circuits and the prevalence of dementia. New England Journal of
human behavior. Archives of Neurology, 50: 873–880. Medicine, 337: 1667–1674.
Cummings, J.L. (1994). Vascular subcortical dementias: Erkinjuntti, T. and Pantoni, L. (2000). Subcortical vas-
Clinical aspects. Dementia, 5: 177–180. cular dementia. In Gauthier, S. and Cummings, J.L.
de la Torre, J.C. (2004). Alzheimer’s disease is a vaso- (eds.), Alzheimer’s Disease and Related Disorders
cognopathy: A new term to describe its nature. Annual. London, United Kingdom: Martin Duniz Ltd,
Neurological Research, 26: 517–524. pp. 101–133.
DeCarli, C. (2004). Vascular factors in dementia: An Fratiglioni, L., Launer, L.J., Andersen, K. et al. (2000).
overview. Journal of the Neurological Sciences, 226: Incidence of dementia and major subtypes in Europe:
19–23. A collaborative study of population-based cohorts.
DeCarli, C. and Scheltens, P. (2002). Structural brain imag- Neurology, 54 (Suppl. 5): S10–S15.
ing. In Erkinjunti, T. and Gauthier, S. (eds.), Vascular Gold, G., Bouras C., Canuto, A. et al. (2002).
Cognitive Impairment, First Edition. London, United Clinicopathological validation study of four sets of
Kingdom: Martin Duniz Ltd, pp. 433–457. clinical criteria for vascular dementia. American Journal
Desmond, D.W. (1996). Vascular dementia: A construct of Psychiatry, 159: 82–87.
in evolution. Cerebrovascular and Brain Metabolism Gold, G., Giannakopoulos, P., Montes-Paixao, J.C. et al.
Reviews, 8: 296–325. (1997). Sensitivity and specificity of newly proposed
Dubois, B., Feldman, H., Jacova, C. et al. (2007). Research clinical criteria for possible vascular dementia.
criteria for the diagnosis of Alzheimer’s disease: Neurology, 49: 690–694.
Revising the NINCDS-ADRDA criteria. Lancet Gorelick, P.B. (1997). Status of risk factors for dementia
Neurology, 6: 734–746. associated with stroke. Stroke, 28 (2): 459–463.
Englund, E., Brun, A. and Alling, C. (1988). White matter Hachinski, V.C. (1990). The decline and resurgence of vas-
changes in dementia of Alzheimer’s type. Biochemical cular dementia. Canadian Medical Journal Association,
and neuropathological correlates. Brain, 111: 142 (2): 107–111.
1425–1439. Hachinski, V. (2008). Shifts in thinking about dementia.
Erkinjuntti, T. (1987). Types of multi-infarct dementia. Acta JAMA, 300: 2172–2173.
Neurologica Scandinavica, 75: 391–399. Hachinski, V.C., Iadecola, C., Petersen C.R. et al. (2006)
Erkinjuntti, T. (1999). Cerebrovascular dementia. National Institute of Neurological Disorders and
Pathophysiology, diagnosis and treatment. CNS Drugs, Stroke – Canadian Stroke Network vascular cogni-
12: 35–48. tive impairment harmonization standards. Stroke, 37:
Erkinjuntti, T. (2007). Vascular cognitive deterioration 2220–2241.
and stroke. Cerebrovascular Diseases, 24 (Suppl. 1): Hachinski V.C., Lassen N.A. and Marshall J. (1974). Multi-
189–194. infarct dementia. A cause of mental deterioration in
Erkinjuntti, T., Bowler, J.V., DeCarli, C. et al. (1999a). the elderly. Lancet, 304 (7874): 207–210.
Imaging of static brain lesions in vascular dementia: Hebert, R. and Brayne, C. (1995). Epidemiology of vascular
Implications for clinical trials. Alzheimer Disease and dementia. Neuroepidemiology, 14 (5): 240–257.
Associated Disorders, 13 (Suppl. 3): S81–S90. Inzitari, D., Erkinjuntti, T., Wallin, A. et al. (2000).
Erkinjuntti, T. and Hachinski, V.C. (1993). Subcortical vascular dementia as a specific target
Rethinking ­vascular dementia. Cerebrovascular for clinical trials. Annals of the New York Academy of
Disease, 3: 3–23. Sciences, 903: 510–521.
640 Dementia

Inzitari, D., Pracucci, G., Poggesi, A. et al. (2009). Changes Lopez, O.L., Larumbe, M.R., Becker, J.T. et al. (1994).
in white matter as determinant of global functional Reliability of NINDS-AIREN clinical criteria for the
decline in older independent outpatients: Three year diagnosis of vascular dementia. Neurology, 44:
follow-up of LADIS (leukoaraiosis and disability) study 1240–1245.
cohort. British Medical Journal, 339: b2477. Mahler, M.E. and Cummings, J.L. (1991). The behav-
Inzitari D., Simoni, M., Pracucci, G. et al. (2007). Risk of ioural neurology of multi-infarct dementia. Alzheimer
rapid global functional decline in elderly patients with Disease and Associated Disorders, 5: 122–130.
severe cerebral age-related white matter changes: McKhann, G., Drachman, D., Folstein, M. et al. (1984).
The LADIS Study. Archives of Internal Medicine, 167: Clinical diagnosis of Alzheimer’s disease: Report of
81–88. the NINCDS-ADRDA Work Group under the aus-
Jokinen, H., Kalska, H., Mäntylä, R. et al. (2005). White pices of Department of Health and Human Services
matter hyperintensities as a predictor of neuropsy- Task Force on Alzheimer’s Disease. Neurology, 34:
chological deficits post-stroke. Journal of Neurology 939–944.
Neurosurgery and Psychiatry, 76: 1229–1233. Melkas, S., Putaala, J., Oksala, N.K. et al. (2011). Small-
Jokinen, H., Kalska, H., Mäntylä, R. et al. (2006). Cognitive vessel disease relates to poor poststroke survival in
profile of subcortical ischemic vascular disease. Journal a 12-year follow-up. Neurology, 76: 734–739.
of Neurology Neurosurgery and Psychiatry, 77: 28–33. Moorhouse, P. and Rockwood, K. (2008). Vascular cogni-
Jokinen, H., Kalska, H., Ylikoski, R. et al. (2009a). MRI- tive impairment: Current concepts and clinical devel-
defined subcortical ischemic vascular disease: Baseline opments. Lancet Neurology, 7: 246–255.
clinical and neuropsychological findings. The LADIS Moroney, J.T., Bagiella, E., Desmond, D.W. et al. (1996).
study. Cerebrovascular Disease, 27: 336–344. Risk factors for incident dementia after stroke.
Jokinen, H., Kalska, H., Ylikoski, R. et al. (2009b). Role of hypoxic and ischemic disorders. Stroke, 27:
Longitudinal cognitive decline in subcortical ischemic 1283–1289.
vascular disease – The LADIS Study. Cerebrovascular Moroney, J.T., Bagiella, E., Desmond, D.W. et al. (1997a).
Disease, 27: 384–391. Cerebral hypoxia and ischemia in the pathogenesis
Jokinen, H., Lipsanen, J., Schmidt, R. et al. (2012). Brain of dementia after stroke. Annals of the New York
atrophy accelerates cognitive decline in cerebral Academy of Sciences, 826: 433–436.
small vessel disease: The LADIS study. Neurology, 78: Moroney, J.T., Bagiella, E., Hachinski, V.C. et al. (1997b).
1785–1792. Misclassification of dementia subtype using the
Jokinen, H., Schmidt, R., Ropele, S. et al. (2013). Diffusion Hachinski Ischemic Score: Results of a meta-analysis of
changes predict cognitive and functional outcome: patients with pathologically verified dementias. Annals
The LADIS study. Annals of Neurology, 73: 576–583. of the New York Academy of Sciences, 826: 490–492.
Kalaria, R.N. and Ballard, C. (1999). Overlap between Neuropathology Group. Medical Research Council
pathology of Alzheimer disease and vascular demen- Cognitive Function and Aging Study (2001).
tia. Alzheimer Disease and Associated Disorders, 13 Pathological correlates of late-onset dementia in a
(Suppl. 3): S115–S123. multicentre, community-based population in England
Kalaria, R.N., Kenny, R.A., Ballard, C.G. et al. (2004). and Wales. Neuropathology Group of the Medical
Towards defining the neuropathological substrates Research Council Cognitive Function and Ageing
of vascular dementia. Journal of the Neurological Study (MRC CFAS). Lancet, 357: 169–175.
Sciences, 226: 75–80. O’Brien, J.T., Erkinjuntti, T., Reisberg, B. et al. (2003).
Kivipelto, M., Ngandu, T., Laatikainen, T. et al. (2006). Risk Vascular cognitive impairment. Lancet Neurology, 2:
score for the prediction of dementia risk in 20 years 89–98.
among middle aged people: A longitudinal, popula- Pantoni, L. (2010). Cerebral small vessel disease: From
tion-based study. Lancet Neurology, 5: 735–741. pathogenesis and clinical characteristics to therapeutic
Langa, K.M., Forster, N.L. and Larson, E.B. (2004). Mixed challenges. Lancet Neurology, 9: 689–701.
dementia: Emerging concepts and therapeutic impli- Pantoni, L. and Garcia, J.H. (1995). The significance of
cations. JAMA, 292: 2901–2908. cerebral white matter abnormalities 100 years after
Leys, D., Henon, H., Mackowiak-Cordoliani, M.-A. and Binswanger’s report. A review. Stroke, 26: 1293–1301.
Pasquier, F. (2005). Poststroke dementia. Lancet Pantoni, L. and Garcia, J.H. (1997). Pathogenesis of leuko-
Neurology, 4: 752–759. araiosis: A review. Stroke, 28: 652–659.
Linden, T., Skoog, I., Fagerber, B. et al. (2004). Cognitive Pantoni, L., Poggesi, A. and Inzitari, D. (2009). Cognitive
impairment and dementia 20 months after stroke. decline and dementia related to cerebrovascular dis-
Neuroepidemiology, 23: 45–52. eases: Some evidence and concepts. Cerebrovascular
Lobo, A., Launer, L.J., Fratiglioni, L. et al. (2000). Disease, 27 (Suppl. 1): 191–196.
Prevalence of dementia and major subtypes in Europe: Pasquier, F. and Leys, D. (1997). Why are stroke patients
A collaborative study of population-based cohorts. prone to develop dementia? Journal of Neurology,
Neurology, 54 (Suppl. 5): S4–S9. 244: 135–142.
What is vascular cognitive impairment? 641

Petersen, R.C., Doody, R., Kurz, A. et al. (2002). Current Roth, M. (1971). Classification and aetiology in mental
concepts in mild cognitive impairment. Archives of disorders of old age: Some recent development. In
Neurology, 58: 1985–1992. Kay, D. and Walk, A. (eds.), Recent Developments in
Pohjasvaara, T., Erkinjuntti, T., Vataja, R. and Kaste, M. Psychogeriatrics. Ashford, United Kingdom: Headley
(1997). Dementia three months after stroke. Baseline Brothers Ltd.
frequency and effect of different definitions of demen- Sachdev, P., Kalaria, R., O’Brien, J. et al. (2014). Diagnostic
tia in the Helsinki Stroke Aging Memory Study (SAM) criteria for vascular cognitive disorders. A VASCOG
cohort. Stroke, 28 (4): 785–792. statement. Alzheimer Disease and Associated
Pohjasvaara, T., Erkinjuntti, T., Ylikoski, R. et al. (1998). Disorders, 28 (3): 206.
Clinical determinants of poststroke dementia. Stroke, Schneider, J.A., Arvanitakis, Z., Bang, W. and Bennet,
29: 75–81. D.A. (2007). Mixed brain pathologies account for most
Pohjasvaara, T., Mäntylä, R., Salonen, O. et al. (2000a). dementia cases in community-dwelling older persons.
How complex interactions of ischemic brain infarcts, Neurology, 69: 2197–2204.
white matter lesions and atrophy relate to post-stroke Seshadri, S. and Wolf, A.P. (2007). Lifetime risk of stroke
dementia. Archives of Neurology, 57: 1295–1300. and dementia: Current concepts, and estimates
Pohjasvaara, T., Mäntylä, R., Ylikoski, R. et al. (2000b). from the Framingham Study. Lancet Neurology, 6:
Comparison of different clinical criteria for the vascular 1106–1114.
cause of vascular dementia (ADDTC, DSM-III, DSM-IV, Shim, Y.S., Yang, D.W., Kim, B.S. et al. (2006). Comparison
ICD-10, NINDS-AIREN). Stroke, 31: 2952–2957. of regional cerebral blood flow in two subsets of
Pohjasvaara, T., Mäntylä, R., Ylikoski, R. et al. (2003). subcortical ischemic vascular dementia: Statistical
Clinical features of MRI-defined subcortical vascular parametric mapping analysis of SPECT. Journal of the
disease. Alzheimer Disease and Associated Disorders, Neurological Sciences, 250: 85–91.
17: 236–242. Skoog, I. (1994). Risk factors for vascular dementia:
Rockwood, K., Howard, K., MacKnight, C. and Darvesh, A review. Dementia, 5: 137–144.
S. (1999). Spectrum of disease in vascular cognitive Skoog, I. (1998). Status of risk factors for vascular demen-
impairment. Neuroepidemiology, 18: 248–254. tia. Neuroepidemiology, 17: 2–9.
Rockwood, K., Wenzel, C., Hachinski, V. et al. (2000). Skoog, I., Kalaria, R.N. and Breteler, M.M.B. (1999).
Prevalence and outcomes of vascular cognitive impair- Vascular factors and Alzheimer’s disease. Alzheimer
ment. Neurology, 54: 447–451. Disease and Associated Disorders, 13 (Suppl. 3):
Roman, G.C. (1985). The identity of lacunar dementia and S106–S114.
Binswanger disease. Medical Hypotheses, 16: 389–391. Snowdon, D.A., Greiner, L.H., Mortimer, J.A. et al.
Roman, G.C. (1987). Senile dementia of the Binswanger (1997). Brain infarction and the clinical expression
type. A vascular form of dementia in the elderly. of Alzheimer disease. The Nun Study. JAMA, 277:
JAMA, 258: 1782–1788. 813–817.
Roman, G.C. (2008). Have we forgotten the cerebrovas- Sulkava, R. and Erkinjuntti, T. (1987). Vascular dementia
cular circulation. Alzheimer Disease and Associated due to cardiac arrhythmias and systemic hypotension.
Disorders, 22: 1–3. Acta Neurologica Scandinavica, 76: 123–128.
Roman, G.C. (2001). Historic evolution of the concept Tatemichi, T.K. (1990). How acute brain failure becomes
of dementia: A systematic review from 2000 BC to chronic. A view of the mechanisms and syndromes of
2000 AD. In Qizilbash, N. et al. (eds.), Evidence- dementia related to stroke. Neurology, 40: 1652–1659.
Based Dementia: A Practical Guide to Diagnosis and Tomlinson, B.E., Blessed, G. and Roth, M. (1970).
Management. Oxford, England: Blackwell Science. Observations on the brains of demented old people.
Roman, G.C., Erkinjuntti, T., Wallin, A. et al. (2002). Journal of the Neurological Sciences, 11: 205–242.
Subcortical ischaemic vascular dementia. Lancet Vataja, R., Pohjasvaara, T., Mäntylä, R. et al. (2003). MRI
Neurology, 1: 426–436. correlates of executive dysfunction in patients with
Roman, G.C. and Kalaria, R.N. (2006). Vascular deter- ischaemic stroke. European Journal of Neurology, 10:
minants of cholinergic deficits in Alzheimer disease 625–631.
and vascular dementia. Neurobiology of Aging, 27: Verdelho, A., Madureira, S., Moleiro, C. et al. (2010). White
1769–1785. matter changes and diabetes predict cognitive decline
Roman, G.C., Sachdev, P., Royall, D.R. et al. (2004). Vascular in the elderly: The LADIS study. Neurology, 75: 60–167.
cognitive disorder: A new diagnostic category updat- Vinters, H.V., Ellis, W.G., Zarow, C. et al. (2000).
ing vascular cognitive impairment and vascular demen- Neuropathologic substrates of ischemic vascular
tia. Journal of the Neurological Sciences, 226: 81–87. dementi. Journal of Neuropathology and Experimental
Roman, G.C., Tatemichi, T.K., Erkinjuntti, T. et al. (1993). Neurology, 60: 658–659.
Vascular dementia diagnostic criteria for research Wallin, A., Blennow, K. and Gottfries, C.G. (2002).
studies: Report of the NINDS-AIREN International Neurochemical abnormalities in vascular dementia.
Work Group. Neurology, 43: 250–260. Dementia, 1: 120–130.
642 Dementia

Wardlaw, J.M., Smith, E.E., Biessels, G.J. et al. (2013). dementia (ADDTC, DSM-IV, ICD-10, NINDS-AIREN).
Neuroimaging standards for research into small Stroke, 27: 30–36.
­vessel disease and its contribution to ageing World Health Organization (1993). ICD-10
and ­neurodegeneration. Lancet Neurology, 8: 822–836. Classification of Mental and Behavioural Disorders:
Wetterling, T., Kanitz, R.D. and Borgis, K.J. (1996). Diagnostic Criteria for Research. Geneva,
Comparison of different diagnostic criteria for vascular Switzerland: WHO.
59
The neuropathology of vascular dementia

RAJ N. KALARIA

results from cortical infarcts attributed to large vessel disease,


59.1 INTRODUCTION whereas subcortical ischaemic or Binswanger type of VaD
involving subcortical structures and the WM results from
Cerebrovascular disease (CVD) is the second most common intracranial small vessel changes (Table 59.1).
cause of age-related dementia. Alzheimer and Kraeplin
(Berrios and Freeman, 1991) both entertained the notion
that gradual strangulation of the brain causes cognitive and
59.2 FROM ARTERIOSCLEROTIC
behavioural deficits. They reasoned that progressive hard-
ening of the arteries led to arteriosclerotic dementia. Until
DEMENTIA TO VASCULAR
the late 1960s, arteriosclerotic dementia was commonly COGNITIVE IMPAIRMENT
described in hospital records and attributed to cerebral soft-
ening with loss of relatively large volume, at least 50 mL, Several terms are commonly used to relate the clinical and
and preferably 100 mL of brain tissue (Tomlinson et al., pathological descriptions of vascular disease to cognitive
1970). It was often clinically overdiagnosed in comparison impairment and dementia. Cerebrovascular dementia or
to Alzheimer’s disease (AD). The current concepts of vascu- more commonly VaD implies a clinically diagnosed demen-
lar dementia (VaD), including pathology, have evolved over tia syndrome comprising subtypes with both ischemic and
a long period, often biased by AD diagnosis. haemorrhagic aetiologies (Román, 2004). Vascular cognitive
VaD is now recognized as a cognitive disorder explained impairment (VCI) (Bowler and Hachinski, 1995) was devised
by vascular causes in the absence of other pathologies. The to incorporate conditions in any cognitive domain that have
first clear recognition of subclasses of VaD, probably, should a vascular origin or impaired brain perfusion (O’Brien et al.,
be credited to Otto Binswanger, who described subcortical 2003). However, it has been challenging to establish the
arteriosclerotic encephalopathy or a type of small vessel dis- degree of pathological burden that relates to impaired cogni-
ease (SVD) related to dementia upon pathological verifica- tion (O’Brien et al., 2003; Gorelick et al., 2011).
tion of cerebral white matter (WM) disorder in patients with Like VCI, VCD (Sachdev, 1999) is a continuum described
hypertensive disease (Berrios and Freeman, 1991). In the more by cognitive disorders of vascular aetiology with diverse
recent times, based on extensive experience, C. Miller Fisher pathologies and clinical manifestations. The Diagnostic and
proposed that cerebrovascular dementia is a matter of both Statistical Manual of Mental Disorders, Fifth Edition (DSM-5)
large and small strokes and provided lucid accounts of lacunar criteria and guidelines propose that the categories of mild
syndromes (Fisher, 1982). Multiple small infarcts in associa- and major vascular cognitive disorders can be recognized
tion with hypertension (état lacunaire) are one of the com- (American Psychiatric Association, 2013). Vascular cognitive
monest pathological changes linked to VaD. It is characterized disorder has also been suggested to indicate the global diag-
by abrupt episodes, which lead to weakness, slowness, dysar- nostic category, restricting the term ‘VCI’ to patients whose
thria, dysphagia, small-stepped gait, brisk reflexes and exten- cognitive impairment falls short of dementia (Román, 2004).
sor plantar responses. All these signs are usually present by The major neurocognitive disorder classification, as a sub-
the time mental deterioration occurs (Hachinski et al., 1974). stitute for VaD, appears to fit better with patients and more
The recognition of subtypes of clinical VaD was an impor- adapted to neurodegenerative cognitive disorders for which
tant step towards current pathological classifications based memory impairment is not predominant but would com-
on vascular aetiology. Multi-infarct dementia predominantly prise substantial frontal lobe pathology (Sachdev et al., 2014).

643
644 Dementia

Table 59.1 VaD subtypes defined by blood vessel size Table 59.2 Key variables to define pathology of VaD
and pathological process
Identify as ischaemic or haemorrhagic infarct(s)
Large vessel dementia (multiple infarcts or MID) Presence of lacunes and lacunar infarcts: état lacunaire
Small vessel dementia (microinfarction) (grey matter) and état crible (WM)
Location of infarcts: cortex, WM, basal ganglia,
Strategic infarct dementia (infarcts in strategic locations
brainstem (pontine), cerebellum
e.g. thalamus)
Circulation involved: arterial territories-anterior, middle
Hypoperfusive dementia
or posterior
Dementia related to angiopathies (hypertension, amyloid) Laterality: right or left anterior and posterior
Haemorrhagic dementia Sizes and number of infarcts = dimension: 0–4 mm,
Other causes of VaD (vasculitis) 5–15 mm, 16–30 mm, 31–50 mm and >50 mm, if size
Familial VaD (CADASIL, CARASIL, RVCL) <5 mm determine as small or microinfarcts
Presence and location of SVD: lipohyalinosis, fibroid
Source: Natte, R. et al., Annals of Neurology, 50, 765–772, 2001.
Notes: Familial or hereditary forms of cerebral amyloid angiopa-
necrosis, CAA
thy involving ischaemic strokes and intracerebral haem- Presence of WM disease: rarefaction or incomplete
orrhages may also lead to cognitive impairment and infarction
stroke. Degree of microgliosis and astocytosis: mild, moderate
Abbreviations: VaD, vascular dementia; MID, multi-infarct demen- or severe
tia; CADASIL, cerebral autosomal dominant arteriopathy
with subcortical infarcts and leukoencephalopathy
Presence of Alzheimer pathology (including NFT and
(Kalimo and Kalaria, 2005); CARASIL, cerebral autosomal neuritic plaque staging). If degree > stage III, the case
recessive arteriopathy with subcortical infarcts and leuko- is mixed AD and VaD
encephalopahty (Hara et al., 2009); RVCL, autosomal Presence of hippocampal sclerosis
dominant retinal vasculopathy with cerebral leukodystro-
phy (Kavanagh et al., 2008). Source: Hachinski, V. et al., Stroke, 37, 2220–2241, 2006.
Notes: 
For reporting purposes, each of above features can be
scored numerically to provide a summary. For example, 0 is
Cognitive impairment or dementia is relatively common absent and 1 means present. Less frequent lesions ­including
following stroke (Leys et al., 2005; Pendlebury and Rothwell, watershed infarcts and laminar necrosis. Increasing numeri-
2009). Incident dementia or post-stroke dementia (PSD) cal value may also be assigned to the infarcts.
Abbreviations: WM, white matter; SVD, small vessel disease; CAA,
may develop within 3 months or after a stabilization period
cerebral ­amyloid angiopathy; NFT, neurofibrillary tangles;
of 1 year or longer after stroke injury (Pohjasvaara et al., AD, Alzheimer’s disease.
1997; Bejot et al., 2011; Allan et al., 2012). PSD can have a
complex aetiology with varying combinations of large SVD
as well as non-vascular pathology. Stroke injury or CVD have been implicated in VaD (Markesbery, 1998). Factors
may unmask other pre-existing disease processes, such as that define pathology in subtypes of VaD include multiplic-
AD although it has been demonstrated that at least 75% of ity, size, anatomical location, laterality and age of the lesions
PSD cases are pathologically confirmed as post-stroke VaD besides genetic influences and previous existence of sys-
with little or no AD pathology (Allan et al., 2012). temic vascular disease. Subcortical ischaemic VaD is likely
Recent advances in neuroimaging and systematic neuro- to be the most significant subtype of VaD (Román, 2002),
pathological examination have enabled better definitions of and it seems that smaller subcortical lesions are key players
clinically diagnosed cerebrovascular disorders, which cause (Table 59.2).
cognitive impairment (Hachinski et al., 2006). The patho-
logical diagnosis of VaD or VCI, however, requires system-
atic evaluation of potentially relevant clinical or phenotypic
59.3 PREVALANCE OF PATHOLOGICALLY
features with particular attention to timing of events
(Ferrer, 2010). It is difficult to define which neuropathologi-
DIAGNOSED VAD
cal changes and to what degree these changes contribute
to dementia because of the heterogeneous localization of As with AD, confirmation of VaD diagnosis is definitive at
lesions and the coexistence of other pathologies including autopsy derived from appropriate sampling of both cerebral
neurodegenerative changes such as those in AD. More than hemispheres and minimal neuropathological examination
one factor may contribute to the overall impairment and the (Hachinski et al., 2006) to rule out significant pathological
VaD phenotype (Table 59.2). These include origin and type changes associated with other dementias. The prevalence
of vascular occlusion, presence of haemorrhage, distribution of early-onset dementia VaD (<65-years old) ranges from
of arterial territories and the size of vessels involved. Thus, 3.1% to 44% in various clinical and population-based stud-
many brain regions including the territories of the anterior, ies across the world (Vieira et al., 2013). These values may
posterior and middle cerebral arteries; the angular gyrus, not reflect true prevalence and incidence rates of VaD due
caudate and medial thalamus in the dominant hemisphere; to inconsistencies in diagnostic criteria, sampling methods,
the amygdala and hippocampus as well as the hippocampus subject or country demographics and variation in morbidity
The neuropathology of vascular dementia 645

Pure Other and colleagues observed that the total volume of infarcts
VaD types in demented stroke patients was usually over 50 mL and in
10% 2%
some cases was greater than 100 mL, exceeding that in non-
AD demented stroke patients (Blessed et al., 1968; Tomlinson
40%
et al., 1970). Subsequent clinicopathological studies reported
Mixed that only 5 out of 23 patients with a pathological diagnosis of
(SVD)
VaD had more than 50 mL of infarcted tissue and 7 had less
40%
than 10 mL (Erkinjuntti et al., 1988). It is now apparent that
Mixed
Mixe widespread small ischaemic lesions or multiple microinfarcts
(LV)
(LV (White, 2009; Westover et al., 2013) distributed throughout
8%
the CNS, correlate with dementia and are key predictors of
Figure 59.1 Pathological outcomes of clinically diagnosed cognitive impairment (Kalaria, 2012). Location of lesions
vascular dementia (VaD). Mixed type 1 revealed large may also be more critical than total volume (Desmond et al.,
infracts, whereas mixed type 2 predominantly exhibited 2000; Erkinjuntti et al., 2000). For example, infarction in
small vessel disease (SVD) with microinfarction. Other the left hemisphere disproportionately increases the risk of
included Lewy body disease, dementia, mild Parkinson dementia (Gorelick et al., 1992; Liu et al., 1992; Censori et al.,
disease and depression. AD, Alzheimer’s disease; LV, large 1996; Pohjasvaara et al., 1998; Desmond et al., 2000). Bilateral
vessel. infarcts with more involvement of the dominant hemisphere
also increase the risk of dementia after stroke (Erkinjuntti
and mortality trends (Figure 59.1). When a range of clini- et al., 1988; del Ser et al., 1990; Liu et al., 1992).
cal criteria have been applied to sample sizes of 59 to 1929, Relatively few prospective studies have validated criteria
autopsy studies demonstrate pathologically diagnosed VaD for VaD. Previous criteria for Binswanger’s disease or cerebral
ranges from as low as 0.03% to as high as 58% with an over- SVD (Bennett et al., 1990) proposed that the clinical diagnosis
all mean estimate of 17% (Markesbery, 1998; Jellinger, 2008). of dementia accompanied by neuromaging evidence (comput-
In western countries, the estimated rates of pathologically erized axial tomography [CT] or magnetic resonance imag-
diagnosed VaD as defined by various criteria lie between ing [MRI]) of bilateral abnormalities and at least two of three
8% and 15%. In studies where diagnosis was restricted to findings included evidence of (1) a vascular risk factor or sys-
the current National Institute of Neurological Disorders temic vascular disease, (2) focal cerebrovascular disease and
and Stroke–Association Internationale pour la Recherché (3) ‘subcortical’ cerebral dysfunction described by gait disor-
et l’Enseignement en Neurosciences (NINDS-AIREN) crite- der, parkinsonism or incontinence. These criteria were vali-
ria (Román et al., 1993), the frequencies were reported to be dated in a prospective series of 184 patients with AD and only
~7%. Taking all these estimates into account, the worldwide 1.6% of the patients were diagnostically misclassified when all
frequency of VaD in autopsy verified cases is determined to the three clinical criteria were met (Bennett et al., 1990).
be 10%–15%, being marginally less than when clinical criteria The Oxford Project to Investigate Memory and Ageing
alone are used (Barker et al., 2002; Knopman et al., 2003). In (OPTIMA) study has developed a simple, novel, image-match-
Japan, the incidence of autopsy verified VaD was previously ing scoring system (Smallwood et al., 2012) to relate the extent
reported to be 35% (Seno et al., 1999) and later 22% (Akatsu of SVD with cognitive function in a study of 70 cases with
et al., 2002). Population-based cohorts should provide the insufficient pathology to the meet criteria involved in diagno-
best estimates for pathology verified VaD, but there are only sis of AD. Severity of SVD pathology was inversely related to
few such studies and all existing ones show that microvas- cognitive scores and 43% of the cases with high SVD scores
cular lesions occur more frequently than neurodegenerative were designated to be demented. To better define clinico-
lesions in elderly c­ ommunity-dwelling subjects with demen- pathological correlation in subtypes of VaD, including SVD,
tia (Neuropathology Group of the Medical Research Council a staging system related to the natural history of cerebrovas-
Cognitive Function and Ageing Study [MRC CFAS], 2001; cular pathology and an algorithm for the neuropathological
White et al., 2005; Schneider et al., 2007; Sonnen et al., 2007). quantification of the CVD burden in dementia have been pro-
posed (Deramecourt et al., 2012). The staging system (I–VI)
needs further evaluation against cognitive function scores to
determine whether this system would be useful in large-scale
59.4 PATHOLOGICAL DIAGNOSIS
studies to understand clinicopathological correlations.
OF VAD Neuropathological diagnosis of VaD should be based
on the absence of a primary neurodegenerative disease
Diagnostic criteria for the neuropathological validation known to cause dementia and the presence of cerebrovas-
of VaD are generally lacking. However, neuroimaging and cular pathology that defines one or more VaD subtypes
clinicopathological studies suggest that VaD is related to the (Table 59.1). These would also include dementia among
extent of cerebral damage caused to a patient. A combination post-stroke survivors who fulfil the NINDS-AIREN cri-
of factors including origin, volume, location and number of teria (Román et al., 1993) for probable VaD. Stroke sur-
lesions, contribute to the development of dementia. Blessed vivors with mild cognitive impairment or VCI (O’Brien
646 Dementia

et al., 2003) may also have sufficient pathology for neu- attributing causes to VaD. As discussed earlier, parenchymal
ropathological diagnosis of VaD. An approach to neuro- abnormalities of neurodegenerative type may be present that
pathological diagnostic evaluation of VaD was previously are not obviously associated with either vascular disease or
proposed by the Newcastle group (Figure 59.2). According systemic factors, i.e. Alzheimer type or hippocampal lesions.
to these criteria, there are two neuropathological diag-
nostic groups: probable VaD is based on the exclusion
of a primary neurodegenerative disease known to cause
dementia plus the presence of cerebrovascular pathology 59.5 DIFFERENT CEREBROVASCULAR
that defines one or more of the VaD subtypes. Possible VaD PATHOLOGIES LEAD TO
is designated to a patient when the brain contains vascu- DEMENTIA
lar pathology that does not fulfil the criteria for one of the
subtypes, but where no other explanation for dementia is Atherothromboembolism is the main cause of infarctions
found. Post-stroke cases are usually included in subtypes associated with major arterial territories, which
I–III. Cases with extensive WM disease in the absence of may be admixed in cortical and subcortical regions.
other significant features are included under SVD. Subtype Thromboembolic events may be responsible for up to 50%
I may result from large vessel occlusion (atherothrombo- of all ischaemic strokes, whereas intracranial SVD causes
embolism), artery-to-artery embolism or cardioembolism. 25% of the infarcts. Small vessel alternations involve
Subtype II usually involves arteriolosclerosis, lipohyalino- arteriolosclerosis and hyalionsis and associated with lacu-
sis and hypertensive, arteriosclerotic, amyloid or collagen nar infarcts and lacunes predominantly occurring in the
angiopathy. Subtypes I, II and V may result from aneu- subcortical structures. WM disease or subcortical leukoen-
rysms, arterial dissections, arteriovenous malformations cephalopathy with incomplete infarction and SVD are com-
and various forms of arteritis (vasculitis). mon pathological changes in CVD associated with dementia
Assessing the neuropathological substrates of VaD involves (Deramecourt et al., 2012). Other features include border
systematic assessment of parenchymal lesions, including zone (watershed) infarctions, laminar necrosis and amyloid
microinfarcts and haemorrhages and the vascular abnor- angiopathy. Complicated angiopathies such as fibromuscu-
malities that may have caused them to relate to progression lar dysplasia, arterial dissections, granulomatous angiitis,
of impairment (Mirra, 1997; Kalaria et al., 2004; Strozyk et al., collagen vascular disease and giant-cell arteritis are rarer
2010; Deramecourt et al., 2012; Smallwood et al., 2012). In causes of cerebrovascular disease and VaD (Table 59.1).
addition to this, systemic factors (e.g. hypotension, hypogly- Previous studies have recorded ischaemic, o ­edematous
cemia) may cause brain or neuronal lesions in the absence of and haemorrhagic lesions induced by pathological changes
severe vascular disease and should be taken in account when in the brain circulation or perfusion to be associated with

Newcastle categorization of the major CV lesions


associated with cognitive impairment
I II III IV V VI

Large infarct or Multiple small or Strategic Cerebral Cerebral CV lesions


several infarcts microinfarcts infarcts hypoperfusion haemorrhage with AD pathology

Multi-infarct White matter Thalamus Hippocampal Lobar Mixed


dementia lesions hippocampus sclerosis ICH dementia
basal forebrain SAH

Figure 59.2 (See colour insert.) Schematic diagram of different cerebrovascular pathologies associated with dementia.
The proposed Newcastle categorization includes six subtypes (Kalaria et al., 2004). In all of the subtypes, the age of the
vascular lesion(s) should correspond with the time when disease began. The post-stroke cases are usually included in
subtypes I–III. While these may not be different from other published subtypes (Jellinger, 2008), they are practical and
simple to use. Cases with extensive white matter (WM) disease in the absence of significant other features are included
under SVD. Subtype I may result from large vessel occlusion (atherothromboembolism), artery-to-artery embolism or
cardioembolism. Subtype II usually involves descriptions of arteriosclerosis, lipohyalinosis, hypertensive, arterioscle-
rotic, amyloid or collagen angiopathy. Subtypes I–II and V may result from aneurysms, arterial dissections, arteriove-
nous malformations and various forms of arteritis (vasculitis). CAA, cerebral amyloid angiopathy; ICH, intracerebral
haemorrhage; SAH, subarachnoid haemorrhage. (Original diagram courtesy of Dr K. Nagata, Akita, Japan.)
The neuropathology of vascular dementia 647

Types of vascular and hippocampal lesions in VaD and smaller vessels, beyond the circle of Willis involving
80
proximal segments of the middle and anterior cerebral arter-
60
ies, is generally rare but may be found in very old subjects
40 (Kalaria et al., 2012). The presence of dolichoectasia and fusi-
20 form aneurysms has also been noted in some cases. In severe
0 cases, medium-sized arteries in the leptomeninges and prox-
% Cases imal perforating arteries are involved. The damage could be
ns

s
ct
io

s
worse depending upon the presence of hypertension.

ts
ct
ar
ct

A
rc
ar
nf
ar

H
CA
fa
nf

IC
Arterial territorial infarctions involve four principal
nf

ri

is
in
oi
na
ei

s
ro
ic
icr
et

cu

areas, particularly, those supplied by the major arteries:


st

cle
l/m
pl

Cy
La

ls
m

al

anterior, middle cerebral artery, posterior artery and the

pa
Co

Sm

am
territory between the anterior and middle cerebral artery.

oc
pp
Hi The intensity of gliosis, both astrocytic and microgliosis, is
an important consideration in judging the degree and age
Figure 59.3 Types of vascular and hippocampal lesions
reported in VaD. Data compiled from 214 cases reported
of infarction. However, there is no clear evidence to suggest
in previous studies (Kalaria et al., 2004; Kalaria et al., that these are related to cognitive impairment. Degrees of
unpublished data). Percentage of cases is averaged from gliosis or glial scars may be noted in VaD brains subjected
two or more reported studies. Cystic infarcts (possibly to global ischaemia i.e. after transient cardiac arrest where
also lacunar) with typically ragged edges were admixed in responses may be observed in vulnerable neuronal groups
both cortical and subcortical structures. Lacunar infarcts within the hippocampus or neocortical laminae.
were mostly in the basal ganglia and white matter.

VaD (Table 59.2). In 10 different studies where VaD was diag-


nosed, 78% of cases revealed cortical and subcortical infarcts 59.7 CEREBRAL SVD
suggesting that other v­ ascular pathologies involving incom-
plete infarction or border zone infarcts could be important SVD per se involves hyalinization of vessels, expansion of the
factors (Figure 59.3). Among other lesions, 25% of cases perivascular space and pallor of adjacent perivascular myelin,
had cystic infarcts whereas 50% showed lacunar infarcts with associated astrocytic gliosis (Figure 59.4). The smaller
or microinfarcts. Lacunar infarcts, however, appear to be vessels of the brain including intracerebral end arteries and
a common category of infarcts and currently recognized as arterioles undergo progressive age-related changes (Lammie,
the most frequent cause of stroke. Severe amyloid angiopathy 2000), which alter perfusion and cause lacunar infarcts (cystic
was present in 10% of the cases. Hippocampal sclerosis and lesions generally < 1 cm) and microinfarcts. The arteriolar
cell atrophy, which may be caused by remote ischaemic changes range from wall thickening by hyalinosis, reduction
injury, was apparent in 55% of cases in a study based on clini- or increment of the intima to severe arteriolosclerosis and
cal diagnosis of ischaemic VaD (Vinters et al., 2000). fibroid necrosis. Arteriolosclerotic changes likely promote loss
of elasticity to dilate and constrict in response to variations
of systemic blood pressure or auto-regulation, which in turn
causes fluctuations in blood flow response and changes in tis-
59.6 L ARGE VESSEL DISEASE sue perfusion. The deep cerebral structures and WM would be
rendered most vulnerable because the vessels are end arteries,
Large infarction or macroinfarction is defined as that vis- almost devoid of anastomoses. Small vessel pathology could
ible upon gross examination of the brain at autopsy. Stenosis also lead to oedema and damage of the blood–brain barrier
arising from atherosclerosis within large vessels is consid- (BBB) with chronic leakage of fluid and macromolecules in the
ered to be the main cause of large infarction, which may WM (Ho and Garcia, 2000; Wardlaw, 2010; Giwa et al., 2012).
sometimes extend beyond the arterial territories. The stages Microvascular disease may also be associated with degrees
of atherosclerosis may vary from accumulation of foam cells of inflammation including the presence of lymphocytes or
causing fatty streaks to complicated atheromas involving macrophages localized on blood vessels (and not necessarily
extracellular matrix components and even viral or bacterial a function of brain ischaemia). In the oldest SVD cases, there
infections. Approximately 15% of VaD assumes occlusion of may also often be evidence of remote haemorrhage in the
the extracranial arteries such as, the internal carotid artery form of perivascular haemosiderin (Deramecourt et al., 2012).
and the main intracranial arteries of the circle of Willis
including the middle cerebral artery lead to multiple infarcts
and dementia (Brun, 1994). The differences between the
anterior versus posterior portions of the circle of Willis and 59.8 LACUNAR INFARCTS
left versus right sides may be variable and stenosis of major
arteries could be up to 75% in very severe cases. Typical ath- Lacunar infarcts, about 1 cm or less in diameter, may occur as
erosclerosis or microatheromatous disease in the meningeal complete or cavitating lesions frequently in both subcortical
648 Dementia

(a) (b) there were no pathological differences between symptom-


atic and asymptomatic patients. Perivascular oedema and
thickening, inflammation and disintegration of the arterio-
lar wall were common, whereas vessel occlusion was rare
(Bailey et al., 2012). In neuropathological studies of elderly
patients with vascular disease, but without evidence of AD or
other neurodegenerative pathologies, dementia was associ-
ated with severe cribriform change and associated with sub-
cortical WM damage and microinfarcts (Esiri et al., 1997).
(c) (d) In the Highly Accelerated Stress Screen (HAAS) analysis
(White, 2009), microvascular infarcts (lacunar and micro-
infarcts) were identified as the sole or dominant lesion in
34% of the definitely impaired decedents (Figure 59.4). Cases
of VaD without significant AD pathology show more severe
cribriform change and deep WM and grey matter lacunar
or microinfarcts than do stroke subjects with macroscopic
infarcts and elderly subjects without dementia (Smallwood
et al., 2012). Similarly, lacunar infarcts and microinfarcts
(e) (f )
were the most common neuropathological features in more
than 50% of elderly patients with ischaemic VaD (Vinters et
al., 2000) and also strong determinants of dementia in the
Geneva brain ageing study (Giannakopoulos et al., 2007).
However, all these findings were also often accompanied by
moderate-to-severe atherosclerosis.

59.9 WM DISEASE
Figure 59.4 (See colour insert.) Pathological features
associated with SVD in VaD. Panels show examples of WM hyperintensities on T2-weighted MRI or leukaraiosis
lacunes, small infarcts and microinfarcts: (a) typical cavi- as a decreased signal on CT is a neuroimaging construct
tated lacunar lesions (arrow) in the putamen of a 65-year- to describe diffuse and focal WM changes. Leukoariaosis
old man. (b) Multiple infarcts in the basal ganglias of an predominantly refers to vascular disease and not only
80-year-old man with vascular and neurofibrillary pathol-
incorporates WM rarefaction, incomplete infarction,
ogy. (c)–(e) Cerebral microvessels with variable hyalinosis,
perivascular rarefaction, microinfarcts and perivascular lacunar strokes, perivascular spacing and demyelination
spaces in three different cases. Moderate gliosis in the but sometimes also axonal and Wallerian degeneration
surrounding region is also evident in case in (c); (f) small (Figure 59.5). Both in areas of leukoaraiosis and zones
cortical infarct in a VaD case with severe CAA. A penetrat- outside the lesions, decreased vascular density is noticed
ing arteriole with CAA is seen in the middle of the infarct indicating that leukoaraiosis appears as a generalized fea-
(arrow). Magnification bar: (a) = 1 cm, (b), (f) = 500 μm; (c), ture of CVD rather than limited to deep WM (Brown et al.,
(d) = 100 μm, (e) = 200 μm. 2007). This is consistent with finding of an association with
unstable carotid plaques and the number of WM lesions,
grey matter and WM in VaD. They represent small foci of suggesting a thromboembolic role in some patients with
ischaemic necrosis resulting from narrowing or occlusion of leukoaraiosis (Altaf et al., 2006).
penetrating arteries branching directly from larger cerebral Neuroimaging and pathological studies demonstrate
arteries (Fisher, 1982). Lacunar infarcts are frequently multi- WM hyperintensisties and represent degeneration of the
ple and bilateral and often coexist with other vascular lesions WM, mostly explained by SVD (Pantoni and Garcia, 1997;
(e.g. large infarcts or diffuse WM damage). Whether single or Pantoni, 2010). Diffuse and focal WM lesions are a hall-
multiple, they may be asymptomatic, depending on their loca- mark of VaD (Ihara et al., 2010), but also occur mostly
tion and the volume of normal brain tissue lost. Lacunes may in ~30% of AD and in dementia with Lewy bodies (DLB)
also represent small haemorrhages or dilated perivascular cases (Englund, 1998). There is some controversy whether
spaces without infarction or haemorrhage. A few lacunes may deep or periventricular lesions are of more importance, but
represent healed or reabsorbed as minute or petechial haem- this depends on the definition of boundaries between the
orrhages. Microlacunes have also been described that are periventricular and deep WM if the coursing of the fibres is
essentially should be thought of as large cystic microinfarcts. used as markers (Kovari et al., 2007). Lacunar infarcts are
Aside from critical lesions occurring often in the internal produced when the ischaemic damage is focal and of suf-
capsule or caudate nucleus, recent meta-analyses suggested ficient severity to result in a small area of necrosis, whereas
The neuropathology of vascular dementia 649

(a) (b) et al., 2012). The projected misery perfusion due to capil-
lary loss or abnormalities occurring prior to leukoaraiosis
corroborates the finding of a chronic hypoxic state in the
deep WM (Fernando et al., 2006), which also releases sev-
eral growth promoting factors (Simpson et al., 2009). Some
of the WM damage in demented patients may simply reflect
Wallerian changes secondary to cortical loss of n ­ eurones.
However, this is unlikely since histological changes char-
acteristic of Wallerian degeneration are not evident as in
(c) (d) WM pallor. Conversely, in AD patients with severe loss of
cortical neurones, similar WM lesions are not apparent
(Englund, 1998).
While WM changes focus on the arterial system, nar-
rowing, in many cases, occlusion of veins and venules by
collagenous thickening of the vessel walls also occur. The
thickening of the walls of periventricular veins and venules
by collagen (collagenosis) increases with age and perivenous
collagenosis is increased further in brains with leukoara-
iosis (Brown et al., 2002b). The presence of apoptotic cells
in WM adjacent to areas of leukoaraiosis suggests that such
lesions are dynamic, with progressive cell loss and expan-
sion (Brown et al., 2002b). Vascular stenosis caused by col-
lagenosis may induce chronic ischaemia or oedema in the
deep WM leading to capillary loss and widespread effects
on the brain (Brown et al., 2007; Brown et al., 2009).
Figure 59.5 (See colour insert.) WM lesions visualized by
conventional histopathological staining in an 85-year-old
man diagnosed with vascular encephalopathy. (a) >75%
stenosis in the internal carotid artery 8 mm above the
bifurcation. The narrowed lumen (arrow) is seen. (b) WM 59.10 CEREBRAL MICROINFARCTS
rarefaction and myelin loss in the medial temporal lobe
but sparing of U fibres; (c), post-mortem T2W magnetic
resonance image of a formalin-fixed block from the parietal The accumulation of small, even miniscule ischaemic lesions
lobe. Area of hypersignal can be seen in the WM (*); as an important substrate of VaD has been emphasized in
(d), H&E stained section from the block in (c) showing recent years (Kalaria, 2012). Microinfarcts are widely accepted
severe deep WM ­pallor in the area of hypertensity (*). to be small lesions visible only upon microscopy (Figure 59.4;
A small cortical infract is also seen (arrow). Magnification
Table 59.2). These lesions of up to 5-mm diameter may or
bar: (a) = 500 mm, (b) = 400 μm, (c) = 1 cm, (d) = 500 μm.
may not involve a small vessel at their centre but are foci with
pallor, neuronal loss, axonal damage (WM) and gliosis. These
diffuse WM change is considered a form of rarefaction or are estimated to occur in thousands. Sometimes, these may
incomplete infarction where there may be selective damage include regions of incomplete infarction or rarefied (subacute)
to some cellular components. Although, the U-fibres are change. Microinfarcts have been described as attenuated
frequently spared, WM disease may comprise several pat- lesions of indistinct nature occurring in both cortical and
terns of alterations including pallor or swelling of myelin, subcortical regions. Such lesions or combinations are reported
loss of oligodendrocytes, axons and myelin fibres, cavita- when there are multiple or at least greater than three present
tions with or without presence of macrophages and areas in any region (Table 59.2). Microvascular infarcts (lacunar
of reactive astrogliosis (Simpson et al., 2007), where the infarcts and microinfarcts) appear central to the most
astrocytic cytoplasm and cell processes may be visible with common cause of VaD (Figure 59.4) and predict poor outcome
standard stains. in the elderly (Ballard et al., 2000; Vinters et al., 2000; Brown
Lesions in the WM also include spongiosis i.e. vacuoliza- et al., 2002a). In autopsied older Japanese-American men, the
tion of the WM structures and widening of the perivascu- importance of microvascular lesions as a likely explanation
lar spaces (Yamamoto et al., 2009). Affected regions do not for dementia was nearly equal to that of Alzheimer lesions
have sharp boundaries, in contrast to the plaques of multi- (White et al., 2002; White, 2009). Microinfarction in the
ple sclerosis. These changes may be associated with chronic subcortical structures has been emphasized as a substrate of
pro-thrombotic endothelial dysfunction in cerebral SVD cognitive impairment (Arvanitakis et al., 2011a; Kalaria, 2012)
(Hassan et al., 2003) also involving the WM (Brown and and correlated with increased Alzheimer type pathology, but
Thore, 2011). However, there may be a cerebral response to cortical microinfarcts also appear to contribute to the progres-
the SVD by increasing endothelial thrombomodulin (Giwa sion of cognitive deficits in brain ageing (Kovari et al., 2004).
650 Dementia

In addition to microinfarction in the subcortical structures, arteriolosclerosis and perivascular spacing and with
it appears increasingly important that multiple cortical areas lacunes in any brain region, but not large vessel disease
of microinfarction are associated with subcortical VaD or whole brain measures of neurodegenerative pathology.
or SVD (Figure 59.4). Thus, these lesions should be taken Higher levels of putamen haemosiderin were correlated
into account when defining the neuropathological criteria. with more microbleeds upon MRI, but it is possible that
Cortical microinfarcts increase in the presence of cerebral brain iron homeostasis and small vessel ischaemic change
amyloid angiopathy (CAA) (Okamoto et al., 2012). In a recent are responsible for these rather than only as a marker for
study, cortical microinfarcts were frequently detected in AD minor episodes of cerebrovascular extravasation.
and associated with CAA, but rarely observed in subcortical
VaD linked to SVD (Suter et al., 2002; Okamoto et al.,
2009). Microinfarcts in the cerebral cortex associated with
59.12 HIPPOCAMPAL ATROPHY AND
severe CAA may be the primary pathological substrate in a
significant proportion to VaD cases (Haglund et al., 2006).
SCLEROSIS
Cortical microinfarcts and to lesser extent periventricular
demyelination were significantly associated with cognitive Neuroimaging studies show that medial temporal lobe and
decline in individuals at high risk of dementia (Gold et al., hippocampal atrophy are associated with VaD (Bastos-Leite
2007). It is proposed that the changes in hemodynamics, e.g. et al., 2007; Firbank et al., 2011) and SVD (O’Sullivan et al.,
hypotension and atherosclerosis may play a role in the genesis 2009; van de Pol et al., 2011) albeit, not to the same extent
of cortical watershed microinfarcts. as in AD (Burton et al., 2009). Pathological evidence shows
that ischaemic VaD and SVD are also associated with hip-
pocampal changes and atrophy remote to ischaemic injury
(Zarow et al., 2005; Matthews et al., 2009). Hippocampal
59.11 CEREBRAL MICROBLEEDS neurones in the Sommer’s sector are highly vulnerable to
disturbances in the cerebral circulation or hypoxia caused
Cerebral microbleeds (CMB) detected by MRI are small, by systemic vascular disease. The focal loss of CA1 neurones
dot-like hypotense abnormalities and have been associated in ischemic VaD has been related to lower hippocampal vol-
with extravasated haemosiderin derived from erythrocytes, ume and memory score (Zarow et al., 2005), but the degree
lipohyalinosis and CAA (Fazekas et al., 1999). They are likely of loss appears to be less in VaD (Kril et al., 2002) than in
to be a surrogate marker of SVD evident on MRI along with AD. However, selective hippocampal neuronal shrinkage is
lacunes and WM changes (Van der Flier and Cordonnier, also an important substrate for VaD. This is also evident in
2012). The prevalence of radiological microbleeds in delayed dementia after stroke in the absence of any neuro-
VaD ranges 35%–85%. Microbleeds mainly result from degenerative pathology (Gemmell et al., 2012). Thus, there
hypertensive vasculopathy, but the frequent co-occurrence of is a clear vascular basis for hippocampal neurodegeneration
lobar microbleeds suggests that neurodegenerative pathology and concurs with the neuroimaging observations of hip-
or CAA is also of importance (Werring et al., 2011). The rel- pocampal atrophy even in population-based incident VaD
evance of this radiological construct is increasingly being (Scher et al., 2011). The simplest mechanistic explanation for
recognized due to their relation to clinical outcome and the atrophy is that the neuronal or dendritic arbour results
occurrence in anti-amyloid vaccination trials (Greenberg in subsequent loss in connectivity, which contributes to
et al., 2009). However, the presence of multiple microbleeds brain’s structural and functional changes. This is consistent
in the context of VaD is related to worse performance on with the finding that soluble synaptophysin was decreased
cognitive tests, mainly in psychomotor speed and executive in VaD as well as AD.
functioning. Since microbleeds are common in cogni- Hippocampal sclerosis is likely a major contributing
tively normal older individuals, attribution of these to VaD factor in the hippocampal atrophy and occurs in approx-
should follow a careful exclusion of other causes of cognitive imately 10% of individuals over the age of 85 and slightly
impairment and only if numerous such lesions are present. higher in VaD. It is characterized by severe cell loss with
Both radiological cerebral microbleeds and foci of the CA fields in the presence or absence of microinfarc-
haemosiderin containing single crystalloids or larger tion and gliosis that is not explained by AD. TAR DNA
perivascular aggregates are found in brains of older protein 43 immunohistochemistry can be used to dem-
subjects including those diagnosed with VaD and AD, but onstrate that hippocampal sclerosis, regardless of accom-
the radiological and pathological relationship between panying pathologies (e.g. AD or VaD), is consistent with
these findings has not been entirely clear. Recent evidence an underlying neurodegenerative pathogenetic mecha-
suggests that cerebral microbleeds detected by MRI are nism (Zarow et al., 2012). Any focal loss or patterns of
a surrogate for ischaemic SVD rather than exclusively hippocampal sclerosis can be graded (Rauramaa et al.,
haemorrhagic diathesis (Janaway et al., 2013). Greater 2013) and recorded together with any microinfarctions.
putamen haemosiderin was significantly associated with Sometimes, the patchy neurone loss and gliosis in some
indices of small vessel ischaemia including microinfarcts, brains with AD pathology may be difficult or impossible
The neuropathology of vascular dementia 651

to distinguish from anoxic-ischaemic change. The aeti- (Brun, 1994; Ferrer et al., 2008) that may fall into the sub-
ology of hippocampal sclerosis is defined in association type IV of VaD pathology.
with neurodegenerative process, a pathologic condition
presumed to arise from hypoxic/ischaemic mechanisms
(Rauramaa et al., 2013). Hippocampal sclerosis pathol-
59.15 SPORADIC CEREBRAL AMYLOID
ogy can be associated with different underlying causes
but a large study (Nelson et al., 2011) found no evidence
ANGIOPATHY IN VAD
for associations between hippocampal sclerosis and lacu-
nar infarcts, large infarcts, circle of Willis atherosclerosis CAA is most common in AD but it often occurs in CVD
or CAA. However, there was a correlation between in the general absence of Alzheimer pathology (Cohen
hippocampal sclerosis and arteriolosclerosis in multiple et al., 1997). In sporadic cases, CAA alone is considered
brain regions outside of the ­h ippocampus including the a substrate for cognitive impairment (Pfeifer et al., 2002;
frontal cortex (Brodmann area 9) (Nelson et al., 2011). Greenberg et al., 2004; Arvanitakis et al., 2011b). Tissue
This is ascribed to a pathogenetic change in aged human microstructural damage caused by CAA prior to pre-
brain arterioles that affects multiple brain areas and intracerebral haemorrhage is independently associated
contributes to hippocampal sclerosis of ageing (Neltner with cognitive impairment (Viswanathan et al., 2008). The
et al., 2014). prevalence of CAA in VaD is not known, but it is a major
cause of intracerebral and lobar haemorrhages leading to
profound ischaemic damage (Reijmer et al., 2015). Several
familial forms of CAA involving ischaemic and haemor-
59.13 BORDER ZONE (WATERSHED) rhagic infarcts (see below) and cerebral hypoperfusion
INFARCTS demonstrate the link between CAA and VaD. In a study of
surgical biopsies exhibiting cerebral and cerebellar infarc-
The border zone or watershed infarctions mostly occur tion, CAA was significantly more common in samples
from haemodynamic events, usually in patients with showing infarction than in age-matched controls with
severe internal carotid artery stenosis. They could occur non-vascular lesions (Cadavid et al., 2000). There is also
bilaterally or unilaterally and are disposed to regions an association between severe CAA and cerebrovascular
between two main arterial territories, deep and superficial lesions coexisting with AD, including lacunar infarcts,
vessel systems. Typical border zone infarctions may be 5 microinfarcts and haemorrhages (Olichney et al., 1995;
mm or wider as wedge-shaped regions of pallor and rar- Ellis et al., 1996; Olichney et al., 1997, 2000a; Okamoto et
efaction extending into the WM. Larger areas of incom- al., 2012). This association apparently is not attributable to
plete infarction may extend into the WM (Ihara et al., apolipoprotein E (APOE) ε4 allele, as the vascular lesions
2010). These are characterized by mild-to-moderate loss of correlated best with severity of CAA, regardless of geno-
oligodendrocytes, myelin and axons in areas where there type (Olichney et al., 2000b). There is some evidence to
may be hylainzed vessels (Brun, 1994). This may be accom- suggest CAA is related to WM changes but not exclusively
panied by astrogliosis, some microgliosis and macrophage in the oldest (Tanskanen et al., 2013). It appears that the
infiltration. The morphology of incomplete or subinfarc- first stroke-like episode triggers multiple cerebral bleeds,
tive changes, though, suspected to be associated with which is preceded by diffuse WM changes that, in turn,
cognitive function, is not consistently described in VaD. lead to rapid decline of cognitive functions.
It may variably manifest as tissue rarefaction assessed by
conventional stains and revealed as injury response, such
as microgliosis and astrocytosis, or the presence of other
59.16 NEUROCHEMICAL PATHOLOGY
‘reactive’ cells or surrogate markers of dendritic, synaptic
or axonal damage.
OF VAD

The neurochemical basis of cognitive decline in CVD is


poorly understood. There are few concerted studies on the
59.14 LAMINAR NECROSIS protein and lipid chemistry of VaD. Various cellular sig-
nalling and regulatory mechanisms including apoptosis,
Laminar necrosis is characterized by neuronal ischaemic autophagy, oxidative stress and inflammation are associ-
changes leading to neuronal loss and gliosis in the cortical ated with VaD by virtue of their involvement in cerebral
ribbon. This is particularly apparent in cases where global ischaemia or oligaemia. VaD subjects also mount to a selec-
ischaemia or hypoperfusion has occurred as in the case of tively attenuated neuroinflammatory response (Mulugeta
cardiac arrest. Typical topographic distribution of spongi- et al., 2008). It was reported that the monocyte chemoat-
form change can be readily apparent with standard stains. tractant protein-1 and interleukin-6 concentrations were
They appear more commonly at the arterial border zones significantly reduced in the frontal lobe of VaD and mixed
652 Dementia

dementia subjects suggesting that these changes had a vas- (Toledo et al., 2013). The burden of mixed pathology is
cular basis rather than due to Alzheimer pathology. even greater in elderly people within the community at
The perivascular nerve plexus (Hamel, 2006) is highly large (Neuropathology Group of the Medical Research
vulnerable, yet only few transmitter specific changes Council Cognitive Function and Ageing Study [MRC
reflecting neurovascular pathology have been described CFAS], 2001; Riekse et al., 2004; Sonnen et al., 2007). In
in subtypes of VaD. Selective transmitter-specific changes one community-based sample, 38% of dementia cases had
have been described in some cases of VaD (Kalaria and mixed pathology, with both Alzheimer-type changes and
Ballard, 1999).Two different groups had previously shown vascular lesions but ‘pure AD’ represented only 21%–24% of
that compared to AD patients, choline acetyltransferase cases (Schneider et al., 2007). WM lesions indicating isch-
activity was also reduced albeit to a lesser degree in the aemic or oligaemic aetiology are also high in community-
­temporal cortex and hippocampus in patients diagnosed dwelling subjects by as much as 94% and this change is
with multi-infarct dementia or VaD (Perry et al., 1977; an independent substrate for dementia (Fernando et al.,
Gottfries et al., 1994). Furthermore, choline acetyltrans- 2004). In addition, atherosclerosis in cerebral arteries and
ferase activities were significantly reduced by 60%– the circle of Willis (Roher et al., 2005; Yarchoan et al., 2012)
70% in frontal and temporal cortices of subjects with is frequently present in AD. The commonest overlapping
Cerebral Autosomal Dominant Arteriopathy with Sub- pathologies involve smaller cerebrovascular lesions rather
cortical Infarcts and Leukoencephalopathy (CADASIL), than large infarcts (Deramecourt et al., 2012) (Figure
which models SVD (Keverne et al., 2007). Choline 59.1). These include most features of SVD such as corti-
acetyltransferase and p75 (neurotrophin receptor) immu- cal infarcts, lacunes, diffuse and periventricular myelin
noreactivities were also affected within the cholinergic loss, WM microvacuolation, microinfarcts, microhaem-
cell bodies of the basal forebrain in CADASIL, but these orrhages, arteriolosclerosis and focal and diffuse gliosis
were not so pronounced. This may depend on severity of (Esiri et al., 1997; Ballard et al., 2000; Vinters et al., 2000).
the WM degeneration (Mann et al., 1986; Keverne et al., AD pathology was three times greater in VaD cases with
2007). However, loss of cholinergic function is consis- small (<15 mL) compared to large infarcts (Ballard et al.,
tently greater in VaD patients with concurrent Alzheimer 2000). These findings also corroborate the importance of
pathology (Román and Kalaria, 2006). Conversely, a novel microvascular disease rather than large vessel disease as
increase in cholinergic activity in the frontal cortex was the critical substrate in VaD and in AD.
revealed in infarct dementia (Sharp et al., 2009). Clinicopathological studies also suggest that vascular
Other studies have reported deficits in monoamines disease influences the burden of the neurodegenerative
including dopamine and 5-hydroxytryptamine (5HT) in lesion (Zekry et al., 2002; Riekse et al., 2004). The den-
the basal ganglia and neocortex in VaD (Gottfries et al., sity of neocortical plaques was lower in AD cases with
1994). To compensate for the loss (Elliott et al., 2009), coexistent vascular lesions interpreted as contributing to
5-HT(1A) and 5-HT(2A) receptors are likely to be increased the dementia (Nagy et al., 1997) as well. In the Religious
in the temporal cortex in multi-infarct but not subcortical Order study involving elderly nuns who exhibited coexis-
VaD. Such findings, albeit fragmentary, reveal distinctions tent AD and brain infarcts at autopsy had poorer cogni-
between the neurochemical pathology of VaD subtypes and tive function and a higher prevalence of dementia than
suggest possibilities of pharmacological manipulation with did those without vascular change (Snowdon et al., 1997).
novel therapies in VaD. There was also loss of glutamater- Compared to pure AD, lower burden of Alzheimer-type
gic synapses, assessed by vesicular glutamate transporter 1 pathology particularly fewer neurofibrillary tangles was
concentrations, in the temporal cortex of VaD (Kirvell et al., required to reach the threshold for dementia when there
2011), but preservation of these in the frontal lobe suggests a were concomitant lacunar infarcts in subcortical struc-
role in sustaining cognition and protecting against demen- tures including the basal ganglia, thalamus or deep WM.
tia following a stroke. Identification of the morphological Similarly, in another religious order study, after account-
equivalents of these changes in types of pyramidal cells in ing for AD lesion burden, the presence of other pathol-
the frontal lobe would be relevant. ogies or infarcts increased the odds of dementia over
fivefold (Schneider et al., 2007) and caused earlier onset of
dementia (Esiri et al., 1999).
59.17 INTERACTION BETWEEN
VASCULAR AND ALZHEIMER-
TYPE PATHOLOGIES 59.18 PATHOLOGICAL FEATURES OF
FAMILIAL CAA AND SVDS AS
Concurrent cerebrovascular disease is a common neuro-
MODELS OF VAD
pathological finding in aged subjects with dementia and
more common in AD than in other neurodegenerative dis- There are more than 10 different hereditary cerebral amy-
orders, especially in younger subjects and lowers the thresh- loid angiopathies caused by mutations in different genes
old for dementia due to AD and alpha-synucleinopathies (Yamamoto et al., 2011). All these angiopathies lead to
The neuropathology of vascular dementia 653

some degree of cognitive impairment or dementia. They are affected subjects exhibit not only severe arteriolosclerosis,
characterized by multiple haemorrhages and haemorrhagic leukoencephalopathy and lacunar infarcts but also spi-
or ischaemic infarcts in addition to severe amyloid deposi- nal anomalies and alopecia. Strokes lead to stepwise dete-
tion within walls of the meningeal and intracerebral ves- rioration with most subjects becoming demented in older
sels. In hereditary cerebral haemorrhage with amyloidosis age. Familial cerebral SVDs, involving progressive visual
of the Dutch type, dementia occurs in most patients sur- impairment (Kalimo and Kalaria, 2005), cause deteriora-
viving their initial stroke (Haan et al., 1990) and may occa- tion in cognitive function. Hereditary endotheliopathy with
sionally be the presenting feature (Wattendorff et al., 1982; retinopathy, nephropathy and stroke (HERNS), cerebro-
Haan et al., 1989). The extensive CAA alone is sufficient to retinal vasculopathy (CRV) and hereditary vascular reti-
cause dementia and this has implications for CAA related nopathy (HVR) were reported independently, but represent
cognitive dysfunction in sporadic CAA and AD (Natte different phenotypes in the same disease spectrum (Jen
et al., 2001). In the Icelandic type of hereditary cerebral et al., 1997; Terwindt et al., 1998; Ophoff et al., 2001). These,
haemorrhage with amyloidosis (HCHWA-I), which is now described as autosomal dominant retinal vasculopathy
associated with a point mutation in the gene encoding the with cerebral leukodystrophy lead to early death and cause
cysteine protease inhibitor Cystatin C (Levy et al., 1989), dementia. Retinal microvessels undergo severe distortions
dementia, occurring in some patients, has been attributed and become tortuous predictive of the SVD type of pathol-
to the multiple vascular lesions. Individuals with gelsolin- ogy with multilaminated vascular basement membranes in
related amyloidosis manifest facial palsy, mild peripheral the brain (Jen et al., 1997).
neuropathy and corneal lattice dystrophy; atrophic bulbar Some rarer and less characterized hereditary SVDs also
palsy, gait ataxia and mild cognitive impairment (Kiuru exist that are associated with clinical SVD features and dif-
et al., 1999). Familial British dementia with severe amy- ferent degrees of cognitive impairment but the pathologies
loid angiopathy (Vidal et al., 1999) is characterized by in these have not been described. They include conditions
dementia, progressive spastic tetraparesis and cerebellar with abnormalities in the skin and eye and multiple lacunar
ataxia, the onset is usually in the sixth decade (Mead et al., infarcts in deep WM and pons (Pavlovic et al., 2005), reti-
2000). Neuropathological features also include Alzheimer- nal arteriolar tortuosity and leukoencephalopathy (Vahedi
type neurofibrillary tangles and neuropil threads in et al., 2003; Gould et al., 2006) and profound WM changes
the anteromedial temporal lobe that may contribute to upon MRI (Verreault et al., 2006).
dementia (Plant et al., 1990; Revesz et al., 2009). Familial
Danish dementia (FDD), also known as heredopathia
ophthalmooto-encephalica (HOOE), is another condition
with severe and widespread CAA (Vidal et al., 2000). FDD 59.19 CONCLUSIONS
is clinically characterized by cataracts, deafness, progres-
sive ataxia and dementia. Defining the neuropathological substrates of VaD relies on
Several familial stroke disorders also appear to cause uniformity in sampling and pathological examination. VaD
cognitive impairment or dementia. A common feature in resulting from severe VCI or vascular cognitive disorder or
these is subcortical SVD often characterized by severe arte- from delayed impairment after stroke appears to result from
riolosclerosis in the perforating vessels (Yamamoto et al., the accumulation of several lesions including cerebral atro-
2011). CADASIL is the most common form of hereditary phy. While robust neuropathological criteria for VaD are
SVDs (Chabriat et al., 2009). Motor deficits, an ataxic hemi- still being developed, multiple microinfarcts, small infarcts
paresis, hemianopsia and dysarthria may be present as key or lacunes in the subcortical structures, rather than mac-
neurological features akin to SVD. Vascular changes includ- roinfarction or large vessel disease appear robustly related
ing apoptotic loss of brain vascular smooth muscle cells to cognitive impairment. Diffuse WM changes involving
(Gray et al., 2007) and vessel wall thickening (Craggs et al., periventricular and deeper regions are frequent in VaD.
2013) are likely to reduce blood flow and affect the vasodila- Concomitant hippocampal pathology including sclerosis
tory response to cause lacunar infarcts and microinfarcts in and Alzheimer pathology compound disease progression.
grey matter and WM (Yamamoto et al., 2011). The exten- Further definitions of the ­­neuropathological correlates of
sive demyelination and axonal damage in the underlying VaD and investigation of genetic models would be ­valuable
WM contributes to cortical atrophy (Craggs et al., 2013) and for exploring the pathogenesis as well as management of
impacts on frontal lobe cognitive functions that is consis- VaD through preventative and treatment strategies in
tent with the disconnection of the fronto-striatal circuits in the future.
CADASIL. Neuronal apoptosis, predominantly in neocorti-
cal layers III and V, is also likely to contribute to dementia REFERENCES
in CADASIL (Gray et al., 2007).
The Maeda syndrome or cerebral autosomal recessive Akatsu, H., Takahashi, M., Matsukawa, N. et al. (2002).
arteriopathy with subcortical infarcts and leukoencepha- Subtype analysis of neuropathologically diagnosed
lopathy (CARASIL) is an autosomal recessive disorder patients in a Japanese geriatric hospital. Journal of the
similar to CADASIL (Hara et al., 2009). The normotensive Neurological Sciences, 196 (1–2): 63–69.
654 Dementia

Allan, L.M., Rowan, E.N., Firbank, M.J. et al. (2012). Bowler, J.V. and Hachinski, V. (1995). Vascular cognitive
Long term incidence of dementia, predictors of impairment: A new approach to vascular dementia.
mortality and pathological diagnosis in older stroke Baillieres Clinical Neurology, 4 (2): 357–376.
survivors. Brain: A Journal of Neurology, 134 (Pt 12): Brown, W.R., Moody, D.M., Challa, V.R. et al. (2002a).
3716–3727. Apoptosis in leukoaraiosis lesions. Journal of the
Altaf, N., Daniels, L., Morgan, P.S. et al. (2006). Cerebral Neurological Sciences, 203–204: 169–171.
white matter hyperintense lesions are associated with Brown, W.R., Moody, D.M., Challa, V.R. et al. (2002b).
unstable carotid plaques. European Journal of Vascular Venous collagenosis and arteriolar tortuosity in leu-
and Endovascular Surgery, 31 (1): 8–13. koaraiosis. Journal of the Neurological Sciences, 203:
Arvanitakis, Z., Leurgans, S.E., Barnes, L.L. et al. (2011a). 159–163.
Microinfarct pathology, dementia, and cognitive sys- Brown, W.R., Moody, D.M., Thore, C.R. et al. (2009).
tems. Stroke: A Journal of Cerebral Circulation, 42 (3): Microvascular changes in the white matter in demen-
722–727. tia. Journal of the Neurological Sciences, 283 (1–2):
Arvanitakis, Z., Leurgans, S.E., Wang, Z. et al. (2011b). 28–31.
Cerebral amyloid angiopathy pathology and cognitive Brown, W.R., Moody, D.M., Thore, C.R. et al. (2007).
domains in older persons. Annals of Neurology, 69 (2): Vascular dementia in leukoaraiosis may be a conse-
320–327. quence of capillary loss not only in the lesions, but
American Psychiatric Association (2013). Diagnostic in normal-appearing white matter and cortex as
and Statistical Manual of Mental Disorders, Vol. 5. well. Journal of the Neurological Sciences, 257 (1–2):
Arlington, VA: American Psychiatric Association. 62–66.
Bailey, E.L., Smith, C., Sudlow, C.L. and Wardlaw, J.M. Brown, W.R. and Thore, C.R. (2011). Review: Cerebral
(2012). Pathology of lacunar ischemic stroke in microvascular pathology in ageing and neurodegen-
humans – A systematic review. Brain Pathology, 22 (5): eration. Neuropathology and Applied Neurobiology,
583–591. 37 (1): 56–74.
Ballard, C., McKeith, I., O’Brien, J. et al. (2000). Brun, A. (1994). Pathology and pathophysiology of cere-
Neuropathological substrates of dementia and brovascular dementia: Pure subgroups of obstruc-
depression in vascular dementia, with a particular tive and hypoperfusive etiology. Dementia, 5 (3–4):
focus on cases with small infarct volumes. Dementia 145–147.
and Geriatric Cognitive Disorders, 11 (2): 59–65. Burton, E.J., Barber, R., Mukaetova-Ladinska, E.B. et al.
Barker, W.W., Luis, C.A., Kashuba, A. et al. (2002). (2009). Medial temporal lobe atrophy on MRI dif-
Relative frequencies of Alzheimer disease, Lewy ferentiates Alzheimer’s disease from dementia with
body, vascular and frontotemporal dementia, and Lewy bodies and vascular cognitive impairment:
hippocampal sclerosis in the State of Florida Brain A prospective study with pathological verification of
Bank. Alzheimer Disease and Associated Disorders, diagnosis. Brain: A Journal of Neurology, 132 (Pt 1):
16 (4): 203–212. 195–203.
Bastos-Leite, A.J., van der Flier, W.M., van Straaten, E.C. Cadavid, D., Mena, H., Koeller, K. and Frommelt, R.A.
et al. (2007). The contribution of medial temporal (2000). Cerebral beta amyloid angiopathy is a
lobe atrophy and vascular pathology to cognitive risk factor for cerebral ischemic infarction. A case
impairment in vascular dementia. Stroke: A Journal of control study in human brain biopsies. Journal of
Cerebral Circulation, 38 (12): 3182–3185. Neuropathology and Experimental Neurology, 59 (9):
Bejot, Y., Aboa-Eboule, C., Durier, J. et al. (2011). 768–773.
Prevalence of early dementia after first-ever stroke: Censori, B., Manara, O., Agostinis, C. et al. (1996).
A 24-year population-based study. Stroke: A Journal Dementia after first stroke. Stroke: A Journal of
of Cerebral Circulation, 42 (3): 607–612. Cerebral Circulation, 27 (7): 1205–1210.
Bennett, D.A., Wilson, R.S., Gilley, D.W. and Fox, J.H. Chabriat, H., Joutel, A., Dichgans, M. et al. (2009).
(1990). Clinical diagnosis of Binswanger’s disease. CADASIL. Lancet Neurology, 8 (7): 643–653.
Journal of Neurology, Neurosurgery and Psychiatry, 53 Cohen, D.L., Hedera, P., Premkumar, D.R. et al. (1997).
(11): 961–965. Amyloid-beta protein angiopathies masquerading as
Berrios, G.E. and Freeman, H.L. (1991). Alzheimer and Alzheimer’s disease? Annals of the New York Academy
the dementiaa. In Berrios, G.E. (ed.), Eponymists in of Sciences, 826: 390–395.
Medicine Series. London, United Kingdom: Royal Craggs, L.J., Yamamoto, Y., Ihara, M. et al. (2013). White
Society of Medicine Services, pp. 69–76. matter pathology and disconnection in the fron-
Blessed, G., Tomlinson, B.E. and Roth, M. (1968). The tal lobe in CADASIL. Neuropathology and Applied
association between quantitative measures of demen- Neurobiology, 40 (5): 591–602.
tia and of senile change in the cerebral grey matter of del Ser, T., Bermejo, F., Portera, A. et al. (1990). Vascular
elderly subjects. The British Journal of Psychiatry, 114 dementia. A clinicopathological study. Journal of the
(512): 797–811. Neurological Sciences, 96 (1): 1–17.
The neuropathology of vascular dementia 655

Deramecourt, V., Slade, J.Y., Oakley, A.E. et al. (2012). Ferrer, I., Kaste, M. and Kalimo, H. (2008). Vascular
Staging and natural history of cerebrovascular pathol- diseases. In Love, S., Louis, D. and Ellison, D. (eds.),
ogy in dementia. Neurology, 78 (14): 1043–1050. Greenfield’s Neuropathology, Eighth Edition.
Desmond, D.W., Moroney, J.T., Paik, M.C. et al. (2000). Oxford, United Kingdom: Oxford University Press,
Frequency and clinical determinants of dementia after pp. 121–240.
ischemic stroke. Neurology, 54 (5): 1124–1131. Firbank, M.J., He, J., Blamire, A.M. et al. (2011). Cerebral
Elliott, M.S., Ballard, C.G., Kalaria, R.N. et al. (2009). blood flow by arterial spin labeling in poststroke
Increased binding to 5-HT1A and 5-HT2A receptors dementia. Neurology, 76 (17): 1478–1484.
is associated with large vessel infarction and rela- Fisher, C.M. (1982). Lacunar strokes and infarcts: A review.
tive preservation of cognition. Brain: A Journal of Neurology, 32 (8): 871–876.
Neurology, 132 (Pt 7): 1858–1865. Gemmell, E., Bosomworth, H., Allan, L. et al. (2012).
Ellis, R.J., Olichney, J.M., Thal, L.J. et al. (1996). Cerebral Hippocampal neuronal atrophy and cognitive func-
amyloid angiopathy in the brains of patients with tion in delayed poststroke and aging-related demen-
Alzheimer’s disease: The CERAD experience, Part XV. tias. Stroke: A Journal of Cerebral Circulation, 43 (3):
Neurology, 46 (6): 1592–1596. 808–814.
Englund, E. (1998). Neuropathology of white matter Giannakopoulos, P., Gold, G., Kovari, E. et al. (2007).
changes in Alzheimer’s disease and vascular dementia. Assessing the cognitive impact of Alzheimer disease
Dementia and Geriatric Cognitive Disorders, 9 (Suppl. pathology and vascular burden in the aging brain: The
1): 6–12. Geneva experience. Acta Neuropathologica, 113 (1):
Erkinjuntti, T., Haltia, M., Palo, J. et al. (1988). Accuracy of 1–12.
the clinical diagnosis of vascular dementia: A prospec- Giwa, M.O., Williams, J., Elderfield, K. et al. (2012).
tive clinical and post-mortem neuropathological study. Neuropathologic evidence of endothelial changes in
Journal of Neurology, Neurosurgery and Psychiatry, 51 cerebral small vessel disease. Neurology, 78 (3): 167–174.
(8): 1037–1044. Gold, G., Giannakopoulos, P., Herrmann, F.R. et al.
Erkinjuntti, T., Inzitari, D., Pantoni, L. et al. (2000). (2007). Identification of Alzheimer and vascular lesion
Limitations of clinical criteria for the diagnosis of thresholds for mixed dementia. Brain: A Journal of
vascular dementia in clinical trials. Is a focus on subcor- Neurology, 130 (Pt 11): 2830–2836.
tical vascular dementia a solution? Annals of the New Gorelick, P.B., Chatterjee, A., Patel, D. et al. (1992). Cranial
York Academy of Sciences, 903: 262–272. computed tomographic observations in multi-infarct
Esiri, M.M., Nagy, Z., Smith, M.Z. et al. (1999). dementia. A controlled study. Stroke: A Journal of
Cerebrovascular disease and threshold for dementia Cerebral Circulation, 23 (6): 804–811.
in the early stages of Alzheimer’s disease. Lancet, 354 Gorelick, P.B., Scuteri, A., Black, S.E. et al. (2011). Vascular
(9182): 919–920. contributions to cognitive impairment and demen-
Esiri, M.M., Wilcock, G.K. and Morris, J.H. (1997). tia: A statement for healthcare professionals from
Neuropathological assessment of the lesions of sig- the American Heart Association/American Stroke
nificance in vascular dementia. Journal of Neurology, Association. Stroke: A Journal of Cerebral Circulation,
Neurosurgery and Psychiatry, 63 (6): 749–753. 42 (9): 2672–2713.
Fazekas, F., Kleinert, R., Roob, G. et al. (1999). Gottfries, C.G., Blennow, K., Karlsson, I. and Wallin, A.
Histopathologic analysis of foci of signal loss on (1994). The neurochemistry of vascular dementia.
gradient-echo T2*-weighted MR images in patients Dementia, 5 (3–4): 163–167.
with spontaneous intracerebral hemorrhage: Evidence Gould, D.B., Phalan, F.C., van Mil, S.E. et al. (2006). Role
of microangiopathy-related microbleeds. American of COL4A1 in small-vessel disease and hemorrhagic
Journal of Neuroradiology, 20 (4): 637–642. stroke. New England Journal of Medicine, 354 (14):
Fernando, M.S., Ince, P.G. and MRC Cognitive Function 1489–1496.
and Ageing Neuropathology Study Group (2004). Gray, F., Polivka, M., Viswanathan, A. et al. (2007).
Vascular pathologies and cognition in a population- Apoptosis in cerebral autosomal-dominant arteriopa-
based cohort of elderly people. Journal of the thy with subcortical infarcts and leukoencephalopa-
Neurological Sciences, 226 (1–2): 13–17. thy. Journal of Neuropathology and Experimental
Fernando, M.S., Simpson, J.E., Matthews, F. et al. (2006). Neurology, 66 (7): 597–607.
White matter lesions in an unselected cohort of the Greenberg, S.M., Gurol, M.E., Rosand, J. and Smith, E.E.
elderly: Molecular pathology suggests origin from (2004). Amyloid angiopathy-related vascular cognitive
chronic hypoperfusion injury. Stroke: A Journal of impairment. Stroke: A Journal of Cerebral Circulation,
Cerebral Circulation, 37 (6): 1391–1398. 35 (11 Suppl. 1): 2616–2619.
Ferrer, I. (2010). Cognitive impairment of vascular origin: Greenberg, S.M., Nandigam, R.N., Delgado, P. et al.
Neuropathology of cognitive impairment of vascular (2009). Microbleeds versus macrobleeds: Evidence
origin. Journal of the Neurological Sciences, 299 (1–2): for distinct entities. Stroke: A Journal of Cerebral
139–149. Circulation, 40 (7): 2382–2386.
656 Dementia

Haan, J., Lanser, J.B., Zijderveld, I. et al. (1990). Kalaria, R.N. (2012). Cerebrovascular disease and mecha-
Dementia in hereditary cerebral hemorrhage with nisms of cognitive impairment: Evidence from clinico-
amyloidosis-Dutch type. Archives of Neurology, pathological studies in humans. Stroke: A Journal of
47 (9): 965–967. Cerebral Circulation, 43 (9): 2526–2534.
Haan, J., Roos, R.A., Briet, P.E. et al. (1989). Hereditary Kalaria, R.N. and Ballard, C. (1999). Overlap between
cerebral hemorrhage with amyloidosis – Dutch pathology of Alzheimer disease and vascular demen-
type. Research-Group hereditary cerebral amyloid-­ tia. Alzheimer Disease and Associated Disorders, 13
angiopathy. Clinical Neurology and Neurosurgery, 91 (Suppl. 3): S115–S123.
(4): 285–290. Kalaria, R.N., Kenny, R.A., Ballard, C.G. et al. (2004).
Hachinski, V., Iadecola, C., Petersen, R.C. et al. (2006). Towards defining the neuropathological substrates
National Institute of Neurological Disorders and of vascular dementia. Journal of the Neurological
Stroke–Canadian Stroke Network vascular cognitive Sciences, 226 (1–2): 75–80.
impairment harmonization standards. Stroke: A Journal Kalaria, R.N., Perry, R.H., O’Brien, J. and Jaros, E. (2012).
of Cerebral Circulation, 37 (9): 2220–2241. Atheromatous disease in small intracerebral vessels,
Hachinski, V.C., Lassen, N.A. and Marshall, J. (1974). Multi- microinfarcts and dementia. Neuropathology and
infarct dementia. A cause of mental deterioration in Applied Neurobiology, 38 (5): 505–508.
the elderly. Lancet, 2 (7874): 207–210. Kalimo, H. and Kalaria, R.N. (2005). Hereditary forms of
Haglund, M., Passant, U., Sjobeck, M. et al. (2006). vascular dementia. In Kalimo, H. (ed.), Cerebrovascular
Cerebral amyloid angiopathy and cortical microin- Diseases, Pathology and Genetics. Basel, Switzerland:
farcts as putative substrates of vascular dementia. ISN Neuropath Press, pp. 324–384.
International Journal of Geriatric Psychiatry, 21 (7): Kavanagh, D., Spitzer, D., Kothari, P.H. et al. (2008). New
681–687. roles for the major human 3'-5’ exonuclease TREX1 in
Hamel, E. (2006). Perivascular nerves and the regulation of human disease. Cell Cycle, 7 (12): 1718–1725.
cerebrovascular tone. Journal of Applied Physiology, Keverne, J.S., Low, W.C., Ziabreva, I. et al. (2007).
100 (3): 1059–1064. Cholinergic neuronal deficits in CADASIL. Stroke:
Hara, K., Shiga, A., Fukutake, T. et al. (2009). Association A Journal of Cerebral Circulation, 38 (1): 188–191.
of HTRA1 mutations and familial ischemic cere- Kirvell, S.L., Elliott, M.S., Kalaria, R.N. et al. (2011).
bral small-vessel disease. New England Journal of Vesicular glutamate transporter and cognition in
Medicine, 360 (17): 1729–1739. stroke: A case-control autopsy study. Neurology, 75
Hassan, A., Hunt, B.J., O’Sullivan, M. et al. (2003). Markers (20): 1803–1809.
of endothelial dysfunction in lacunar infarction and Kiuru, S., Salonen, O. and Haltia, M. (1999). Gelsolin-
ischaemic leukoaraiosis. Brain: A Journal of Neurology, related spinal and cerebral amyloid angiopathy. Annals
126 (Pt 2): 424–432. of Neurology, 45 (3): 305–311.
Ho, K.L. and Garcia, J.H. (2000). Neuropathology of Knopman, D.S., Parisi, J.E., Boeve, B.F. et al. (2003).
the small blood vessels in selected disease of the Vascular dementia in a population-based autopsy
cerebral white matter. In Pantoni, L., Inzitari, D. and study. Archives of Neurology, 60 (4): 569–575.
Wallin, A. (eds.), The Matter of White Matter. Current Kovari, E., Gold, G., Herrmann, F.R. et al. (2004). Cortical
Issues in Neurodegenerative Diseases. Utrecht, The microinfarcts and demyelination significantly affect
Netherlands: Academic Pharmaceutical Productions, cognition in brain aging. Stroke: A Journal of Cerebral
pp. 247–273. Circulation, 35 (2): 410–414.
Ihara, M., Polvikoski, T.M., Hall, R. et al. (2010). Kovari, E., Gold, G., Herrmann, F.R. et al. (2007). Cortical
Quantification of myelin loss in frontal lobe white microinfarcts and demyelination affect cognition in
matter in vascular dementia, Alzheimer’s disease, and cases at high risk for dementia. Neurology, 68 (12):
dementia with Lewy bodies. Acta Neuropathologica, 927–931.
119 (5): 579–589. Kril, J.J., Patel, S., Harding, A.J. and Halliday, G.M. (2002).
Janaway, B.M., Simpson, J.E., Hoggard, N. et al. (2013). Patients with vascular dementia due to microvascular
Brain haemosiderin in older people: Pathological evi- pathology have significant hippocampal neuronal loss.
dence for an ischaemic origin of magnetic resonance Journal of Neurology, Neurosurgery and Psychiatry, 72
imaging (MRI) microbleeds. Neuropathology and (6): 747–751.
Applied Neurobiology, 40 (3): 258–269. Lammie, G.A. (2000). Pathology of small vessel stroke.
Jellinger, K.A. (2008). The pathology of "vascular British Medical Bulletin, 56 (2): 296–306.
­dementia": A critical update. Journal of Alzheimer’s Levy, E., Lopez-Otin, C., Ghiso, J. et al. (1989). Stroke in
Disease, 14 (1): 107–123. Icelandic patients with hereditary amyloid angiopa-
Jen, J., Cohen, A.H., Yue, Q. et al. (1997). Hereditary thy is related to a mutation in the cystatin C gene,
endotheliopathy with retinopathy, nephropathy, and an inhibitor of cysteine proteases. The Journal of
stroke (HERNS). Neurology, 49 (5): 1322–1330. Experimental Medicine, 169 (5): 1771–1778.
The neuropathology of vascular dementia 657

Leys, D., Henon, H., Mackowiak-Cordoliani, M.A. and O’Brien, J.T., Erkinjuntti, T., Reisberg, B. et al. (2003).
Pasquier, F. (2005). Poststroke dementia. The Lancet Vascular cognitive impairment. The Lancet Neurology,
Neurology, 4 (11): 752–759. 2 (2): 89–98.
Liu, C.K., Miller, B.L., Cummings, J.L. et al. (1992). A quan- O’Sullivan, M., Ngo, E., Viswanathan, A. et al. (2009).
titative MRI study of vascular dementia. Neurology, 42 Hippocampal volume is an independent predictor of
(1): 138–143. cognitive performance in CADASIL. Neurobiology of
Mann, D.M., Yates, P.O. and Marcyniuk, B. (1986). The Aging, 30 (6): 890–897.
nucleus basalis of Meynert in multi-infarct (vascular) Okamoto, Y., Ihara, M., Fujita, Y. et al. (2009). Cortical
dementia. Acta Neuropathologica, 71 (3–4): 332–337. microinfarcts in Alzheimer’s disease and subcortical
Markesbery, W. (1998). Vascular dementia. In Markesbery, vascular dementia. Neuroreport, 20 (11): 990–996.
W.R. (ed.), Neuropathology of Dementing Disorders. Okamoto, Y., Yamamoto, T., Kalaria, R.N. et al. (2012).
London, United Kingdom: Arnold, pp. 293–311. Cerebral hypoperfusion accelerates cerebral amyloid
Matthews, F.E., Brayne, C., Lowe, J. et al. (2009). angiopathy and promotes cortical microinfarcts. Acta
Epidemiological pathology of dementia: Attributable- Neuropathologica, 123 (3): 381–394.
risks at death in the Medical Research Council Olichney, J.M., Ellis, R.J., Katzman, R. et al. (1997). Types
Cognitive Function and Ageing Study. PLoS Medicine, of cerebrovascular lesions associated with severe cere-
6 (11): e1000180. bral amyloid angiopathy in Alzheimer’s disease. Annals
Mead, S., James-Galton, M., Revesz, T. et al. (2000). of the New York Academy of Sciences, 826: 493–497.
Familial British dementia with amyloid angiopa- Olichney, J.M., Hansen, L.A., Hofstetter, C.R. et al. (1995).
thy: Early clinical, neuropsychological and imaging Cerebral infarction in Alzheimer’s disease is associated
findings. Brain: A Journal of Neurology, 123 (Pt 5): with severe amyloid angiopathy and hypertension.
975–991. Archives of Neurology, 52 (7): 702–708.
Mirra, S.S. (1997). The CERAD neuropathology protocol Olichney, J.M., Hansen, L.A., Hofstetter, C.R., et al.
and consensus recommendations for the postmor- (2000a) Association between severe cerebral amyloid
tem diagnosis of Alzheimer’s disease: A commentary. angiopathy and cerebrovascular lesions in Alzheimer
Neurobiology of Aging, 18 (4 Suppl.): S91–S94. disease is not a spurious one attributable to apolipo-
Mulugeta, E., Molina-Holgado, F., Elliott, M.S. et al. protein E4. Archives of Neurology, 57 (6): 869–874.
(2008). Inflammatory mediators in the frontal lobe Olichney, J.M., Hansen, L.A., Lee, J.H. et al. (2000b).
of patients with mixed and vascular dementia. Relationship between severe amyloid angiopathy,
Dementia and Geriatric Cognitive Disorders, 25 (3): apolipoprotein E genotype, and vascular lesions in
278–286. Alzheimer’s disease. Annals of the New York Academy
Nagy, Z., Esiri, M.M., Jobst, K.A. et al. (1997). The effects of Sciences, 903: 138–143.
of additional pathology on the cognitive deficit in Ophoff, R.A., DeYoung, J., Service, S.K. et al. (2001).
Alzheimer disease. Journal of Neuropathology and Hereditary vascular retinopathy, cerebroretinal vascu-
Experimental Neurology, 56 (2): 165–170. lopathy, and hereditary endotheliopathy with retinopa-
Natte, R., Maat-Schieman, M.L., Haan, J. et al. (2001). thy, nephropathy, and stroke map to a single locus on
Dementia in hereditary cerebral hemorrhage with chromosome 3p21.1-p21.3. The American Journal of
amyloidosis-Dutch type is associated with cerebral Human Genetics, 69 (2): 447–453.
amyloid angiopathy but is independent of plaques and Pantoni, L. (2010). Cerebral small vessel disease: From
neurofibrillary tangles. Annals of Neurology, 50 (6): pathogenesis and clinical characteristics to therapeutic
765–772. challenges. The Lancet Neurology, 9 (7): 689–701.
Nelson, P.T., Schmitt, F.A., Lin, Y. et al. (2011). Pantoni, L. and Garcia, J.H. (1997). Pathogenesis of
Hippocampal sclerosis in advanced age: Clinical and leukoaraiosis: A review. Stroke: A Journal of Cerebral
pathological features. Brain: A Journal of Neurology, Circulation, 28 (3): 652–659.
134 (Pt 5): 1506–1518. Pavlovic, A.M., Zidverc-Trajkovic, J., Milovic, M.M. et al.
Neltner, J.H., Abner, E.L., Baker, S. et al. (2014). (2005). Cerebral small vessel disease in pseudoxan-
Arteriolosclerosis that affects multiple brain regions thoma elasticum: Three cases. Canadian Journal of
is linked to hippocampal sclerosis of ageing. Brain: Neurological Sciences, 32 (1): 115–118.
A Journal of Neurology, 137 (Pt 1): 255–267. Pendlebury, S.T. and Rothwell, P.M. (2009). Prevalence,
Neuropathology Group of the Medical Research Council incidence, and factors associated with pre-stroke and
Cognitive Function and Ageing Study (MRC CFAS) post-stroke dementia: A systematic review and meta-
(2001). Pathological correlates of late-onset demen- analysis. The Lancet Neurology, 8 (11): 1006–1018.
tia in a multicentre, community-based population Perry, E.K., Gibson, P.H., Blessed, G. et al. (1977).
in England and Wales. Neuropathology Group of Neurotransmitter enzyme abnormalities in senile
the Medical Research Council Cognitive Function dementia. Choline acetyltransferase and glutamic
and Ageing Study (MRC CFAS). Lancet, 357 (9251): acid decarboxylase activities in necropsy brain tissue.
169–175. Journal of the Neurological Sciences, 34 (2): 247–265.
658 Dementia

Pfeifer, L.A., White, L.R., Ross, G.W. et al. (2002). Sachdev, P., Kalaria, R.N., O’Brien, J.T. et al. (2014).
Cerebral amyloid angiopathy and cognitive func- Diagnostic criteria for vascular cognitive disorders:
tion: The HAAS autopsy study. Neurology, 58 (11): A VASCOG statement. Alzheimer Disease and
1629–1634. Associated Disorders, 28 (3): 206.
Plant, G.T., Revesz, T., Barnard, R.O. et al. (1990). Familial Scher, A.I., Xu, Y., Korf, E.S. et al. (2011). Hippocampal
cerebral amyloid angiopathy with nonneuritic amyloid morphometry in population-based incident
plaque formation. Brain: A Journal of Neurology, 113 Alzheimer’s disease and vascular dementia: The HAAS.
(Pt 3): 721–747. Journal Neurology, Neurosurgery and Psychiatry, 82
Pohjasvaara, T., Erkinjuntti, T., Vataja, R. and Kaste, M. (4): 373–376.
(1997). Dementia three months after stroke. Baseline Schneider, J.A., Arvanitakis, Z., Bang, W. and Bennett,
frequency and effect of different definitions of demen- D.A. (2007). Mixed brain pathologies account for most
tia in the Helsinki Stroke Aging Memory Study (SAM) dementia cases in community-dwelling older persons.
cohort. Stroke: A Journal of Cerebral Circulation, 28 Neurology, 69 (24): 2197–2204.
(4): 785–792. Seno, H., Ishino, H., Inagaki, T. et al. (1999). A neuro-
Pohjasvaara, T., Erkinjuntti, T., Ylikoski, R. et al. (1998). pathological study of dementia in nursing homes
Clinical determinants of poststroke dementia. over a 17-year period, in Shimane Prefecture, Japan.
Stroke: A Journal of Cerebral Circulation, 29 (1): Gerontology, 45 (1): 44–48.
75–81. Sharp, S.I., Francis, P.T., Elliott, M.S. et al. (2009). Choline
Rauramaa, T., Pikkarainen, M., Englund, E. et al. (2013). acetyltransferase activity in vascular dementia and
Consensus recommendations on pathologic changes stroke. Dementia and Geriatric Cognitive Disorders, 28
in the hippocampus: A postmortem multicenter (3): 233–238.
inter-rater study. Journal of Neuropathology and Simpson, J.E., Fernando, M.S., Clark, L. et al. (2007).
Experimental Neurology, 72 (6): 452–461. White matter lesions in an unselected cohort of the
Reijmer, Y.D., van Veluw, S.J. and Greenberg, S.M. (2015). elderly: Astrocytic, microglial and oligodendrocyte
Ischemic brain injury in cerebral amyloid ­angiopathy. precursor cell responses. Neuropathology and Applied
Journal of Cerebral Blood Flow and Metabolism, Neurobiology, 33 (4): 410–419.
36 (1): 40–54. Simpson, J.E., Hosny, O., Wharton, S.B. et al. (2009).
Revesz, T., Holton, J.L., Lashley, T. et al. (2009). Genetics Microarray RNA expression analysis of cerebral white
and molecular pathogenesis of sporadic and matter lesions reveals changes in multiple functional
hereditary cerebral amyloid angiopathies. Acta pathways. Stroke: A Journal of Cerebral Circulation, 40
Neuropathologica, 118 (1): 115–130. (2): 369–375.
Riekse, R.G., Leverenz, J.B., McCormick, W. et al. (2004). Smallwood, A., Oulhaj, A., Joachim, C. et al. (2012).
Effect of vascular lesions on cognition in Alzheimer’s Cerebral subcortical small vessel disease and its rela-
disease: A community-based study. Journal of the tion to cognition in elderly subjects: A pathological
American Geriatrics Society, 52 (9): 1442–1448. study in the Oxford Project to Investigate Memory
Roher, A.E., Esh, C., Kokjohn, T. et al. (2005). and Ageing (OPTIMA) cohort. Neuropathology and
Atherosclerosis and AD: Analysis of data from the U.S. Applied Neurobiology, 38 (4): 337–343.
National Alzheimer’s Coordinating Center. Neurology, Snowdon, D.A., Greiner, L.H., Mortimer, J.A. et al. (1997).
65 (6): 974. Brain infarction and the clinical expression of Alzheimer
Román, G.C. (2002). Vascular dementia ­revisited: disease. The Nun Study. JAMA, 277 (10): 813–817.
Diagnosis, pathogenesis, treatment, and Sonnen, J.A., Larson, E.B., Crane, P.K. et al. (2007).
­prevention. Medical Clinics of North America, 86 Pathological correlates of dementia in a longitudinal,
(3): 477–499. population-based sample of aging. Annals of
Román, G.C. (2004). Brain hypoperfusion: A critical factor Neurology, 62 (4): 406–413.
in vascular dementia. Neurological Research, 26 (5): Strozyk, D., Dickson, D.W., Lipton, R.B. et al. (2010).
454–458. Contribution of vascular pathology to the clinical
Román, G.C. and Kalaria, R.N. (2006). Vascular determi- expression of dementia. Neurobiology of Aging, 31
nants of cholinergic deficits in Alzheimer disease and (10): 1710–1720.
vascular dementia. Neurobiology of Aging, 27 (12): Suter, O.C., Sunthorn, T., Kraftsik, R. et al. (2002). Cerebral
1769–1785. hypoperfusion generates cortical watershed micro-
Román, G.C., Tatemichi, T.K., Erkinjuntti, T. et al. (1993). infarcts in Alzheimer disease. Stroke: A Journal of
Vascular dementia: Diagnostic criteria for research Cerebral Circulation, 33 (8): 1986–1992.
studies. Report of the NINDS-AIREN International Tanskanen, M., Kalaria, R.N., Notkola, I.L. et al. (2013).
Workshop. Neurology, 43 (2): 250–260. Relationships between white matter hyperintensities,
Sachdev, P. (1999). Vascular cognitive disorder. cerebral amyloid angiopathy and dementia in a
International Journal of Geriatric Psychiatry, 14 (5): population-based sample of the oldest old. Current
402–403. Alzheimer Research, 10 (10): 1090–1097.
The neuropathology of vascular dementia 659

Terwindt, G.M., Haan, J., Ophoff, R.A. et al. (1998). Wattendorff, A.R., Bots, G.T., Went, L.N. and Endtz, L.J.
Clinical and genetic analysis of a large Dutch fam- (1982). Familial cerebral amyloid angiopathy present-
ily with autosomal dominant vascular retinopathy, ing as recurrent cerebral haemorrhage. Journal of the
migraine and Raynaud’s phenomenon. Brain: A Journal Neurological Sciences, 55 (2): 121–135.
of Neurology, 121 (Pt 2): 303–316. Werring, D.J., Gregoire, S.M. and Cipolotti, L. (2011).
Toledo, J.B., Arnold, S.E., Raible, K. et al. (2013). Cerebral microbleeds and vascular cognitive impair-
Contribution of cerebrovascular disease in autopsy ment. Journal of the Neurological Sciences, 299 (1–2):
confirmed neurodegenerative disease cases in the 131–135.
National Alzheimer’s Coordinating Centre. Brain: Westover, M.B., Bianchi, M.T., Yang, C. et al. (2013).
A Journal of Neurology, 136 (Pt. 9): 2697–2706. Estimating cerebral microinfarct burden from autopsy
Tomlinson, B.E., Blessed, G. and Roth, M. (1970). samples. Neurology, 80 (15): 1365–1369.
Observations on the brains of demented old White, L. (2009). Brain lesions at autopsy in older
­people. Journal of the Neurological Sciences, Japanese-American men as related to cogni-
11 (3): 205–242. tive impairment and dementia in the final years
Vahedi, K., Massin, P., Guichard, J.P. et al. (2003). Hereditary of life: A summary report from the Honolulu-Asia
infantile hemiparesis, retinal arteriolar tortuosity, and aging study. Journal of Alzheimer’s Disease, 18
leukoencephalopathy. Neurology, 60 (1): 57–63. (3): 713–725.
van de Pol, L., Gertz, H.J., Scheltens, P. and Wolf, H. (2011). White, L., Petrovitch, H., Hardman, J. et al. (2002).
Hippocampal atrophy in subcortical vascular dementia. Cerebrovascular pathology and dementia in autopsied
Neurodegenerative Diseases, 8 (6): 465–469. Honolulu-Asia Aging Study participants. Annals of the
Van der Flier, W.M. and Cordonnier, C. (2012). New York Academy of Science, 977: 9–23.
Microbleeds in vascular dementia: Clinical aspects. White, L., Small, B.J., Petrovitch, H. et al. (2005). Recent
Experimental Gerontology, 47 (11): 853–857. clinical-pathologic research on the causes of demen-
Verreault, S., Joutel, A., Riant, F. et al. (2006). A novel tia in late life: Update from the Honolulu-Asia Aging
hereditary small vessel disease of the brain. Annals of Study. Journal of Geriatric Psychiatry and Neurology,
Neurology, 59 (2): 353–357. 18 (4): 224–227.
Vidal, R., Frangione, B., Rostagno, A. et al. (1999). Yamamoto, Y., Craggs, L., Baumann, M. et al. (2011).
A ­stop-codon mutation in the BRI gene associated Molecular genetics and pathology of hereditary small
with familial British dementia. Nature, 399 (6738): vessel diseases of the brain. Neuropathology and
776–781. Applied Neurobiology, 37 (1): 94–113.
Vidal, R., Revesz, T., Rostagno, A. et al. (2000). A decamer Yamamoto, Y., Ihara, M., Tham, C. et al. (2009).
duplication in the 3’ region of the BRI gene originates Neuropathological correlates of temporal pole white
an amyloid peptide that is associated with dementia matter hyperintensities in CADASIL. Stroke: A Journal
in a Danish kindred. Proceedings of the National of Cerebral Circulation, 40 (6): 2004–2011.
Academy of Sciences of the United States of America, Yarchoan, M., Xie, S.X., Kling, M.A. et al. (2012).
97 (9): 4920–4925. Cerebrovascular atherosclerosis correlates with
Vieira, R.T., Caixeta, L., Machado, S. et al. (2013). Alzheimer pathology in neurodegenerative dementias.
Epidemiology of early-onset dementia: A review of the Brain: A Journal of Neurology, 135 (Pt 12): 3749–3756.
literature. Clinical Practice and Epidemiology in Mental Zarow, C., Vinters, H.V., Ellis, W.G. et al. (2005). Correlates
Health, 9: 88–95. of hippocampal neuron number in Alzheimer’s disease
Vinters, H.V., Ellis, W.G., Zarow, C. et al. (2000). and ischemic vascular dementia. Annals of Neurology,
Neuropathologic substrates of ischemic vascular 57 (6): 896–903.
dementia. Journal of Neuropathology and Zarow, C., Weiner, M.W., Ellis, W.G. and Chui, H.C. (2012).
Experimental Neurology, 59 (11): 931–945. Prevalence, laterality, and comorbidity of hippocampal
Viswanathan, A., Patel, P., Rahman, R. et al. (2008). Tissue sclerosis in an autopsy sample. Brain and Behavior, 2
microstructural changes are independently associated (4): 435–442.
with cognitive impairment in cerebral amyloid angi- Zekry, D., Duyckaerts, C., Moulias, R. et al. (2002).
opathy. Stroke: A Journal of Cerebral Circulation, 39 Degenerative and vascular lesions of the brain have
(7): 1988–1992. synergistic effects in dementia of the elderly. Acta
Wardlaw, J.M. (2010). Blood-brain barrier and cerebral Neuropathologica, 103 (5): 481–487.
small vessel disease. Journal of the Neurological
Sciences, 299 (1–2): 66–71.
60
Therapeutic strategies for vascular cognitive
disorders

GUSTAVO C. ROMÁN

double-blind randomized controlled trial (RCT) in


60.1 INTRODUCTION Finland (Ngandu et al., 2015), on 1260 subjects ages 60–77
years, demonstrated that after 2 years of a multidomain
The therapeutic management of vascular cognitive dis- intervention (diet, exercise, cognitive training, vascular
orders (VCDs) including vascular dementia (VaD) is par- risk monitoring), cognition improved or remained bet-
ticularly challenging because of the multiple types of ter in the intervention group compared with the control
cerebrovascular injuries and circulatory events that may group that received only general health advice.
lead to cognitive decline and eventual dementia. The mul- Lastly, VaD is often complicated by neuropsychiatric
tiplicity of pathogenetic mechanisms results in a variety symptoms, in particular depression, apathy, aggressiveness
of clinical presentations that have hampered the formula- and other behavioural changes requiring careful manage-
tion of diagnostic criteria and the ascertainment of cases ment (Román, 2014). With exception of the dextromethor­
for controlled clinical trials. The International Society for phan plus ultra low-dose quinidine trial showing reduction
Vascular Behavioural and Cognitive Disorders (VASCOG) of pseudobulbar effect and some of the above manifesta-
recently proposed a unified set of criteria for VCD (Sachdev tions (Pioro et al., 2010), few clinical trials have addressed
et al., 2014) that addressed most of those problems. The these issues.
VASCOG criteria recognized the continuum of vascular
cognitive impairment (VCI) ranging from mild VCD at one
end to VaD or major VCD at the other end, and addressed
the important role of brain imaging in clinical trials of VaD
(Black, 2007; Román et al., 2010). 60.2 DEFINITIONS
A recent development is the recognition of the impor-
tance of vascular lesions in the pathogenesis and clinical 60.2.1 VASCULAR COGNITIVE DISORDERS
presentation of most cases of late-onset Alzheimer’s dis- (VCDs)
ease (AD) (Roman, 2002a), supporting the concept that
vigorous treatment of vascular risk factors in patients Sachdev (1999) first indicated the need for a categorical
with early cognitive decline could delay the onset or pre- entity – a disorder – to include the large numbers of patients
vent dementia (Román et al., 2012; Willis and Hakim, with dementia and cognitive decline of vascular aetiol-
2013; Román and Boller, 2014). It has been estimated that ogy and proposed the name ‘vascular cognitive disorder’.
one-third of AD cases worldwide might be attributable to VASCOG formally proposed VCD as a diagnostic category
potentially modifiable vascular risk factors (Norton et al., (Sachdev et al., 2014). VCD recognizes the need for a cat-
2014). According to Barnes and Yaffe (2011) ‘A 10%–25% egorical diagnosis that encompasses mild impairment, pre-
reduction in all seven risk factors (diabetes, midlife hyper- dementia and dementia syndromes. VASCOG included
tension, midlife obesity, smoking, depression, cognitive the recommendations of expert groups from the National
inactivity or low educational attainment and physical Institute of Neurological Disorders and Stroke (NINDS)–
inactivity) could potentially prevent as many as 1.1–3.0 Canadian Stroke Network (Hachinski et al., 2006) and the
million AD cases worldwide’. A recent, population-based statement from the American Heart Association/American
660
Therapeutic strategies for vascular cognitive disorders 661

Stroke Association (Gorelick et al., 2011). VCD is defined pharmacological research in this domain of major public
as a diagnostic category that embraces many syndromes health importance.
and diseases, and acknowledges the common occurrence of The VASCOG statement (Sachdev et al., 2014) unified
non-cognitive syndromes such as depression, anxiety and seven sets of criteria for VaD, including Hachinski Ischaemic
psychosis. It includes all forms of cognitive decline result- Score (HIS) (Hachinski et al., 1975), National Institute of
ing from cerebrovascular disease (CVD) ranging from Neurological Disorders and Stroke–Association Internationale
mild cognitive deficits (mild VCD) to VaD (Román et al., pour la Recherche et l’Enseignement en Neurosciences
2004). VCD excludes isolated cognitive dysfunctions result- (NINDS-AIREN) (Román et al., 1993), Alzheimer’s Disease
ing from stroke (e.g. aphasia, apraxia or agnosia). VCD Diagnostic and Treatment Center (ADDTC) (Chui et al.,
includes all the syndromes and diseases characterized by 1992), Diagnostic and Statistical Manual of Mental Disorders,
cognitive impairment resulting from cerebrovascular aeti- 4th Edition (DSM-IV) (American Psychiatric Association,
ologies including post-stroke VaD, Cerebral Autosomal 1994), International Statistical Classification of Diseases, 10th
Dominant Arteriopathy with Subcortical Infarcts and Revision (ICD-10) (World Health Organization, 2008), the
Leukoencephalopathy (CADASIL), Binswanger’s disease subcortical VaD criteria (Erkinjuntti et al., 2000) and the vas-
(BD) and mixed dementia (AD+CVD). Regarding specific cular cognitive impairment without dementia (VCI-ND) crite-
aetiologies of VCD (Table 60.1), Mendez and Cummings ria (Rockwood et al., 2000; Ingles et al., 2002). Although all of
(2003) collected more than 100 potential vascular causes them are useful in selecting VaD patients with varying sensitiv-
of cognitive impairment and dementia. This concep- ity and specificity, they are not interchangeable (Verhey et al.,
tual clarification should facilitate epidemiological and 1996; Wetterling et al., 1996; Chui et al., 2000; Pohjasvaara

Table 60.1 Pathological basis for vascular cognitive disorders according to VASCOG criteria
Parenchymal lesions of vascular aetiology
(1) Large vessel or atherothromboembolic disease
(a) Multiple infarcts
(b) Single strategically placed infarct
(2) Small vessel disease
(a) Multiple lacunar infarcts in white matter and deep grey matter nuclei
(b) Ischemic white matter change
(c) Dilatation of perivascular spaces
(d) Cortical/subcortical microinfarcts and microhemorrhages
(3) Haemorrhage
(a) Intracerebral haemorrhage
(b) Multiple cortical and subcortical microbleeds
(c) Subarachnoid haemorrhage
(4) Hypoperfusion
(a) Hippocampal sclerosis
(b) Laminar cortical sclerosis
Types of vascular lesions
(1) Atherosclerosis
(2) Cardiac, atherosclerotic and systemic emboli
(3) Arteriolosclerosis
(4) Lipohyalinosis
(5) Amyloid angiopathy
(6) Vasculitis – infectious and non-infectious
(7) Venous collagenosis
(8) Arteriovenous fistulae – dural or parenchymal
(9) Hereditary angiopathies – cerebral autosomal dominant arteriopathy with subcortical infarcts and
leukoencephalopathy (CADASIL), cerebral autosomal recessive arteriopathy with subcortical autosomal recessive
leukoencephalopathy (CARASIL), etc.
(10) Giant cell arteritis
(11) Berry aneurysms
(12) Miscellaneous vasculopathies – fibromuscular dysplasia, Moya-Moya
(13) Systemic microangiopathies without vascular inflammatory cell infiltrates
(14) Cerebral venous thrombosis
Source: Sachdev, P. et al., Alzheimer Disease and Associated Disorders, 28, 206–218, 2014.
Abbreviation: VASCOG, International Society for Vascular Behavioural and Cognitive Disorders.
662 Dementia

et al., 2000; Wiederkehr et al., 2008a, 2008b). The VASCOG haemorrhagic stroke, as well as from ischaemic hypoper-
criteria provide a scientific and coherent approach to the study fusive brain injuries, affecting brain regions important for
of this group of disorders unifying the terminology under the memory, cognition and behaviour.
name VCD. Also, VASCOG provides diagnostic thresholds Table 60.1 lists the most common neuropathological
and proposes clinical and neuroimaging criteria for establish- lesions capable of producing VaD including brain haemor-
ing a vascular aetiology, describes subtypes and emphasizes the rhages and hypoperfusive and ischaemic (occlusive) vas-
frequent co-occurrence of AD and vascular pathology. cular brain injuries (Brun, 1994; Parnetti, 1999; Mikol and
Vallat-Decouvelaere, 2000; Vinters et al., 2000; Jefferson
60.2.2 MILD VCD – MILD VCI et al., 2015a, 2015b).
The VASCOG criteria for VaD (Sachdev et al., 2014)
In 2004, an International Working Group on Mild Cognitive require that in addition to the criteria of mild cognitive
Impairment (VCI) proposed a set of recommendations for disorder listed above, the objective deficits in neurocogni-
the diagnosis of VCI (Winblad et al., 2004). According to tive tests typically should fall ≥2 standard deviations (SDs)
VASCOG (Sachdev et al., 2014), the term ‘impairment’ in this below the mean (or below the third percentile) of people of
context should be discouraged because of two major limita- similar age, sex, education and sociocultural background,
tions: (1) impairment is used in medicine to indicate a reduc- or an equivalent level as judged by the clinician; also, the
tion or loss of function applied either as a statistical construct cognitive deficits are sufficient to interfere with the subject’s
based on normative data or as a demonstration of disability independence.
and (2) VCI has sometimes been used to denote mild cogni-
tive impairment (MCI) due to vascular factors, insufficiently 60.2.3.1 The NINDS-AIREN VaD criteria
severe to be diagnosed as dementia (Román et al., 2004) by
analogy to MCI and considered as the earliest manifestation The criteria developed by the NINDS-AIREN (Román
of AD (Petersen et al., 1999). Also, VCI is sometimes equated et al., 1993) were developed specifically for research stud-
with dysexecutive MCI (Winblad et al., 2004) and with ies in VaD, including most vascular aetiologies and clini-
VCI without dementia or VCI-ND (Rockwood et al., 2000; cal manifestations (Table 60.3). These criteria have the
Ingles et al., 2002). Instead of VCI and to harmonize with highest specificity (0.84–0.94) (Gold et al., 1997, 2002) and
Diagnostic and Statistical Manual of Mental Disorders, 5th have been used in most RCTs of VaD (Rosen et al., 1980;
Edition (DSM-5) (American Psychiatric Association, 2013), López-Pousa et al., 1997; Kittner, 1999; Erkinjuntti et al.,
VASCOG recommended using the categories mild VCD, at 2002a; Orgogozo et al., 2002; Wilcock et al., 2002; Black et al.,
one end of severity, and VaD or major VCD, at the other end. 2003; Wilkinson et al., 2003; Black, 2007). These criteria rec-
Table 60.2 summarizes the VASCOG criteria for mild VCD. ognize patients with multiple forms of stroke and CVD, but
establish strict requirements to classify patients appropri-
60.2.3 VASCULAR DEMENTIA ately. Two cardinal elements of the clinical syndrome of VaD
must be identified: the cognitive syndrome of dementia and
Román (2002b, 2003a) defined VaD as an aetiological the definition of the vascular cause of the dementia. CVD is
category of dementia characterized by acquired intellec- defined by the presence of focal neurological signs and brain
tual impairment, severe enough to interfere with social imaging showing evidence of stroke or ischaemic changes in
and personal independence, resulting from ischaemic or the brain. A defining element of the criteria is the need for a

Table 60.2 VASCOG diagnostic criteria for mild cognitive disorder


A. Acquired decline from a documented or inferred previous level of performance in ≥1 of the following cognitive
domains: (1) attention and processing speed, (2) frontal executive function, (3) learning and memory, (4) language,
(5) visuoconstructional–perceptual ability, (6) praxis-gnosis-body schema, (7) social cognition, as evidenced by the
following:
(a) Concerns of a patient, knowledgeable informant or a clinician of mild levels of decline from a previous level of
cognitive functioning. Typically, the reports will involve greater difficulty in performing the tasks or the use of
compensatory strategies.
(b) Evidence of modest deficits on objective cognitive assessment based on a validated measure of neurocognitive
function (either formal neuropsychological testing or an equivalent clinical evaluation) in ≥1 cognitive domains listed
above. The test performance is typically in the range between 1 and 2 standard deviations below appropriate norms
(or between the 3rd and 16th percentiles) when a formal neuropsychological assessment is available or an equivalent
level as judged by the clinician.
B. The cognitive deficits are not sufficient to interfere with independence (i.e. instrumental activities of daily living are
preserved), but greater effort, compensatory strategies or accommodation may be required to maintain
independence.
Source: Sachdev, P. et al., Alzheimer Disease and Associated Disorders, 28, 206–218, 2014.
Therapeutic strategies for vascular cognitive disorders 663

Table 60.3 NINDS-AIREN diagnostic criteria for vascular dementia (VaD)


I. The criteria for the diagnosis of probable VaD include all of the following:
(a) DEMENTIA: Impairment of memory and two or more cognitive domains (including executive function), interfering with
ADLs and not due to physical effects of stroke alone. Exclusion criteria: Alterations of consciousness, delirium,
psychoses, severe aphasia or deficits precluding testing, systemic disorders, Alzheimer’s disease or other forms of
dementia.
(b) CEREBROVASCULAR DISEASE (CVD): Focal signs on neurological examination (hemiparesis, lower facial weakness, Babinski
sign, sensory deficit, hemianopsia, dysarthria) consistent with stroke (with or without history of stroke) and evidence
of relevant CVD by brain CT or MRI including multiple large vessel infarcts or a single strategically placed infarct
(angular gyrus, thalamus, basal forebrain, or PCA or ACA territories), as well as multiple basal ganglia and white
matter lacunes or extensive periventricular white matter lesions, or combinations thereof. Exclusion criteria:
Absence of cerebrovascular lesions on CT or MRI.
(c) A RELATIONSHIP BETWEEN THE ABOVE TWO DISORDERS: Manifested or inferred by the presence of one or more of the following:
(i) Onset of dementia within 3 months following a recognized stroke.
(ii) Abrupt deterioration in cognitive functions or fluctuating, stepwise progression of cognitive deficits.
  II. Clinical features consistent with the diagnosis of probable VaD include the following:
(a) Early presence of gait disturbances (small step gait or marche à petits pas, or magnetic, apraxic–ataxic or
parkinsonian gait).
(b) History of unsteadiness and frequent, unprovoked falls.
(c) Early urinary frequency, urgency and other urinary symptoms not explained by urologic disease.
(d) Pseudobulbar palsy.
(e) Personality and mood changes, abulia, depression, emotional incontinence, or other deficits including psychomotor
retardation and abnormal executive function.
III. Features that make the diagnosis of VaD uncertain or unlikely include:
(a) Early onset of memory deficit and progressive worsening of memory and other cognitive functions such as language
(transcortical sensory aphasia), motor skills (apraxia) and perception (agnosia), in the absence of corresponding
focal lesions on brain imaging.
(b) Absence of focal neurological signs, other than cognitive disturbances.
(c) Absence of CVD on CT or MRI.
Source: Román, G.C. et al., Neurology, 43, 250–260, 1993.
Abbreviations: INDS-AIREN, National Institute of Neurological Disorders and Stroke–Association Internationale pour la Recherche et
l’Enseignement en Neurosciences; ADLs, activities of daily living; CT, computerized tomography; MRI, magnetic resonance imaging;
PCA, posterior cerebral artery; ACA, anterior cerebral artery.

temporal relationship between dementia and CVD whereby from placebo groups in AD (Román and Rogers, 2004) as
dementia onset should occur within 3 months following a well as in mixed AD plus CVD trials (Erkinjuntti et al.,
recognized stroke. The temporal relationship proved to be 2002a, 2002b), further indicating the separate identity of the
difficult to fulfil due to silent strokes and subcortical VaD. VaD cases selected (Pratt, 2002). The latest donepezil trial in
The accuracy of the NINDS-AIREN criteria increases by VaD recruited 974 patients (mean age 73.0 years) with prob-
excluding subjects with pre-existing diagnosis of AD, those able or possible VaD who were randomized 2:1 to receive
with isolated memory impairment (MCI), as well as those donepezil 5 mg/day or placebo (Román et al., 2010). Thus,
with history of obvious progressive cognitive decline prior almost 2200 patients with unmixed VaD identified with the
to a stroke or pre-stroke dementia (Hénon et al., 1997, 2001). NINDS-AIREN criteria have been studied in clinical trials
VaD dementia typically occurs in patients with stroke and using a cholinesterase inhibitor (ChEI).
multiple vascular risk factors. The HIS quantifies this profile
with high interrater reliability (kappa = 0.61). A score >4 60.2.4 VaD WITH AD
in the HIS usually excludes AD. Using the NINDS-AIREN
criteria (Román et al., 1993), three international trials of Specific brain pathological lesions define the two most com-
donepezil in VaD (Black et al., 2003; Wilkinson et al., 2003; mon forms of senile dementia, AD and VaD. Stroke and
Román et al., 2010) recruited 2193 patients with relatively CVD are the hallmarks of VaD, while senile plaques and
pure or unmixed VaD enrolled at 111 investigational sites neurofibrillary tangles typify AD. When these two patholo-
in the United States, Europe, Canada and Australia (Román gies are combined, the neuropathological diagnoses of
et al., 2005). The VaD population selected was older and AD+CVD, VaD with AD, or ‘mixed dementia’ are invoked
had a male preponderance, in agreement with popula- (Zekry et al., 2002a). However, the term ‘mixed dementia’ is
tion-based epidemiological data on VaD (Román, 2008b). ambiguous and is not recommended by VASCOG.
Furthermore, the 6-month progression of the placebo In the experience of the Consortium to Establish a
groups selected in the donepezil VaD studies was different Registry for AD (CERAD), vascular pathology also coexists
664 Dementia

with Lewy body disease in isolation or in combination with The first and foremost requirement for a valid trial in
AD pathology (Mirra, 1997). Coexistent lesions occur in dementia, and VaD in particular, is the use of appropriate
up to one-third of unselected elderly demented patients inclusion and exclusion criteria. This requirement is crucial
(Kalaria and Ballard, 1999; Zekry et al., 2002a, 2002b, for the selection of a homogeneous population that would
2002c) or in 10%–15% of cases of post-stroke dementia allow the generalized application of the trial results. Given the
(PSD) (Hénon et al., 1997, 2001; Barba et al., 2000, 2001), clinical variety of VaD, some authors have suggested limiting
but mixed lesions also occur in patients without dementia the trials to patients with subcortical VaD using various sets
(Román and Royall, 2004). of criteria (Pantoni et al., 1996, 2000; Erkinjuntti et al., 2000;
A solid body of clinical, imaging, epidemiological and Moretti et al., 2001, 2002, 2004) or to cases of mixed demen-
neuropathological evidence has accumulated over the past tia, including AD+CVD (Erkinjuntti et al., 2002a, 2002b,
two decades confirming the critical role of CVD in AD 2003a) or AD plus arterial hypertension (Kumar et al., 2000).
and other dementias of the aged subjects (Chui et al., 2006; However, as discussed above, the criteria proposed by Román
Schneider et al., 2007; Fotuhi et al., 2009; Gorelick et al., et al. (1993) are appropriate for VaD trials. Several instru-
2011; Wharton et al., 2011; De Carli, 2012; Yarchoan et al., ments such as the Mini-Mental State Examination (MMSE)
2012; Bennett et al., 2013; Yates et al., 2014). Data from the (Folstein et al., 1975), the Cambridge Cognitive Examination
National Alzheimer’s Coordinating Center (Toledo et al., (CAMCOG) (Roth et al., 1986; Huppert et al., 1995), the
2013) showed the presence of vascular pathology in 80% Alzheimer’s Disease Assessment Scale-cognitive subscale
of 4629 brains from patients with neuropathologically (ADAS-cog) (Rosen et al., 1984), the Clinical Dementia Rating
confirmed AD. Lesions included large vessel disease with (CDR) (Berg, 1988) and the Montreal Cognitive Assessment
atherosclerosis in the arteries of the circle of Willis and its (MoCA) (Nasreddine et al., 2005) are used to ensure that
branches resulting in large territorial infarcts, small vessel dementia severity is comparable across the trial.
disease with arteriolosclerosis and small infarcts includ- The next requirement is an appropriate number of par-
ing lacunes and multiple microinfarcts, Binswanger’s type ticipants (sample size). The sample size is a function not
ischaemic periventricular leukoencephalopathy and brain only of the drug’s effectiveness but also of the degree of
haemorrhages. Notably, cerebral amyloid angiopathy (CAA) responsiveness – or lack thereof – of the measuring instru-
was present in less than half of the brains (41%). Population- ments, as well as of the accuracy of the end points selected
based autopsy studies find pure AD and pure cases of VaD for the trial. Instruments that are relatively insensitive to
to be uncommon in older demented subjects. In this age change require larger sample sizes in order to demonstrate
group, the neuropathology rule is the presence at autopsy of a statistically significant difference from the placebo group.
mixed dementia with a predominant vascular component Other requirements include randomization with appro-
(Zekry et al., 2002a). According to Matthews et al. (2009), priate group allocation concealment, placebo-controlled,
the main attributable risks at death for dementia are age double-blinded design with parallel group, and specific
(18%), small brain (12%), neocortical neuritic plaques (8%) outcome measures, adequate instruments and well-defined
and neurofibrillary tangles (11%), small vessel disease (12%), end points. Potential targets for the symptomatic treatment
multiple vascular pathologies (9%), hippocampal atrophy of VaD include: (1) symptomatic improvement of the core
(10%), CAA (7%) and Lewy bodies (3%). The confirmation symptoms (cognition, function and behaviour), (2) slow-
of the role of vascular lesions in AD explains some of the ing of progression and (3) treatment of neuropsychiatric
difficulties with consensus criteria for VaD (Black, 2007) symptoms.
and offers hope for the treatment and prevention of late- All the current VaD trials have used the same primary
life dementia (Brookmeyer et al., 2007; Polidori et al., 2012; outcome measures used in AD trials, as recommended
Anstey et al., 2013). by the US Food and Drug Administration (FDA). These
instruments include the ADAS-cog (Rosen et al., 1984),
the Clinician’s Interview-Based Impression of Change plus
caregiver input (CIBIC-plus) (Schneider et al., 1997) or the
Clinical Global Impression of Change (CGIC) (Berg, 1988).
60.3 METHODOLOGICAL ASPECTS OF
The European Medicines Agency (EMEA) Committee
CLINICAL TRIALS IN VaD for Medicinal Products for Human Use (CHMP) also
requires positive impact on activities of daily living (ADLs)
The methodological requirements for RCTs in patients with using scales such as the Alzheimer’s Disease Functional
dementia were reviewed by the International Working Group Assessment and Change Scale (ADFACS), which pro-
on Harmonization of Dementia Drug Guidelines at the vides a measure of instrumental ADLs (IADLs) and basic
Sixth International Congress on AD and Related Disorders ADLs (Mohs et al., 2001), or the Disability Assessment for
in Amsterdam (Whitehouse et al., 1998), and at the 1998 Dementia (DAD) scale (Gélinas et al., 1999). The CHMP
Osaka Conference on VaD (Erkinjuntti and Sawada, 1999; also requires a responder analysis. To the extent that ADAS-
Erkinjuntti et al., 1999); for more recent reviews, see Mills cog and CIBIC-plus assess the multiple cognitive domains
and Chow (2003), Mani (2004), Erkinjuntti et al. (2004), affected in all forms of dementia, it is not unreasonable to
Hachinski et al. (2006) and Sachdev et al. (2014). use the same measures in both AD and VaD populations.
Therapeutic strategies for vascular cognitive disorders 665

However, Quinn et al. (2000) showed difficulties with (multi-infarct dementia or MID). VaD can result from a
CIBIC-plus ratings in instances of clinical improvement; single stroke located in one of the following three possible
often physicians failed to recognize successful disease treat- areas (Delay and Brion, 1962):
ment beyond reversal of progression.
More importantly, current tests are relatively insensitive 1. Posterior cerebral artery (PCA) territory, involving
to frontal/subcortical executive dysfunction, a key cognitive ventral-medial temporal lobe, hippocampus, occipital
domain in VaD (Román and Royall, 1999; Pohjasvaara et al., structures and thalamus.
2002). To address this issue, at least two VaD clinical trials, 2. Anterior cerebral artery (ACA) territory and medial
one of donepezil in CADASIL – a genetic, predominantly frontal lobe lesions, often from an anterior communi-
subcortical form of VaD (Dichgans et al., 2008; Chabriat cating artery aneurysm rupture.
et al., 2009), and in VaD (Román et al., 2010), incorpo- 3. Basal ganglia and thalamus, usually from small vessel
rated tests such as the vascular equivalent of the ADAS-cog disease; this is the so-called ‘thalamic dementia of
called the Vascular-Alzheimer’s Disease Assessment Scale- vascular origin’ caused by butterfly-shaped bilateral
cognitive subscale (V-ADAS-cog) (Ferris and Gauthier, paramedian thalamic artery polar infarcts. Some cases
2002), as well as other tests that include formal measurement of caudate stroke may also result in VaD.
of executive function. These include CLOX, an executive
clock drawing task (Royall et al., 1998) and the EXIT25, an Numerous reports have confirmed the occurrence of
executive interview (Royall et al., 1992). ADLs are consid- true ‘lacunar dementia,’ i.e. acute VaD resulting from a
ered a proxy evaluation of executive function. Pohjasvaara single lacunar stroke involving the inferior genu of the
et al. (2002) confirmed that executive dysfunction was the internal capsule and causing ipsilateral blood flow reduc-
main determinant of abnormalities in both basic and IADLs tion to the inferomedial frontal cortex by a mechanism of
in patients with post-stroke VaD. diaschisis (Tatemichi et al., 1992, 1995; Chukwudelunzu
Recent clinical trials in dementia have relied on brain et al., 2001). This thalamo-cortical disconnection syndrome
imaging end points such as quantification of cortical atrophy is manifested by sudden change in cognitive function, often
over time (de Jager et al., 2012). In comparison with neuropsy- associated with fluctuating attention, memory loss, confu-
chological end points, brain imaging methods in dementia sion, abulia, striking psychomotor retardation, inattention
trials are much more sensitive to change and allow a decrease and other features of frontal lobe dysfunction but with mild
in the sample size for RCTs (Román and Pascual, 2012). focal findings such as hemiparesis or dysarthria.
For instance, Douaud et al. (2013) in the Oxford Project to
Investigate Memory and Ageing (OPTIMA) trial used mag- 60.4.2 SUBACUTE–SUBCORTICAL FORMS
netic resonance imaging (MRI) scans with optimized voxel- OF VaD
based morphometry in 80 subjects with MCI (Petersen et al.,
1999; Petersen, 2004) compared with 76 subjects on placebo. Subcortical ischaemic vascular dementia (SIVD) (Román
After 24 months of treatment, B-group vitamins (the active et al., 2002) is the prototype of subacute VaD. This frequent
arm) resulted in significant decrease of the rate of grey matter clinical form is defined clinically by insidious onset of sub-
atrophy resulting from neurodegeneration. Compared with cortical dementia with frontal lobe deficits, depressive mood
the usual trials in dementia, the sample size reduction in changes – the so-called ‘vascular depression’ (Alexopoulos
this trial was one order of magnitude. Thus, the use of brain et al., 2002), along with psychomotor slowing, parkinsonian
imaging end points could decrease substantially the cost of features, urinary disturbances and pseudobulbar palsy, usu-
controlled clinical trials in VaD and other forms of VCD. ally resulting from small vessel disease (Román et al., 2002).
Symptoms are due to interruption by ischaemic lesions
of prefrontal–subcortical circuits for executive control of
working memory, organization, language, mood, regulation
60.4 CLINICAL FORMS OF VaD of attention, constructional skills, motivation and socially
responsive behaviours (Cummings, 1993). Prefrontal–
Despite the anatomical and pathophysiological complexity subcortical loops may be severed by lacunes in the striatum,
of VaD, Román (1999a, 2002b) has simplified the diverse by globus pallidus or thalamus or by white matter lesions
clinical forms into two main groups, acute and subacute, that disconnect the prefrontal or the anterior cingulate cor-
according to the temporal profile of presentation. tex from their basal ganglia or thalamo-cortical connections
(Swartz et al., 2003; Vataja et al., 2003).
60.4.1 ACUTE FORMS OF VaD Loss of executive function is a major component of VaD
(Román and Royall, 1999) leading to cognitive disability
Acute VaD is defined by new onset of dementia after a and clinically significant loss of planning capacity, working
demonstrated stroke, either from a single ‘strategic stroke’ memory, attention, concentration, discrimination, abstrac-
resulting from occlusion (or rupture) of a large-sized ves- tion, conceptual flexibility and self-control (Román and
sel, following a symptomatic lacunar stroke caused by Royall, 1999; Vataja et al., 2003). Some patients are unable
ischaemic small vessel disease, or from recurrent strokes to initiate the required behaviour, others fail to inhibit
666 Dementia

irrelevant behaviours and most can perform individual dementia was uncommon or mild. Population-based stud-
steps of a complex problem but cannot provide a correct ies of elderly subjects indicate that in the vast majority of
strategy to solve it. These symptoms often lead to post- cases lacunes are silent (Longstreth et al., 1998); however,
stroke depression and functional disability (Pohjasvaara silent lacunes particularly in the thalamus cause more than
et al., 1998, 2002). double the risk of dementia (hazard ratio [HR] 2.26, 95%
The main clinical forms of subacute VaD are BD (état confidence interval [CI] 1.09–4.70) (Vermeer et al., 2003).
lacunaire), CADASIL and rare instances of CAA, including Hospital-based series of lacunar infarctions show that
those that present with leukoencephalopathy (Gray et al., about 30% of these patients develop dementia (Babikian
1985; Sarazin et al., 2002). The temporal profile of presenta- et al., 1990; Tatemichi et al., 1993). In a clinicopathological
tion of these conditions is typically subacute, with a chronic review of 100 consecutive autopsies of patients with lacu-
course marked by fluctuations and progressive worsening. nar strokes at the Hôpital de la Salpêtrière (Román, 1985b),
72 cases with multiple lacunes were found, but only 36%
60.4.2.1 Binswanger disease were clinically demented (état lacunaire). In six cases AD
and lacunes coexisted and an additional six patients had BD
In 1894, Otto Binswanger described a disease characterized (Mikol, 1968). In the demented patients, dilatation of the
by the presence of ischaemic periventricular leukoencepha- lateral ventricles was the only morphological feature that
lopathy typically sparing the arcuate subcortical U-fibres separated état lacunaire patients from cases with multiple
(Binswanger, 1894; Román, 1987). Brain imaging (com- lacunes but without dementia, suggesting that either white
puted tomography [CT] and MRI) usually correlates well matter disease or normal pressure hydrocephalus could
with the pathological features that include diffuse myelin have contributed to dementia (Román, 1991).
pallor and ventricular dilatation. Microscopically there is The Secondary Prevention of Small Subcortical Strokes
loss of myelin and axons resulting in rarefaction, spongio- (SPS3) trial was a large multicentre international study con-
sis and vacuolization without definite necrosis (incomplete ducted in relatively young patients (mean age 63 ± 11 years)
white matter infarction); there is loss of oligodendrocytes with acute lacunar stroke causing no major functional defi-
and astrocytic gliosis with clasmatodendrosis (Sahlas et al., cit or cognitive loss (Benavente et al., 2011, 2014). Of the 2916
2002), axonal thinning, demyelination, gliosis at the dorso- participants recruited, 56% were English-speaking from the
lateral angles of the frontal horns, along with loss of epen- United States or Canada, 40% were Spanish-speaking from
dymal lining (ependymitis granularis) allowing leakage of Latin America, Spain and the United States and 4% were
ventricular cerebrospinal fluid (CSF) (Black et al., 2009). French-speaking Canadians (White et al., 2013). Cognitive
Moody et al. (1995) found that a non-inflammatory, peri- function was determined by the Cognitive Abilities
ventricular venulopathy with concentric collagen deposi- Screening Instrument (CASI) (Teng et al., 1994; McCurry
tion causing intramural thickening, stenosis and ultimately et al., 1999); participants were tested annually for up
venous luminal occlusion could be relevant in the patho- to 5 years. The SPS3 trial showed that a treatment that
genesis. This periventricular occlusive venous collagenosis reduces systolic blood pressure (BP) (<130 mm Hg) lowers
increases markedly with age (65% after age 65 years and the risk of recurrent stroke compared with a higher target
older) and correlates with the severity of the periventricular (130–149 mm Hg) (The SPS3 Study Group, 2013; Pearce
leukoencephalopathy. In addition, small vessel disease with et al., 2014); the addition of clopidogrel to aspirin (ASA)
arteriolosclerosis and other hypertensive lesions (Lammie, increased bleeding risk and was not beneficial compared
2002), and état criblé or widening of perivascular spaces are with ASA alone (The SPS3 Investigators, 2012). MCI was
present. Advanced white matter lesions include focal necro- found in 46% of participants at study entry (Jacova et al.,
sis and cavitations (lacunes); there are also lacunar infarcts 2012, 2015); of the remaining, 27% developed MCI in the
in the basal ganglia and the pons. Román (1985a) suggested absence of a stroke during the study: 6% amnestic, 19%
the identity of BD and a remarkably similar clinical condi- non-amnestic and 1% multidomain (Pearce et al., 2014).
tion, état lacunaire (Marie, 1901), characterized by multiple Cognitive decline beyond age-expected changes over a
lacunes, ventricular dilatation and lesions of the white mat- median follow-up of 3 years was not observed in the entire
ter with a moth-eaten appearance. The syndrome included group and cognition was not affected by short-term dual
mild residual hemiparesis, a peculiar gait with short steps antiplatelet treatment or BP reduction (Pearce et al., 2014).
(marche à petits pas), dysarthria, pseudobulbar palsy and Neuropathological studies show that as many as 60% of
extrapyramidal features such as inexpressive facies, slow- patients with AD have Binswanger-type white matter lesions
ness of movement, axial rigidity, loss of postural reflexes (Brun and Englund, 1988; Englund et al., 1988). These
and frequent falls. lesions have been postulated as being caused by cerebral
Dementia is not a salient feature of état lacunaire (Marie, hypoperfusion resulting from cardiac disease or orthostatic
1901), but some intellectual deficit is constantly present. hypotension; this form could be called AD with Binswanger-
However, Fisher (1965) remarked that lacunar strokes typi- type lesions (AD+BD). Moreover, Snowdon et al. (1997)
cally do not cause disturbances in higher cerebral func- in the Nun Study reported that in very old subjects lacu-
tions, and when present ‘always exclude a lacunar diagnosis’. nes are an important factor in the clinical expression of
Among 114 patients with lacunes he examined at necropsy, AD. This was confirmed in the Religious Orders Study
Therapeutic strategies for vascular cognitive disorders 667

(Schneider et al., 2009), the Honolulu-Asian Aging Study embryonic development. The large extracellular domain of
(Petrovitch et al., 2005) and the Medical Research Council Notch3 contains 34 tandem epidermal growth factor (EGF)-
Cognitive Function and Ageing Study (MRC-CFAS) like repeats where the mutations result in gain or loss of a
(Matthews et al., 2009). Likewise, according to Jellinger and cysteine residue.
Mitter-Ferstl (2003), the presence of cerebrovascular pathol- Other rare genetic forms of VCD include: cerebral
ogy in patients with AD is significantly higher at autopsy autosomal recessive arteriopathy with subcortical autoso-
than in controls (48% versus 7.4%). On post-mortem exami- mal recessive leukoencephalopathy (CARASIL); heredi-
nation, Zekry et al. (2002b, 2002c) demonstrated the syner- tary endotheliopathy retinopathy, nephropathy and stroke
gistic effect of vascular lesions in patients with AD dementia. (HERNS); pontine autosomal dominant microangiopathy
Presence of vascular lesions had a direct correlation with and leukoencephalopathy (PADMAL); retinal vasculopathy
dementia severity. Furthermore, the density of AD lesions with cerebral leukodystrophy (RVCL) and, collagen type IV,
(neuritic plaques, focal amyloid β deposits and neurofibril- α1 (COL4A1)-related disorders (Leblanc et al., 2006).
lary tangles) was significantly lower when vascular lesions
were present. 60.4.2.3 Cerebral amyloid angiopathy

60.4.2.2 CADASIL CAA is a heterogeneous group of disorders characterized


by amyloid deposition in the walls of leptomeningeal and
CADASIL was described in France in the early 1990s cerebral cortical blood vessels, manifested clinically by
(Bousser and Tournier-Lasserve, 1994). This condition was recurrent lobar haemorrhages, cognitive deterioration and
originally called ‘familial Binswanger’ (van Bogaert, 1955) ischaemic strokes. Cerebral MRI displays diffuse white
and ‘hereditary MID’ (Sourander and Walinder, 1977; matter abnormalities along with ischemic or haemorrhagic
Sonninen and Savontaus, 1987). CADASIL appears to be focal brain lesions. On histology, the vessels show amyloid
the most common genetic form of VaD (Dichgans, 2002; deposition, microaneurysms and fibrinoid necrosis.
Kalimo et al., 2002; Chabriat and Bousser, 2003; Chabriat There are several autosomal dominant forms of CAA
et al., 2009). This autosomal dominant disorder of cere- with differences in their clinical, genetic, biochemical and
bral small vessels maps to chromosome 19 and is caused by pathologic findings. A-β, the major amyloid component in
Notch3 gene mutations. The underlying vascular lesion is a the Dutch-, Flemish- and Iowa-type of familial CAA, is also
unique non-amyloid non-atherosclerotic microangiopathy the major amyloid component in sporadic CAA and in AD.
involving arterioles (100–400 μm in diameter) and capil- Familial British dementia with amyloid angiopathy (FBD)
laries, primarily in the brain but also in other organs. The is an autosomal dominant condition characterized by VaD,
diagnosis may be established by skin biopsy (Ruchoux et al., progressive spastic paraparesis and cerebellar ataxia with
1995) with confirmation by immunostaining using a Notch3 onset in the sixth decade (Mead et al., 2000). A point muta-
monoclonal antibody. tion in the BRI gene is the genetic abnormality. On brain
CADASIL offers a natural model for the study of MRI, Binswanger-type deep white matter hyperintensities
subcortical–subacute forms of VaD, in particular BD. and lacunar infarcts are seen, but without intracerebral
Numerous pedigrees have been described in Europe and haemorrhages. The corpus callosum is severely affected and
North America. Clinical manifestations include transient atrophic. Plaques and tangles are present but the amyloid
ischaemic attacks (TIAs) and strokes (80%); cognitive defi- subunit (ABri) found in FBD brains is entirely different
cits, apathy and VaD (50%); migraine with focal deficits and unrelated to other amyloid proteins. FBD combines
(40%); mood disorders (30%) and epilepsy (10%). Onset neurodegeneration and dementia with systemic amyloid
is usually in early adulthood (mean age: 46 years) in the deposition.
absence of risk factors for vascular disease, culminating in
dementia and death usually about 20 years after symptom
onset. The dementia is slow in onset, subcortical, frontal in
type, accompanied by gait and urinary disturbances and 60.5 RISK FACTORS FOR VaD
pseudobulbar palsy clinically identical to that of sporadic
BD. MRI reveals a combination of small lacunar lesions and A number of case–control studies of PSD have provided
diffuse white matter abnormalities, including typical lesions quantification of risk factors for VaD (Tatemichi et al., 1990;
of the corpus callosum and tip of the temporal lobes; MRI Pohjasvaara et al., 1997; Barba et al., 2001; Leys et al., 2005;
lesions are often present in asymptomatic relatives. Also, Brainin et al., 2015). In the Framingham study, the occur-
cerebral blood flow (CBF) reactivity to inhaled carbon diox- rence of stroke doubled the risk of dementia (Ivan et al.,
ide (CO2) is impaired. Widespread smooth muscle cell loss 2004); the prevalence of PSD in stroke survivors is about
results in decreased regional cerebral blood volume (CBV); 30% and the incidence of new-onset dementia after stroke
these values correlate inversely with cognitive performance. increases from 7% after 1 year to 48% after 25 years (Leys
The disease is caused by highly stereotyped mutations in the et al., 2005). In patients with ischaemic stroke, the most
Notch3 gene that encodes a phyllogenetically old transmem- important risk factors for the development of PSD are older
brane cell surface receptor that regulates cell fate during age, lower educational level and loss of independence or
668 Dementia

cognitive decline prior to stroke, as well as recurrent stroke, smoking (PAR = 16.7%), diabetes mellitus (PAR = 7.7%),
presence of dysphagia, gait limitations and urinary impair- excessive alcohol drinking (PAR = 5.4%) and atrial fibril-
ment. The left-sided predominance of lesions in demented lation (PAR = 2%). The seven potentially modifiable vas-
patients with large vessel stroke in the territories of the cular risk factors include diabetes, midlife hypertension,
anterior and posterior cerebral arteries is well established midlife obesity, smoking, depression, cognitive inactivity or
(Leys and Pasquier, 2000). Also important are silent cere- low educational attainment and physical inactivity (Barnes
bral infarcts, and global and medial temporal lobe atrophy. and Yaffe, 2011; Norton et al., 2014). As noted, at least one
Less often considered are obstructive sleep apnoea, hyper- population-based controlled trial in Finland (Ngandu et al.,
homocysteinaemia and mutations of the methylenetetrahy- 2015) demonstrated positive cognitive effects after 2 years
drofolate reductase (MTHFR) gene (Román, 2015). of a multidomain intervention that included diet, exercise,
Other risk factors include diabetes mellitus (Pasquier cognitive training and vascular risk monitoring.
et al., 2006; Umegaki, 2010), atrial fibrillation (Lefebvre A brief review of the main results of antihypertensive tri-
et al., 2006), as well as conditions resulting in cerebral hypo- als and dementia now follows.
perfusion (Román, 2004) (e.g. myocardial infarction, car-
diac arrhythmias and congestive heart failure) (Jefferson 60.5.1 HYPERTENSION AND
et al., 2015a, 2015b). Johnston et al. (2004) observed progres- PREHYPERTENSION
sive decline in cognitive function among Cardiovascular
Health Study participants with stenosis of the left internal Several prospective studies have shown that hypertension
carotid artery (ICA), but not in those with stenosis of the increases dementia risk (Tzourio et al., 1999; Birkenhäger
right ICA, in the absence of stroke. Possible explanations and Staessen, 2006; Nagai et al., 2010, 2012). Also, disrup-
include the fact that the left hemisphere has larger choliner- tion of diurnal BP variation is closely associated with cogni-
gic innervation than the right one and that most cognitive tive impairment (Sakakura et al., 2007; Nagai et al., 2008).
tests rely heavily on language functions. Therefore, there is heightened interest in the diagnosis and
Other risk factors for PSD include current smoking, treatment of the large population with prehypertension,
lower BP, orthostatic hypotension and larger periventricu- defined as BP in the range of >120–139 mm Hg systolic or
lar white matter ischaemic lesions by brain CT or MRI (Liu >80–89 mm Hg diastolic (Schunkert, 2006). In the Trial of
et al., 1991; Gorelick et al., 1992). Orthostatic hypotension Preventing Hypertension (TROPHY), Julius et al. (2006)
is an important factor in VaD (Román, 2004) and a strong studied the use of the angiotensin receptor blocker cande-
predictor of periventricular white matter ischaemic lesions sartan or placebo for 2 years in persons with prehyperten-
(Longstreth et al., 1996). Hypoxic and ischaemic complica- sion (first phase). Then, all participants received placebo
tions of acute stroke (e.g. cardiac arrhythmias, aspiration for another 2 years (second phase). TROPHY showed that
pneumonia, sepsis, etc.) are strong and independent risk after 2 years of active treatment, candesartan may interfere
factors for PSD (Moroney et al., 1996); these complications with development of hypertension and prevent target organ
increase more than fourfold the risk of developing PSD damage with a RR reduction of 15.6% for the development
(odds ratio [OR] 4.3, 95% CI 1.9–9.6, after adjustment for of hypertension after 4 years. Schunkert (2006) noted that
demographic factors, recurrent stroke and baseline cogni- most people with prehypertension have additional risk fac-
tive function). Seizures following stroke (Cordonnier et al., tors including obesity, and most of the complications asso-
2007) are independent predictors of new-onset dementia ciated with prehypertension result from these cofactors
within 3 years after stroke (HR 3.81, 95% CI 1.13–12.82). including diabetes mellitus and chronic kidney disease. The
Pendlebury and Rothwell (2009) reviewed 21 hospital-based JNC7 guidelines recommended antihypertensive treatment
and 8 population-based cohorts (7511 patients) described in of such patients if a trial of lifestyle modification failed to
73 articles and concluded that PSD is associated with mul- reduce BP below 130/80 mm Hg (Chobanian et al., 2003).
tiple strokes rather than with underlying vascular risk fac- More recent experience indicates that there is insufficient
tors. However, as noted by Hennerici (2009), most studies information to recommend drug therapy for prehyperten-
were from the 1970s and 1980s, emphasizing the concept sion (Marta et al., 2014).
of MID and stroke recurrences rather than distinct mecha- Shah et al. (2009) reviewed the role of antihypertensive
nisms of stroke causation. drugs on dementia incidence and progression. These authors
PSD is a strong predictor of poor outcome (Desmond et al., analysed 536 publications from 1999 to 2008 that included
1998; Barba et al., 2001; Linden et al., 2004; Leys et al., 2005). In calcium channel blockers (CCBs), diuretics and angiotensin-
Rochester, Minnessota, 10% of 482 incident cases of dementia converting enzyme inhibitors (ACE-I). All three drug types
had onset or worsening of the dementia within 3 months of a appeared to be beneficial in dementia, but only ACE-I plus
stroke (Knopman et al., 2003). Overall, patients with VaD had diuretics showed a significant reduction of dementia risk and
worse mortality than matched subjects (relative risk [RR] 2.7), progression. A meta-analysis of the Systolic Hypertension
particularly among cases of PSD (RR 4.5). in the Elderly Program (SHEP), Systolic Hypertension in
The main correctable population-attributable risk (PAR) Europe (Syst-Eur) and Study on Cognition and Prognosis in
factors (Nyenhuis and Gorelick, 2007) include hyperten- the Elderly (SCOPE) trials (McGuinness et al., 2009) showed
sion (PAR = 65%), hypercholesterolemia (PAR = 36.5%), that antihypertensive therapy resulted in non-statistically
Therapeutic strategies for vascular cognitive disorders 669

significant reduction of the risk of dementia by 11% (OR Study on Cognitive function And systolic blood pressure
0.89, 95%CI 0.69–1.16). Statistically significant reduction (SBP) Reduction (OSCAR) that lowering of SBP by the ARB
of the risk of dementia (HR 0.87, 95%CI 0.76–1.00) with eprosartan slowed cognitive decline (OR 0.77, 95% CI 0.73–
BP control was found in a meta-analysis of the SHEP, Syst- 0.82). According to Fogari et al. (2006), antihypertensive
Eur, Perindopril Protection Against Recurrent Stroke Study therapy with an ARB (telmisartan) plus HCTZ showed sig-
(PROGRESS) and Hypertension in the Very Elderly Trial nificant improvement in cognitive function compared with
(HYVET) trials (Peters et al., 2008). As mentioned, in SPS3, ACE-I plus diuretic (lisinopril and HCTZ). ARBs have been
strict control of BP post-lacunar stroke produced no change shown to reduce stroke risk, in part independently of their
in cognitive decline beyond age-expected changes over a BP-lowering effect (Diener, 2009).
median follow-up of 3 years; dual antiplatelet treatment did
not improve the prognosis (Pearce et al., 2014). 60.5.2 VASCULAR CARE AND STROKE
PREVENTION IN PATIENTS WITH
60.5.1.1 Diuretics DEMENTIA
Although diuretics remain the mainstay of hypertension treat- Richard et al. (2010) in the Evaluation of Vascular Care in
ment (Prince, 1997; Chobanian et al., 2003), there were no AD (EVA) study demonstrated that improved vascular care
differences in cognition in those treated with diuretics alone in patients with AD+CVD slows the progression of white
versus controls in the Medical Research Council’s (MRC) matter lesions on MRI. There was no difference in the num-
study (Prince et al., 1996), in the SHEP cohort (Applegate ber of new lacunes or change in global cortical atrophy or
et al., 1994) nor in Hypertension in the Very Elderly Trial medial temporal lobe atrophy between the standard care
Cognitive Function Assessment (HYVET-cog) trial (Peters and the improved vascular care group.
et al., 2008). However, at population level, in the Cache County Savva and Stephan (2010) have emphasized the need for
Study (Kachaturian et al., 2006) potassium-sparing diuretics stroke prevention given that a history of stroke doubles the
reduced (HR 0.26, 95% CI 0.08–0.64) the incidence of AD. risk of incident dementia in older populations. This increase
is not explained by demographic or cardiovascular risk fac-
60.5.1.2 Calcium channel blockers tors or by pre-stroke cognitive decline. The excess risk of
incident dementia diminishes with time after stroke and
In the Syst-Eur trial (Forette et al., 1998), treatment with the may be higher in those without an apolipoprotein E ε4
CCB nitrendipine decreased 42% the primary end point of fatal (APOEε4) allele. There is no excess risk of incident dementia
and non-fatal stroke. Of 2418 participants in the dementia in those aged ≥85 years with a history of stroke. The effect
sub-study, 60% of the treatment group received nitrendipine of stroke on dementia incidence in the population is not
alone; the rest also had enalapril and/or a hydrochlorothiazide explained by common risk factors.
(HCTZ). Compared with the controls, antihypertensive ther- Staekenborg et al. (2009) showed that patients with MCI
apy reduced the risk of dementia by 55% (Forette et al., 2002). who converted to VaD were older and had lower baseline
MMSE than non-converters, and on baseline MRI showed
60.5.1.3 Angiotensin-converting enzyme more severe white matter hyperintensities, higher preva-
inhibitors lence of lacunes in the basal ganglia and microbleeds. Deep
white matter lesions (HR = 5.7, 95%CI 1.2–26.7), periven-
In PROGRESS trial (Tzourio et al., 1999, 2003), perindopril tricular hyperintensities (HR = 6.5, 95%CI 1.4–29.8) and
plus a diuretic (indapamide) – but not ACE-I alone – caused microbleeds (HR = 2.6, 95% CI 0.9–7.5) increased risk of
a RR reduction of dementia of 23% (95% CI 0–41%). In the progression to VaD. Medial temporal lobe atrophy and
Heart Outcomes Prevention Evaluation (HOPE) study, markers of CVD predict the development of different types
ramipril compared with placebo significantly reduced (RR of dementia in MCI. It is important to control all vascular
0.59, 95%CI 0.37–0.94) the rate of cognitive decline (Bosch risk factors in patients with vascular MCI, particularly when
et al., 2002). MRI brain imaging shows white matter hyperintensities,
lacunes in the basal ganglia and microbleeds. Homocysteine
60.5.1.4 Angiotensin II receptor blockers may play a role in the conversion from MCI to AD.
(ARBs)
In SCOPE trial of candesartan versus placebo (Lithell et al., 60.6 PHARMACOLOGICAL THERAPY
2003) and in Prevention Regimen for Effectively Avoiding
OF VaD
Second Strokes (PRoFESS), a trial of telmisartan versus pla-
cebo with over 20,332 subjects (Diener et al., 2008), there
were no significant differences in the rate of cognitive decline A review of the history of VaD (Román, 2002c, 2002d) sug-
or dementia with either one of the two ARBs compared gests that the prevailing concepts of pathogenesis at a given
with placebo. Hanon et al. (2008) reported in Observational time influence not only the elaboration of clinical criteria but
670 Dementia

also the approach to therapy. Until the 1970s, it was believed 2007). Selden et al. (1998) described in the human brain two
that the cause of VaD was a progressive strangulation of blood highly organized and discrete bundles of cholinergic fibres
flow to the brain resulting in chronic cerebral hypoperfusion. extending from the nucleus basalis of Meynert (nbM) to the
Therefore, beginning with nicotinic acid, a number of vaso- cerebral cortex and amygdala. Mesulam et al. (2003) dem-
dilating agents were recommended for the treatment of non- onstrated cholinergic denervation from ischaemic pathway
specific senile symptoms, cognitive decline and dementia. lesions in CADASIL, a pure genetic form of VaD, entirely
Following the same logic, some authors (Walsh, 1968; Walsh unmixed with AD lesions.
et al., 1978) claimed significant improvement of symptoms Cholinergic mechanisms play a role in the modula-
of presenile and senile dementia with a combination of war- tion of regional CBF (Sato et al., 2001). Stimulation of the
farin anticoagulation and psychotherapy. A vascular patho- nbM results in increased blood flow in the cerebral cortex
genesis mediated by recurrent multiple strokes (multi-infarct (Biesold et al., 1989). This cholinergic vasodilatory system
dementia) also underlay the use of antithrombotic agents, relies upon activation of both muscarinic and nicotinic
ergot alkaloids, nootropics, thyrotropin-releasing hormone cholinergic receptors and the response declines with age
(TRH) analogue, Ginkgo biloba extract, plasma viscosity (Lacombe et al., 1997; Uchida et al., 2000). Therefore, there
drugs, hyperbaric oxygen, antioxidants, serotonin and his- is loss of cholinergic function in patients with VaD and this
tamine receptor antagonists, vasoactive agents, xanthine is associated with reductions in CBF (Court et al., 2002).
derivates and calcium antagonists (Román, 2000). With These observations provide reasonable arguments to jus-
increasing recognition of AD and new knowledge of neu- tify the use of ChEIs in patients with VaD, both unmixed
rotransmitters, neuronal metabolism and biochemical tissue with AD, as well as in patients with AD+CVD (Zekry and
reactions to ischaemia-hypoxia and oxidative stress, more Gold, 2010). Three of the ChEIs approved for use in AD –
specific medications were developed leading to neuroprotec- donepezil, galantamine and rivastigmine – have been stud-
tion (Mondadori, 1993) and neurorepair; including agents ied also in VaD.
that may have effects similar to those of stem cells, such as
citicoline (Álvarez-Sabín and Román, 2013). The most recent
advance in the treatment of VaD has been the development
60.6.1.1 Donepezil
of medications to enhance specific neurotransmitters, such Donepezil hydrochloride is a reversible, non-competitive
as acetylcholine (ACh). Furthermore, these products have ChEI with highly selective central action, approved for the
been studied by strict controlled clinical trials. The ChEIs treatment of mild, moderate and advanced AD (see Chapter
will be discussed first, followed by memantine, CCBs, noo- 52). Donepezil is unrelated to other ChEIs and has greater
tropic agents, xanthines, ergot derivatives, vasodilators and selectivity for cerebral acetylcholinesterase (AChE) than for
then other treatments for VaD. butirylcholinesterase (BuChE).
With the demonstration of reduced regional CBF in late- The safety and efficacy of donepezil have been studied
life dementias (Román, 2008a), the pendulum has returned in three of the largest (N = 2193) clinical trials of pure VaD
to the concept of brain hypoperfusion as a critical element in (Goldsmith and Scott, 2003; Román et al., 2005; Román et al.,
dementia pathogenesis. This has led to a proposal to treat VaD 2010). A total of 1219 subjects were recruited for a 24-week,
using sphenopalatine ganglion (SPG) stimulation to augment randomized, placebo-controlled, multicentre, multinational
CBF (Henninger and Fisher, 2007; Khurana et al., 2009). study, which is divided into two identical trials comprising
307 (Black et al., 2003) and 308 (Wilkinson et al., 2003). The
60.6.1 CHOLINERGIC DYSFUNCTION IN patients were randomized to one of three groups: placebo,
VaD donepezil at a dosage of 5 mg/day or donepezil, 10 mg/day.
The group receiving 10 mg/day initially received 5 mg/day for
Cholinergic dysfunction is well documented in VaD, 4 weeks; the dosage was then titrated up to 10 mg/day. Most
independently of any concomitant AD pathology; these patients (73%) fulfilled diagnosis of probable VaD accord-
deficits consist of decreased levels of ACh in the CSF and ing to the NINDS-AIREN criteria (Román et al., 1993). All
reduced cholinergic markers such as choline acetyltrans- had brain imaging prior to enrolment (CT or MRI) with
ferase (ChAT) in the brain (Szilagyi et al., 1987; Wallin demonstration of cerebrovascular lesions. Patients with
et al., 1989; Wallin and Gottfries, 1990; Gottfries et al., 1994; pre-existing AD and those with ‘mixed dementia’ (AD plus
Tohgi et al., 1996; Wang et al., 2009). Hippocampal ChAT CVD) were excluded. Exclusion criteria also included other
deficits of up to 60% were reported in the brains of both AD neurodegenerative or psychiatric disorders (e.g. schizo-
and VaD patients (Perry et al., 1977; Sakurada et al., 1990; phrenia, major depression), an MMSE (Folstein et al., 1975)
Kalaria et al., 2001). The cholinergic basal forebrain nuclei score >26 or <10, the occurrence of new strokes within the
are irrigated by penetrating arterioles and are therefore sus- 28 days prior to baseline or myocardial infarction within 3
ceptible to the effects of arterial hypertension (Román and months of enrolment. These two VaD studies enrolled more
Kalaria, 2006). Moreover, ischaemic lesions in the white men than women (58% versus 38%) and their mean age
matter and the basal ganglia can interrupt the cholinergic was older than in AD (74.5 ± 0.2 versus 72 ± 0.2 years). The
projections (Swartz et al., 2003; Bocti et al., 2005; Behl et al., more severe vascular pathology of VaD was reflected in the
Therapeutic strategies for vascular cognitive disorders 671

high HIS score (6.6 ± 0.2 versus <4 in AD); most subjects hippocampal size. Donepezil was relatively well tolerated;
had hypertension, hypercholesterolemia, cardiovascular dis- AEs were consistent with current labelling. Mortality in the
ease, diabetes, smoking, previous stroke and TIAs. The third placebo group was unexpectedly low (Román et al., 2010).
study (319 trial) recruited 974 subjects (mean age 73.0 years) In comparison with AD patients, cognitive decline in
with probable or possible VaD that were randomized 2:1 to untreated VaD patients during 24 weeks of study in these
receive donepezil 5 mg/day or placebo. Coprimary outcome trials was less severe, using similar instruments. These dif-
measures were the V-ADAS-cog (Ferris and Gauthier, 2002) ferences were also noted for global effects, measured by
and the CIBIC-Plus (Schneider et al., 1997). the CIBIC-plus. Substantial decline of IADLs was noted in
There were three main end points: (1) cognition, mea- untreated VaD patients. A combined analysis of the total
sured with the ADAS-cog (Rosen et al., 1984) and the population confirmed the effects on cognition and global
MMSE; (2) global function, evaluated with the CIBIC-plus function. Donepezil-treated patients demonstrated statis-
and with the sum of boxes of the Clinical Dementia Rating tically significant improvements in IADLs compared with
(CDR-SB) (Berg, 1988) and (3) ADLs, measured with the decline in the placebo group (LS mean change from base-
ADFACS (Mohs et al., 2001). line at end point: placebo 0.60; 5 mg/day −0.09, p < 0.05;
In study 307 (Black et al., 2003), both donepezil treatment 10 mg/day −0.18, p < 0.01). These results suggest that donepe-
groups showed statistically significant cognitive improve- zil treatment may improve or maintain the patients’ abilities
ment measured with the MMSE and the ADAS-cog; the mean to perform ADLs. Executive dysfunction is a major compo-
changes from baseline ADAS-cog scores were the following: nent of disability in patients with VaD and the beneficial
donepezil 5 mg/day −1.90 (p = 0.001); donepezil 10 mg/day effect of donepezil on IADLs may be related to improvement
−2.33 (p < 0.001). The donepezil 5 mg/day group showed sig- or stabilization of executive function.
nificant improvement in global function on the CIBIC-plus but Donepezil was generally well tolerated, although more
did not reach significance in the 10 mg/day group (p = 0.27). The adverse effects were reported in the 10 mg/day group than
CDR-SB showed non-significant benefit in the 5 mg/day group in the 5 mg/day or placebo groups. The adverse effects were
and statistical significance in the 10 mg/day group (p = 0.022). assessed as mild to moderate and transient, and were typi-
Compared with placebo, there was significant benefit in ADLs cally diarrhoea, nausea, arthralgia, leg cramps, anorexia
(p < 0.05) in both donepezil groups using the ADFACS. and headache. The incidence of bradycardia and syncope
Similar results were observed in study 308 (Wilkinson et al., was not significantly different from the placebo group. The
2003); donepezil treatment resulted in significant improvement discontinuation rates were 15% for placebo, 18% for 5 mg
in cognition measured with the MMSE and the ADAS-cog. For and 28% for the 10 mg group. There was no significant inter-
the latter, the mean changes from baseline score were the fol- action with cardiovascular medications or antithrombotic
lowing: donepezil 5 mg/day −1.65 (p = 0.003); donepezil 10 agents. These three large trials offer evidence that donepezil
mg/day −2.09 (p = 0.0002). Global function on the CIBIC-plus is effective and well tolerated in VaD patients.
was significantly better in the 5 mg/day group (p = 0.004) and Dichgans et al. (2008) conducted a trial of donepezil
was barely below significance in 10 mg/day group (p = 0.047). in patients with CADASIL that randomized 168 patients
The CDR-SB showed benefits with 5 mg/day (non-significant), (mean age 54.8 years) to 10 mg donepezil per day (n = 86) or
and it reached significance with 10 mg/day (p = 0.03). ADLs placebo (n = 82). Inclusion criteria included an MMSE score
improved in patients treated with donepezil when compared of 10–27 or a Trail Making Test (TMT) B time score at least
with placebo using the ADFACS, but did not reach significance 1.5 SD below the mean, after adjustment for age and edu-
at the end of the study. Comparison of cortical versus subcorti- cation. The primary end point was the V-ADAS-cog at 18
cal forms of VaD showed no differences in the overall results weeks. Secondary end points included ADAS-cog, MMSE,
(Salloway et al., 2003). TMT A and B times, Stroop test, EXIT25, CLOX, DAD and
In trial 319 (Román et al., 2010), donepezil-treated CDR sum of boxes. Donepezil (5–10 mg/day) had no effect
patients showed significant improvement from baseline on V-ADAS-cog but had significant effect on two measures
to end point on the V-ADAS-cog (least squares [LS] mean of executive function: EXIT25 and TMT-B. These subjects
difference −1.156; 95% CI −1.98 to −0.33, p < 0.01) but not were younger than most VaD patients and most (75%) had
on the CIBIC-Plus. Donepezil-treated patients with hip- no clinically significant memory dysfunction. The positive
pocampal atrophy demonstrated stable cognition versus effect of donepezil on executive function, however, indicates
decline in the placebo-treated group; in those without some cholinergic deficit in executive dysfunction.
atrophy, cognition improved with donepezil versus relative
stability with placebo. Results on secondary efficacy mea- 60.6.1.2 Galantamine
sures were inconsistent. The incidence of adverse events
(AEs) was similar across groups. Eleven deaths occurred Galantamine is approved for the treatment of mild-
in the donepezil group (1.7%) – similar to rates previously to-moderate AD. It is a reversible specific inhibitor of AChE,
reported for donepezil trials in VaD – whereas no deaths and in addition it modulates central nicotinic receptors to
occurred in the placebo group. Donepezil 5 mg/day showed increase cholinergic neurotransmission. The safety and
significant improvement in cognitive, but not global, func- efficacy of galantamine in AD have been studied in trials
tion, with differential treatment response of VaD patients by recruiting over 2600 patients (see Chapter 52).
672 Dementia

Birks et al. (2013) conducted a Cochrane review of galan- galantamine 1.9, p = 0.06) (Erkinjuntti et al., 2003a). More
tamine in VCD. Among others (Craig and Birks, 2006), patients treated with galantamine than with placebo main-
galantamine has been studied in VaD in the GAL-INT-6 tained or improved global function (CIBIC-plus, 31% versus
study, a large Phase III, randomized, multicentre, double- 23%, not statistically significant). In these patients, the cog-
blind, placebo-controlled RCT in patients with probable VaD nitive benefits of galantamine were maintained at least up
or with AD combined with CVD (Erkinjuntti et al., 2002a). to 12 months, demonstrating a mean change of −2.1 in the
Patients received galantamine 24 mg/day (n = 396) or placebo ADAS-cog score compared to baseline, and the active group
(n = 196) for 6 months; the age of the patients ranged from 40 was still close to baseline at 24 months (Erkinjuntti et al.,
to 90 years. Eligible subjects met either the clinical criteria of 2003a). During the 12-month trial, the most frequently
probable VaD by NINDS-AIREN criteria (Román et al., 1993) reported AEs were depression (13%), agitation (12%) and
or of possible AD according to the NINCDS-ADRDA crite- insomnia (11%).
ria (McKhann et al., 1984). Radiological evidence of CVD on Erkinjuntti et al. (2008) performed responder analyses
brain CT or MR was required (i.e. AD plus CVD), including for cognitive, behavioural and functional outcome mea-
multiple large vessel infarcts or a single, strategically placed sures in GAL-INT-6. Galantamine treatment resulted in sig-
infarct (angular gyrus, thalamus, basal forebrain, territory of nificant cognitive and functional improvements compared
the PCA or ACA) or at least two basal ganglia and white mat- with placebo at 6 months. Significantly higher percentage
ter lacunes or white matter changes involving at least 25% of treatment responders were found for the ADAS-cog/11
of the total white matter. Mild-to-moderate dementia was (60.5% for galantamine versus 46.0% for placebo, p = 0.013);
defined by scores of ≥12 in the ADAS-cog/11 (Rosen et al., the CIBIC-plus (75% versus 53.6% with placebo, p = 0.0006);
1984) and 10–25 on the MMSE. behaviour (64.9% versus 56.6%, p = 0.024), and numerically
Primary end points were cognition, as measured using favourable responder rates were seen with galantamine for
the ADAS-cog/11 and global functioning as measured ADLs, indicating a broad range of cognitive, functional and
using the CIBIC-plus (Schneider et al., 1997). Secondary behavioural benefits with galantamine across the spectrum
end points included assessments of ADLs, using the DAD of AD and AD+CVD.
(Gélinas et al., 1999), and behavioural symptoms using the In another trial of galantamine in VaD, the GAL-INT-26
Neuropsychiatric Inventory (NPI) (Cummings et al., 1994). study (Auchus et al., 2007), 788 patients with probable VaD
Galantamine demonstrated greater efficacy than placebo on and strict centrally read MRI criteria were randomized to
all outcome measures in analyses of both groups of patients galantamine or placebo. Efficacy was evaluated using mea-
as a whole (Corey-Bloom, 2013): ADAS-cog (2.7 points, sures of cognition, daily function, and behaviour using the
p ≤ 0.001) on the CIBIC-plus (74% versus 59% of patients NPI (Cummings et al., 1994); EXIT-25 was used for the
remained stable or improved, p ≤ 0.001). ADLs and behav- assessment of executive functioning. Safety and tolerability
ioural symptoms in the NPI were also significantly improved were also monitored. Galantamine showed improvement in
compared with placebo (both p < 0.05). Galantamine was ADAS-cog/11 after 26 weeks compared with placebo (−1.8
safe and well tolerated; however, nausea was six times com- versus −0.3; p < 0.001), but there was no difference on the
moner in patients on galantamine than those on placebo. ADCS-ADL score (0.7 versus 1.3; p = 0.783). Improvement
In comparison, nausea was twice as common in patients in global functioning by the CIBIC-plus approached sig-
with AD treated with galantamine compared with placebo- nificance (p = 0.069). In patients with VaD, galantamine
treated patients (Erkinjuntti et al., 2002a), suggesting lesser significantly improved cognition, including executive func-
tolerability in VaD patients. tion, but ADLs were similar to the placebo group (Auchus
In an open-label extension (Erkinjuntti et al., 2003a), the et al., 2007).
original galantamine group of patients with probable VaD In Thai patients, Thavichachart et al. (2006) reported
or AD+CVD showed similar sustained benefits in terms the results of a 6-month trial of galantamine in AD with
of maintenance of or improvement in cognition (ADS- or without CVD and VaD. Galantamine showed favourable
cog), functional ability (DAD), and behaviour (NPI) after effects on ADLs; behavioural symptoms and sleep quality
12 months. Although not designed to detect differences were also significantly improved (p < 0.05). Galantamine
between subgroups, the subgroup of patients with AD+CVD was well tolerated.
on galantamine (N = 188, 48%) showed greater efficacy than In the Cochrane review of galantamine for VaD, Birks
placebo (N = 97, 50%) at 6 months on the ADAS-cog (p ≤ et al. (2013) concluded that there is positive evidence of
0.001) and the CIBIC-plus (p = 0.019) (Erkinjuntti et al., improved cognition and executive functioning in VCDs;
2002a). In the open-label extension, patients with AD+CVD however, galantamine produced higher rates of gastrointes-
on galantamine maintained cognition at baseline for 12 tinal side effects.
months (Erkinjuntti et al., 2003a).
In the subgroup of patients with VaD treated with 60.6.1.3 Rivastigmine
galantamine for 6 months, the ADAS-cog scores improved
significantly (mean change from baseline 2.4 points, Rivastigmine is a slowly reversible AChE inhibitor and
p < 0.0001), compared with no response in placebo group BuChE inhibitor approved for use in mild-to-moderate AD
(mean change from baseline 0.4, treatment difference versus (see Chapter 52). Rivastigmine is metabolized at the synapse
Therapeutic strategies for vascular cognitive disorders 673

rather than by hepatic cytochrome enzymes; hence, there and Machado, 2008). The patch is associated with three
are no drug–drug interactions (Polinsky, 1998). The role of times fewer reports of nausea and vomiting (Winblad and
BuChE inhibition in glial cells in AD remains unclear. After Machado, 2008). The rivastigmine patch has not been evalu-
treatment of AD with rivastigmine for 12 months, CSF and ated in VaD.
plasma AChE activity decreased by 46% and BuChE by 65%
from baseline (Darreh-Shori et al., 2002). There was a sig- 60.6.2 MEMANTINE
nificant correlation between plasma ChE inhibition and
cognition, particularly in relation to attention (Almkvist Memantine is a potent antagonist of the N-methyl-
et al., 2004). d-aspartate (NMDA) receptor (Farlow, 2004) (see Chapter 52).
Rivastigmine was used in small open-label studies of Experimentally, memantine inhibits and reverses the abnor-
patients with subcortical VaD followed for 12 months mal activity of a protein phosphatase (PP-2A) that leads to tau
(Moretti et al., 2001) and 22 months (Moretti et al., 2002); hyperphosphorylation and to neurofibrillary degeneration in
rivastigmine resulted in the stabilization of cognition and AD (Li et al., 2004). Memantine is effective and well toler-
ADLs, with slight improvement of executive function and ated in patients with severe AD (Reisberg et al., 2003) and was
planning, less caregiver stress and improved behaviour. approved by the FDA for the treatment of moderate-to-severe
More recently, Moretti et al. (2008) observed significant AD (Möbius, 2003). Furthermore, Tariot et al. (2004) showed
improvement in behavioural symptoms in two forms of that in patients with moderate-to-severe AD receiving stable
VaD, MID and subcortical VaD, except delusions, suggest- doses of donepezil, memantine resulted in significantly better
ing that rivastigmine helps reduce concomitant neuroleptic outcomes than placebo in cognition, ADLs, global outcome,
and benzodiazepine use. and behaviour. These findings suggest that this dual therapy
Rivastigmine effects in VaD were studied in the VaD could be useful.
trial studying Exelon (VantagE), a Phase III, double-blind, Memantine has been used in patients with VaD based
placebo-controlled RCT (Moretti et al., 2004). This 24-week, on its experimental efficacy in animal models of ischaemic
multicentre study randomized 710 VaD patients aged 50–85 lesions. Memantine acts on potentially contributing factors
years to rivastigmine capsules (3–12 mg/day) or placebo such as neuronal depolarization, mitochondrial dysfunc-
(Ballard et al., 2008). Efficacy assessments included global tion, magnesium effects on NMDA receptors and chronic
and cognitive performances, ADLs and neuropsychiatric glutamatergic overstimulation; it also has shown positive
symptoms. Rivastigmine was superior to placebo on three effects on long-term potentiation and cognitive tests in stan-
measures of cognitive performance (but no other out- dard animal models of impaired synaptic plasticity (Möbius,
comes). Cognitive improvement occurred in older patients, 1999). The pivotal memantine 9M-Best study by Winblad
probably likely to have concomitant AD as evidenced by and Poritis (1999) in severe dementia included both AD and
medial temporal atrophy. VaD patients. A statistically significant effect was detected
Regarding the use of rivastigmine in patients with in both dementias. Orgogozo et al. (2002) and Wilcock et al.
AD+CVD, Kumar et al. (2000) compared the outcomes of (2002) completed two 6-month placebo-controlled RCTs of
AD patients with or without concurrent vascular risk fac- memantine (20 mg/day) in mild-to-moderate probable VaD,
tors; cognitive effects were seen in both groups, but patients diagnosed by NINDS-AIREN criteria (Román et al., 1993).
with AD and vascular risk had greater clinical benefit. These In study MMM 300, Orgogozo et al. (2002) random-
findings were confirmed by Erkinjuntti et al. (2002b) in an ized 147 patients to memantine and 141 to placebo. After
open-label extension study of 104 weeks. Compared with 28 weeks, the mean ADAS-cog scores were significantly
non-hypertensive patients with AD, significant treatment improved relative to placebo: the memantine group gained
differences were observed in the hypertensive subgroup on an average of 0.4 points versus a decline of 1.6 in the pla-
the Progressive and Global Deterioration Scales (PDS and cebo group, a difference of 2.0 points (p = 0.0016). The
GDS). Furthermore, Erkinjuntti et al. (2003b) stratified 725 CIBIC-plus that improved or remained stable reached 60%
patients with AD treated with rivastigmine, according to with memantine compared with 52% with placebo (p =
the presence or absence of arterial hypertension at baseline. 0.227). The Gottfries–Bråne–Steen (GBS) Scale (Gottfries
Rivastigmine 6–12 mg/day provided better outcomes than et al., 1982) and the Nurses’ Observation Scale for Geriatric
placebo on the PDS in the hypertensive (p = 0.031) and non- Patients (NOSGER) (Spiegel et al., 1991) total scores at week
hypertensive (p = 0.035) subgroups. All patients receiving 28 did not differ significantly between the two groups.
rivastigmine 6–12 mg/day had superior CIBIC-plus scores However, the GBS Scale intellectual function sub-score
to those receiving placebo. The additional apparent benefits and the NOSGER disturbing behaviour dimension also
on disease progression detected in patients with AD and favoured memantine (p = 0.04 and p = 0.07, respectively).
hypertension may be linked to drug effects on cerebrovas- Wilcock et al. (2002) in study MMM 500 random-
cular risk factors, or to a larger underlying cholinergic defi- ized 277 patients to memantine and 271 to placebo. At 28
cit in patients with AD and hypertension. weeks, the active group gained 0.53 points and the placebo
The problems associated with gastrointestinal side effects declined by 2.28 points in ADAS-cog, a significant dif-
requiring slow oral titration of rivastigmine were solved ference of 1.75 ADAS-cog points between the groups (p <
to a large extent with the use of a dermal patch (Winblad 0.05). There were no differences in CGIC, MMSE, GBS or
674 Dementia

NOSGER between groups. Memantine was well tolerated hippocampus, caudate nucleus and cerebral cortex (Traber
in both studies. Möbius and Stoffler (2002) performed a and Gibsen, 1989). Nimodipine binds to slow l-type calcium
post hoc pooled analysis of these two placebo-controlled receptors, preventing influx of calcium into vascular smooth
trials of memantine in VaD. Baseline severity, as assessed muscle cells and into ischaemic neurones (Greenberg et al.,
by MMSE, showed larger cognitive benefit in patients with 1990); experimentally, it protects against age-associated
more advanced disease. Also, the cognitive treatment effect microvascular abnormalities in the rat brain (De Jong et al.,
for memantine was more pronounced in the small ves- 1990 Mark et al., 1992). Although a neuroprotective effect
sel type group without cortical infarctions by CT or MRI. has been postulated in stroke patients, nimodipine showed
The placebo subgroup of patients with large vessel disease only a trend for better outcome when used within the first 12
showed less cognitive decline than the other subgroup. hours of stroke onset (Gelmers and Hennerici, 1990).
Möbius and Stoffler (2002, 2003) reviewed the results of A reanalysis of the Scandinavian Trial of Nimodipine
these studies of memantine in VaD in a post hoc analysis in MID (Pantoni et al., 2000) showed a beneficial effect in
and showed that the small vessel disease subgroup of VaD tests of attention and psychomotor performance in the sub-
accounted for most of the outcome (responders) while the group of patients with subcortical small vessel VaD, but not
large vessel disease subgroup (non-responders) was not sta- in patients with PSD of the MID type. A pilot open-label
tistically significant compared with placebo. Solving this trial of nimodipine in patients with small vessel VaD was
issue would probably require a separate trial of memantine positive (Pantoni et al., 1996). In this study, subcortical VaD
in small vessel VaD. was defined according to ICD-10 criteria (World Health
In a review, Thomas and Grossberg (2009) concluded Organization, 2008), and inclusion criteria required the
that memantine shows promise for the treatment of patients patients to have mild-to-moderate dementia (MMSE 12–24,
with VaD because of its safety and efficacy. However, GDS 3–5), as well as CT evidence of extensive leukoara-
Kavirajan and Schneider (2007) performed a meta-analysis iosis and at least one lacunar infarct. The primary measure
of 6-month RCTs in VaD (three donepezil, two galantamine, of efficacy was the Sandoz Clinical Assessment – Geriatric
one rivastigmine and two memantine trials), comprising (SCAG) scale (Shader et al., 1974).
3093 patients on the study drugs and 2090 patients on pla- Using similar criteria and outcome measures, a large
cebo. They found that all drugs produced small but signifi- multicentre, double-blind, placebo-controlled RCT of
cant cognitive effects of uncertain clinical significance in nimodipine in subcortical VaD was conducted in Italy and
patients with mild-to-moderate VaD. Compared with pla- Spain (Pantoni et al., 2005). Of 242 patients randomized to
cebo, more dropouts and AEs (anorexia, nausea, vomiting, nimodipine 90 mg/day or placebo, 230 patients (nimodipine
diarrhoea and insomnia) occurred with all three ChEIs, but N = 121, placebo N = 109) were included in the intention-to-
not with memantine. The authors concluded that the data treat analysis. At 52 weeks, there was no difference in the
were insufficient to support widespread use of these drugs primary outcome measure (SCAG scale) between the two
in VaD. Wong et al. (2009) analysed the cost-effectiveness groups. However, patients on nimodipine performed better
of using ChEIs and memantine in VaD and concluded that than placebo in lexical production (p < 0.01), MMSE (p <
treatment with ChEIs or memantine was more effective, but 0.01) and GDS (p < 0.05). More dropouts and AEs occurred
more costly, than standard care for mild-to-moderate VaD. among the placebo group (cardiovascular and cerebrovas-
cular events, and behavioural disturbances requiring inter-
60.6.3 CALCIUM CHANNEL BLOCKERS vention). The authors concluded that nimodipine offers
benefit in subcortical VaD and might protect against car-
The main calcium channels blockers used for the treatment diovascular and CVD comorbidities in this high-risk group.
of VaD include nimodipine, nicardipine, lacidipine and A Cochrane review on nimodipine (López-Arrieta and
fasudil. Birks, 2002) found no convincing evidence for its efficacy in
AD, VaD or mixed forms of dementia.
60.6.3.1 Nimodipine
60.6.3.2 Nicardipine
Nimodipine is a dihydropyridine-type CCB used as anti-
hypertensive agent. Due to its highly lipophilic properties it Nicardipine is a dihydropyridine-type CCB with high
readily crosses the blood–brain barrier (BBB), affects auto- vascular selectivity, strong antihypertensive activity and
regulation of CBF and produces vasodilatation of small cere- cerebrovascular effects (Amenta et al., 2008). Nicardipine
bral blood vessels (Jansen et al., 1991). Nimodipine reduces protects from atherogenesis in experimental vascular injury
the severity of neurological outcome after subarachnoid (Weinstein and Heider, 1989); has antioxidant effects, pre-
haemorrhage (SAH) secondary to ruptured aneurysms, venting endothelial cell damage from free radical injury
probably by decreasing cerebral vasospasm, although this (Mak et al., 1995) and inhibits platelet aggregation (Yamada
effect has not been demonstrated by angiography or tran- et al., 1990). Nicardipine, like all CCBs, lowers arterial pres-
scranial Doppler (Ohman et al., 1991). In addition to its sure by reducing peripheral vascular resistance, thereby
vascular effects, nimodipine appears to have nootropic prop- decreasing both systolic and diastolic BP and improving
erties. There is a high density of nimodipine-binding sites in myocardial oxygen supply by vasodilating coronary arteries
Therapeutic strategies for vascular cognitive disorders 675

(Amenta et al., 2008). Therefore, nicardipine is an anti- not taking antihypertensives. Nicardipine was well tolerated
angina and cardioprotective agent (Pepine and Lambert, and no major side effects were seen.
1990). CCBs are more effective in BP lowering in patients Based on this evidence, Amenta et al. (2008, 2009) con-
older than 55 years or those of black ethnic origin at any cluded that the anti-hypertensive effect of nicardipine, its
age (Messerli et al., 2007). Amenta et al. (2009) recommend safety and effectiveness in improving cognition and func-
nicardipine for the treatment of hypertension after acute tional domains make this a recommended drug in the treat-
ischaemic stroke or intracerebral haemorrhage for the con- ment of cognitive impairment of vascular origin.
trol of vasospasm in SAH, as well as for stroke prevention
(Martí-Massó and Lozano, 1990). 60.6.3.3 Lacidipine and lercanidipine
Amenta et al. (2009) reviewed the effects of nicardipine
on cognition in 24 studies performed primarily in Japan, Lacidipine and lercanidipine are third-generation calcium
Spain and Italy on about 6000 patients with ‘chronic cere- antagonists that cause systemic vasodilatation by block-
brovascular insufficiency’ due to several forms of CVD ing calcium entry through l-type calcium channels in cell
including PSD and ranging in severity from vascular MCI membranes; these agents also improve carotid atherosclero-
to VaD. Positive effects of treatment with nicardipine were sis (Zanchetti et al., 2004), small vessel disease (Frishman,
observed in over 60% of patients; more favourable results 2002) and cerebral flow (Semplicini et al., 2000) in hyper-
occurred in hypertensive patients resulting in decreased tensive patients; their use results in less peripheral oedema
systolic and diastolic BP, improvement of neurological than with older dihydropyridine CCBs.
symptoms and signs, as well as amelioration of cognition
assessed with the SCAG scale, MMSE and other batteries. 60.6.3.4 Fasudil
The Spanish Group of Nicardipine Study in VaD (Grupo
Español de Estudio de Nicardipino, 1999) conducted a dou- Fasudil hydrochloride is a novel intracellular calcium
ble-blind, placebo-controlled RCT to investigate the effect antagonist, with Rho-kinase inhibitory activity on arte-
of nicardipine on cognitive function in patients with VaD. rial walls affecting vessel remodelling (Pearce et al., 2004).
The effect of daily treatment with nicardipine (20 mg three Fasudil has been used to prevent and treat vasospasm asso-
times per day) for 1 year was investigated in 156 patients ciated with SAH (Suzuki et al., 2008). Kamei et al. (1996)
with cognitive impairment of vascular origin, random- reported the use of fasudil in the treatment of two patients
ized to nicardipine (N = 81) or placebo (N = 75). A total of with BD using 31P-magnetic resonance spectroscopy and
142 subjects completed the study (nicardipine N = 73, pla- Xenon-CT. Treatment with fasudil at 30 or 60 mg/day orally
cebo N = 60) on intention-to-treat analysis. The primary for 8 weeks controlled the fluctuating symptoms in both
efficacy variable was the loss of >10% of the MMSE basal patients. Mental tests and imaging also improved.
score; other end points were the Short Portable Mental
Status Questionnaire (SPMSQ) score (Pfeiffer, 1975), func- 60.6.4 NOOTROPIC AGENTS
tional disability and drug safety. At 1 year, 21.1% of patients
treated with nicardipine had lowered the MMSE score ver- The name nootropic (Greek nous, mind; trophikos, nour-
sus 34.6% with placebo. Favourable effects of nicardipine ishing) describes a category of agents with neuroprotective
were found on females (40.9% versus 10.5%, p = 0.01876), capacity against anoxic or oxidative injuries. Piracetam,
previously untreated subjects (46.2% versus 13.3%, p = oxiracetam and citicoline will be reviewed.
0.00748) and patients on concomitant antiplatelet treat-
ment (35.0% versus 15.9, p = 0.03836). Survival analysis 60.6.4.1 Piracetam
found patients on nicardipine took longer to lose cognitive
capacities (p = 0.031, RR 1.15–3.99). In summary, nicardip- Piracetam (2-oxo-1-pyrrolidine acetamide), the first noo-
ine significantly delayed cognitive decline, producing better tropic agent, is a cyclic derivative of γ–aminobutyric acid
evolution in females. The drug was remarkably safe and was (GABA), which can cross the BBB (Hitzenberger et al.,
well tolerated for 1 year; side effects secondary to vasodila- 1998) to concentrate selectively in the brain cortex (Vernon
tation were short lived and of low intensity. and Sorkin, 1991). At low doses, piracetam increases both
The largest (N = 6375 patients) nicardipine trial on vas- oxygen and glucose utilization via adenosine triphosphate
cular MCI and VaD was an open, prospective, multicen- (ATP) pathways and the release of some neurotransmit-
tre, 6-month study to test the efficacy and tolerability of ters – in particular dopamine metabolites – while at higher
a single 40 mg/day oral dose of nicardipine retard (slow dosages, it is associated with platelet anti-aggregation and
release) in patients with cognitive deterioration of vas- rheological effects with antithrombotic properties (Moriau
cular origin (González-González and Lozano, 2000). BP, et al., 1993). It enhances the microcirculation by promoting
ADLs and cognition with the SPMSQ score (Pfeiffer, 1975) erythrocyte deformability and reducing adherence to endo-
were obtained. Nicardipine improved functional capac- thelial cells. However, studies of piracetam in acute stroke
ity in 65.5% of the patients; systolic/diastolic BP decreased have been inconclusive, although improvement of aphasia
11.3/7.4 mm Hg in hypertensive patients treated with in patients treated within 7 hours of stroke onset was seen
other antihypertensive drugs and 5.9/4.3 mm Hg in those (Hitzenberger et al., 1998). Several animal studies showed
676 Dementia

effects of piracetam on memory and facilitation of retention in a rat model of chronic cerebral hypoperfusion. Silveri
over 24 hours. A Cochrane review (Flicker and Grimley et al. (2008) demonstrated that citicoline treatment for 6
Evans, 2001) concluded that the usefulness of piracetam in weeks produced significant increases in phosphocreatine
patients with AD and VaD in small clinical trials is unclear, (+7%), in beta-nucleoside triphosphates (largely brain ATP
based mainly on subjective Global Impression of Change. [+14%]), and in the ratio of phosphocreatine to inorganic
These findings are supported by a larger Russian trial phosphate (+32%), as well as significant changes in mem-
(Batysheva et al., 2009) on 70 patients (37 women and 33 brane phospholipids in the anterior cingulate cortex.
men, mean age 62 years), including 29 (41.5%) with PSD and Based on a large body of experimental animal evidence
41 (58.5%) with chronic cerebral ischaemia. Improvements (Bustamante et al., 2012), the neuroprotective effects of citi-
in cognition (MMSE) and behaviour were better at a dose coline have been well documented in at least 64 ­animal stud-
of 2400 mg/day. Piracetam was well tolerated. A review ies of stroke therapy. The accepted mechanisms of action of
of 19 trials in patients with several forms of dementia citicoline are the following: (1) protects cell membranes by
(Waegemans et al., 2002) and a protective effect in patients accelerating phospholipid resynthesis, (2) inhibits activation
undergoing coronary artery bypass surgery (Uebelhack of certain phospholipases, (3) increases glutamate transport
et al., 2003) appear to provide some support for a moderate in astrocytes (Hurtado et al., 2008), (4) increases cholin-
effect of piracetam on cognitive impairment. ergic transmission, (5) restores the activity of mitochon-
drial ATPase and membrane Na+/K+ ATPase, (6) increases
60.6.4.2 Oxiracetam ­neuronal ATP levels in ischemia, (7) decreases ischaemic
lesion volume, (8) accelerates reabsorption of cerebral
Oxiracetam is a structural analogue of piracetam. This oedema and (9) inhibits apoptosis mechanisms.
compound has enhancing effects on vigilance and memory. In experimental animals, citicoline enhances numerous
In comparison with piracetam, oxiracetam exhibits greater neuroregenerative processes including inhibition of apop-
improvement in memory (Itil et al., 1986) and has been tosis, angiogenesis, neurogenesis, gliagenesis, synaptogen-
used extensively for the treatment of dementia, including esis, and modulation of neurotransmitters; these effects are
AD, VaD, MID (Baumel et al., 1989; Dysken et al., 1989), similar to those induced by stem cells (Gutiérrez-Fernández
and mixed forms. Green et al. (1992) failed to demonstrate a et al., 2012; Krupinski et al., 2012). Citicoline can produce
difference from placebo in AD patients. In contrast, Maina regrowth of dendritic spines in an experimental stroke
et al. (1989) studied 289 patients with MID in a double-blind model (Hurtado et al., 2007). Furthermore, Hurtado et al.
placebo-controlled trial and after 12 weeks of treatment (2013) demonstrated in experimental stroke that citicoline
found that oxiracetam reduced behavioural symptoms. increases SIRT1 protein levels in the brain concomitantly
with neuroprotection. Moreover, resveratrol elicited a
60.6.4.3 Citicoline strong synergistic neuroprotective effect with CDP-choline,
suggesting that these two products probably should be
Citicoline, also known as CDP-choline or cytidine-5′- used together. SIRT1 plays a major role in the prevention
diphosphatecholine, is a naturally occurring endogenous of ageing-related conditions, including neurodegenerative
nucleoside. Citicoline functions as an intermediate in three diseases such as Alzheimer’s and Parkinson’s diseases and
major metabolic pathways (Conant and Schauss, 2004): amyotrophic lateral sclerosis (ALS).
(1) synthesis of phosphatidylcholine (lecithin), one of the In a pooled analysis of clinical trials on acute isch-
major cell membrane phospholipids with an important role aemic stroke, citicoline enhanced the possibility of recov-
in lipoprotein formation. Citicoline formation is the rate- ery (Dávalos et al., 2002; Saver, 2008). However, the ICTUS
limiting step in the synthesis of phosphatidylcholine. (2) trial (Dávalos et al., 2012) failed to show a beneficial effect
Synthesis of ACh: citicoline by providing choline for this in acute ischaemic stroke. Reviews of the effects of citicoline
neurotransmitter could limit choline availability for mem- in acute stroke patients (Secades et al., 2016; Álvarez-Sabín
brane synthesis (Ulus et al., 1989). (3) Oxidation to betaine, and Román, 2013) and post-stroke VCD (Álvarez-Sabín
a methyl donor. The main components of citicoline, choline and Román, 2011) concluded that citicoline may possess
and cytidine, are readily absorbed in the gastrointestinal substantial neuroregenerative potential that would explain
tract and cross the BBB (Secades and Lorenzo, 2006). As a better its long-term beneficial effects in post-stroke patients.
dietary supplement, choline is part of the vitamin-B group. Álvarez-Sabín et al. (2013) conducted a study to assess the
A related product, choline alphoscerate, has also been used effect of citicoline on cognitive function in 347 patients with
less frequently (Parnetti et al., 2007). acute stroke who received citicoline 2 g/day for 6 weeks. After
In animal studies, citicoline enhances repair of ischaemic 6 weeks, they were randomized in two groups of 175 subjects
neuronal injury and increases levels of ACh and dopamine each: one group stopped citicoline (controls) and the other
(Secades and Frontera, 1995; Secades, 2002; Secades and received citicoline 1 g/day for 6 months. Cognitive functions
Lorenzo, 2006; Saver, 2008; Secades, 2011). In aged animals, improved 6 months after stroke in the entire group, but in
citicoline increases dopamine release, improving learning comparison with controls citicoline-treated patients showed
and memory tasks (Cacabelos et al., 1993). Lee et al. (2009) statistically significant better outcome in attention-executive
showed that citicoline protects against cognitive impairment functions (OR 1.7, 90% CI 1.1–2.7, p = 0.019) and temporal
Therapeutic strategies for vascular cognitive disorders 677

orientation (OR 1.7, 90% CI 1.02–2.9, p = 0.042). These results 60.6.5.1 Pentoxifylline


suggest that citicoline treatment within 24 hours of stroke
onset, following by continuous treatment for 6 months is Haemorheological alterations are found in AD patients
safe, and benefits prevention of post-stroke VCD compared and in some forms of VaD, such as BD, in comparison with
to placebo. Furthermore, continuous citicoline treatment in age-matched controls (Solerte et al., 2000). Abnormalities
post-stroke patients for up to 12 months has excellent toler- included hyperviscosity, increased sedimentation rate, hyper-
ability and safety and continued improvement in post-stroke fibrinogenemia and increased acute-phase reactants; these
VCD especially in temporal orientation, executive functions changes correlate with increased levels of TNF-α and IFN-γ.
and attention (Álvarez-Sabín et al., 2016). Pentoxifylline treatment lowers fibrinogen and TNF-α levels.
Citicoline has been used in a number of trials in AD Black et al. (1992) demonstrated beneficial effects of pentoxi-
patients showing consistent moderate improvement of fylline in patients with MID. These preliminary findings were
memory and behaviour (Cacabelos et al., 1993; Álvarez confirmed in the larger, multicentre European Pentoxifylline
et al., 1997). A placebo-controlled trial on 30 patients with Multi-Infarct Dementia Study (1996) that demonstrated sig-
VaD showed no evidence of cognitive improvement (Cohen nificant cognitive improvement in the MID form of VaD in
et al., 2003). Cotroneo et al. (2013) in the IDEALE study comparison with placebo. Sha and Callahan (2003) reviewed
demonstrated modest cognitive effects in 349 patients with 20 articles on pentoxifylline use in VaD but only four stud-
mild VCD. Previously, a Cochrane review (Fioravanti and ies met systematic review criteria. A trend towards improved
Yanagi, 2005) included 14 studies on more than 1000 aged cognitive function was found in pentoxifylline-treated
individuals with symptoms ranging from memory disor- patients. A subgroup analysis of three studies of VaD noted
ders to patients with vascular MCI, VaD or senile dementia. statistically significant improvement in cognitive function
Studies’ duration ranged between 20 and 30 days, one study with pentoxifylline compared with placebo and suggested a
was of 6 weeks’ duration, four studies lasted 2 and 3 months potential therapeutic role for pentoxifylline in VaD.
and one study lasted 12 months. Multiple doses, inclusion
criteria and outcome measures were used. Overall results 60.6.5.2 Propentofylline
(884 patients) showed evidence of benefit of CDP-choline
on memory and behaviour but not attention. There was sig- Beneficial effects on learning and memory were observed
nificant improvement on the Global Impression of Change, in several European and Canadian double-blind, placebo-
in comparison with the placebo group. The effect size was controlled, parallel group RCTs of PPF in AD and VaD
very large (OR 8.89, 95% CI 5.19–15.22, p < 0.001) indicat- (Kittner et al., 1997; Mielke et al., 1998; Pischel, 1998; Rother
ing a strong drug effect for improvement under treatment et al., 1998; Kittner, 1999). Despite these results, the clinical
(Fioravanti and Yanagi, 2005). These authors concluded that development of PPF was halted.
the cognitive effects of citicoline are clearly evident at the
behavioural level and can be easily appreciated with clinical 60.6.5.3 Denbufylline
observation of patients, irrespective of the methods used to
measure them. Citicoline is very well tolerated; more side Denbufylline, a xanthine derivative and PDE4 inhibitor, has
effects were seen with placebo than with active treatment. effects such as vasodilatation of cerebral vessels (Willette
et al., 1997) and potent activation of the hypothalamic–
60.6.5 XANTHINE DERIVATIVES pituitary–adrenal axis. PDE4 is one of the cyclic adenos-
ine monophosphate (cAMP)–specific phosphodiesterases
The main xanthine derivatives used to treat VaD are pent- whose tissue distribution is important in pathologies related
oxifylline or oxpentifylline, denbufylline, a phosphodi- to the central nervous and immune systems. In experimen-
esterase 4 (PDE4) inhibitor, and propentofylline (PPF). tal animals, denbufylline increases adrenocorticotropic
Pentoxifylline has haemorheological effects both on the hormone (ACTH), circulating corticosterone, luteinizing
microcirculation and on peripheral vascular disease (in par- hormone, corticotrophin-releasing hormone, and cAMP
ticular for treating intermittent claudication). Pentoxifylline content of the hypothalamic tissue, but is without effect
has immunomodulatory properties, increasing the rate of on arginine vasopressin (Kumari et al., 1997). Treves and
neutrophil migration, suppressing monocyte production Korczyn (1999) studied patients with AD, mixed demen-
of tumour necrosis factor (TNF-α) and inhibiting leuco- tia, and VaD treated with denbufylline. No significant dif-
cyte stimulation by TNF-α and interleukin-1 (IL-1). These ferences were found in comparison with placebo for the
properties are strong contributors to its haemorheological treatment of AD or VaD, although patients who received
effects (Samlaska and Winfield, 1994). PPF exhibits ade- denbufylline tended to improve their cognitive scores.
nosine-mediated nootropic properties against post-anoxic
neuronal cell damage and glial modulation effects with 60.6.6 VASODILATORS
inhibition of microglial activation. Bruno et al. (2009)
showed improvement of neurological and neurochemical Yesavage et al. (1979) reviewed 102 studies on the use of
deficits in rats subjected to transient brain ischaemia treated vasodilating agents in senile dementias; the postulated effect
with pentoxifylline. of these agents was to counteract ‘hardening of the arteries’.
678 Dementia

Recommendations for most of these agents were based in tri- maintained for 6–12 months. Cognitive assessment (MMSE)
als invalidated by small numbers of participants, short open was performed in 261 patients. The difference between treat-
treatment periods, and variations in diagnostic criteria and ment and control groups on the MMSE favoured nicergoline;
clinical end points. Cochrane (1979) strongly criticized the at 12 months, the effect size was 2.86. Herrmann et al. (1997)
poor quality of this evidence. The principal pharmacologi- conducted a double-blind placebo-controlled RCT involving
cal agents with primary effect on smooth muscle resulting in 136 patients with MID. After 6 months of treatment, cogni-
vasodilatation are cyclandelate, papaverine, isoxsuprine, cin- tion in the nicergoline group was significantly better than in
narizine and nafronyl. There were no significant effects with the placebo group. Another placebo-controlled pilot study
the use of vasodilating agents in VaD (Cook and James, 1981). by Bes et al. (1999) recruited 72 elderly hypertensive patients
Nicotinic acid (niacin), long used for its vasodilating with VCI and evidence of leukoaraiosis, randomly assigned to
properties, has received renewed attention due to its effects either nicergoline (n = 36) or placebo (n = 36) for 24 months.
in the treatment of primary hypercholesterolemia and Nicergoline (30 mg twice daily [b.i.d.] for 24 months) was
mixed dyslipidemia. It is the only drug that primarily low- well tolerated and attenuated cognitive decline. Winblad et al.
ers concentrations of non-sterified fatty acids and thereby (2008) found that up to 89% of patients treated with nicer-
lowers very low-density lipoprotein (VLDL) triglycerides. goline 30 mg b.i.d. showed improvements in cognition and
Nicotinic acid also seems to improve insulin resistance behaviour; patients remained stable after 12 months of treat-
and stimulate cholesterol mobilization from macrophages, ment. In addition, nicergoline has favourable safety and toler-
offering an avenue for regression of atherosclerotic vascular ability (Fioravanti et al., 2014).
lesions (Karpe and Frayn, 2004).
60.6.8 POSATIRELIN
60.6.7 ERGOT DERIVATIVES
Posatirelin is a TRH analogue. In a rat experimental model
The main ergot derivatives used for the treatment of VaD of brain cholinergic deficit by lesion of the nucleus basa-
are nicergoline and codergocrine or ergoloid mesylates – a lis magnocellularis (nbM), posatirelin treatment rescued
mixture (in a 3:3:2:1 ratio) of four dehydrogenated mesyl- cholinergic neurones of the nbM and their cholinergic
ated ergot peptide derivatives, i.e. dihydroergocornine, projections to the cerebral cortex (Sabbatini et al., 1998).
dihydroergocristine, dihydro-α-ergocryptine and dihydro- Posatirelin has been used in AD and VaD patients (Parnetti
β-ergocryptine (Wadworth and Chrisp, 1992). et al., 1995, 1996). VaD patients treated with posatirelin
showed a significant improvement in intellectual perfor-
60.6.7.1 Ergoloid mesylates mance, orientation, motivation and memory as compared
to controls. The drug was well tolerated.
The metabolic effects of ergoloid mesylates (Hydergine) may
include enhancement of noradrenergic, dopaminergic and 60.6.9 ANTITHROMBOTIC AGENTS
serotoninergic neurotransmission (Weil, 1988) and reduc-
tion of free radicals. Schneider and Olin (1994) reviewed 151 Antiplatelet drugs are effective to prevent TIAs and isch-
reports on the use of Hydergine in senile dementia, but only aemic stroke (Meschia et al., 2014). Antiplatelet agents
47 of these trials (31%) met strict criteria for meta-analysis. A include ASA, sulfinpyrazone, ticlopidine, clopidogrel,
Cochrane review (Olin et al., 2001) confirmed that patients dipyridamole, and orally administered IIb/IIIa inhibitors.
with VaD appeared to benefit more than patients with AD The Antithrombotic Trialists’ Collaboration (Easton, 2003)
in terms of cognition, clinical global ratings and combined assessed the effect of antiplatelet therapy in 135,000 patients.
measures. However, compared with placebo, efficacy was very Therapy reduces the combined odds of stroke, myocardial
modest. Thus, there are no grounds to recommend its use. infarction or vascular death by 22%, and reduces the odds of
a non-fatal stroke by 25% in patients with or without a history
60.6.7.2 Nicergoline of stroke. Likewise, among patients with nonvalvular atrial
fibrillation, anticoagulation (International Normalized Ratio
Nicergoline is an ergot derivative that may protect against [INR] ≥ 2.0) reduces the frequency, severity and mortality of
degeneration of cholinergic neurones (Giardino et al., 2002). ischaemic stroke (Hart, 2003). Substantial progress has been
Nicergoline has a broad spectrum of action (Winblad et al., achieved with the use of new oral anticoagulants, particu-
2008): (1) as α1-adrenoceptor antagonist it induces vasodilata- larly for stroke prevention in atrial fibrillation (January et al.,
tion and increases arterial blood flow; (2) it enhances cholin- 2014). However, few trials of antiplatelet agents have been
ergic and catecholaminergic neurotransmission; (3) it inhibits conducted in patients with VaD.
platelet aggregation; (4) it promotes metabolic activity, result-
ing in increased oxygen and glucose utilization; and (5) it has 60.6.9.1 Aspirin
neurotrophic and antioxidant properties. Nicergoline has been
used for the treatment of various dementias, including AD and A single population-based study (Sturmer et al., 1996)
VaD (Fioravanti and Flicker, 2001). The therapeutic effects of showed that ASA users have a slight protection against
nicergoline were evident by 2 months of treatment and were cognitive decline (OR 0.97–0.87), but a Cochrane review
Therapeutic strategies for vascular cognitive disorders 679

(Williams et al., 2000) concluded that there is no evidence 60.6.11 JIANNAO YIZHI


that ASA treats VaD effectively.
Jiannao Yizhi, a Chinese herbal medicine, was studied for
60.6.9.2 Triflusal the treatment of VaD (Zhang et al., 2002) in a multicentre,
double-blind, placebo-controlled RCT of 242 patients with
Triflusal is an antiplatelet agent structurally related to the mild-to-moderate VaD; 89 cases were randomized to the
salicylates, but it is not derived from ASA. Triflusal and its active group (Jiannao Yizhi granules), 106 cases to the west-
metabolite (3-hydroxy-4-trifluoro-methylbenzoic acid or ern medicine group and 47 to placebo. MMSE and Blessed
HTB) produce specific inhibition of platelet arachidonic acid Dementia Scale were used to evaluate the therapeutic effect
metabolism (McNeely and Goa, 1998). A single 12-month after 60 days. Jiannao Yizhi treatment was superior to west-
open-label trial of triflusal in 73 VaD patients (López-Pousa ern medicine and to placebo (Obulesu and Rao, 2011).
et al., 1997) showed fewer declines in MMSE scores in
the active group compared with untreated subjects. More 60.6.12 VINPOCETINE
recently, triflusal was used in patients with amnesic MCI;
257 patients were randomized to receive 900 mg of triflusal Vinpocetine is a synthetic ethyl ester of apovincamine.
or placebo for 18 months. Triflusal therapy was associated Vinpocetine has been used for cognitive impairment, but
with a significantly lower rate of conversion to dementia the mechanism of action is unclear. A Cochrane review
(Gómez-Isla et al., 2008). (Szatmari and Whitehouse, 2003) of three short-term stud-
ies involving 583 patients with dementia (AD, VaD, mixed)
60.6.9.3 Ginkgo concluded that patients treated with vinpocetine (30–60 mg/
day) showed modest benefit compared to placebo.
Ginkgo, extract EGb 761 from the G. biloba tree (the ‘maiden-
hair’ tree), has been widely used in several European countries 60.6.13 DEHYDROEPIANDROSTERONE
and in North America for the treatment of ‘chronic cerebral (DHEA)
circulatory insufficiency’ (Kleijnen and Knipschild, 1992;
Gertz and Kiefer, 2004). Ginkgo biloba extracts have vasodi- DHEA is a weakly androgenic adrenal steroid and an inter-
lating and antioxidant properties, as well as haemorheological mediary in the biosynthesis of androgens and oestrogens. It
and nootropic effects, and decrease platelet aggregability and is a neurohormone; small quantities of DHEA are produced
blood viscosity, but the mechanism of action remains poorly in the brain (Knopman and Henderson, 2003). DHEA or its
understood (Zimmermann et al., 2002; Gertz and Kiefer, sulphate ester metabolite (dehydroepiandrosterone sulphate
2004). Several trials used ginkgo in patients with MID or with [DHEAS]) is the most abundant circulating steroid and
AD plus CVD, with modest positive results (Le Bars et al., declines in serum and CSF with age. Bicikova et al. (2004)
1997; Kanowski and Hoerr, 2003; van Dongen et al., 2003). suggested that levels of these neurosteroids could discrimi-
Using Cochrane data, Kurz and van Baelen (2004) showed sig- nate between AD and VaD; however, CSF levels of DHEAS
nificant but minimal benefit versus placebo with ginkgo treat- are low in both AD and VaD (Kim et al., 2003).
ment only when all doses were pooled. Ginkgo treatment in DHEA is classified as a dietary supplement; a 6-month,
the elderly has been associated with SAH and other haemor- double-blind RCT of DHEA for AD (Wolkowitz et al., 2003)
rhagic complications such as subdural haematoma, intracra- showed modest improvement with DHEA but increas-
nial and intraocular bleeding (Vale, 1998; Fong and Kinnear, ing confusion, agitation, and paranoid reactions were
2003; Meisel et al., 2003). Snitz et al. (2009) conducted the seen among DHEA-treated participants, resulting in high
Ginkgo Evaluation of Memory (GEM) trial and concluded dropout. A single open-label trial of DHEA in the MID
that compared with placebo, the use of G. biloba, 120 mg b.i.d., type of VaD (Azuma et al., 1999) used intravenous DHES
did not prevent cognitive decline in older adults with normal (200 mg/day) for 4 weeks; the treatment markedly increased
cognition or MCI. The minimal therapeutic results should be serum and CSF levels of DHEAS in seven MID patients;
tempered by the potential side effects (Brown et al., 2010). however, improvement of ADL and emotional disturbances
were seen in only three patients.
60.6.10 CHOTO-SAN
60.6.14 CEREBROLYSIN
Choto-san (Gouteng-san) is a Japanese (Kampo) herbal
medicine with apparent neuroprotective effect against glu- Cerebrolysin is a peptidergic drug produced from purified
tamate-induced neuronal death (Itoh et al., 1999; Watanabe porcine brain proteins, with postulated neurotrophic and
et al., 2003). Choto-san was administered for 12 weeks to 10 neuroprotective effects (Windisch et al., 1998; Rockenstein
patients with post-stroke VCI; P3 event-related brain poten- et al., 2003; Plosker and Gauthier, 2009; Gauthier et al.,
tials, MMSE and verbal fluency tests significantly improved 2015). A small open-label trial in patients with AD and VaD
with treatment (Yamaguchi et al., 2004). In a larger trial, (Rainer et al., 1997) showed minimal improvement in cog-
Choto-san resulted in improvement of ADLs, global rating nitive tests and clinical global impression. The main side
and subjective and psychiatric symptoms (Itoh et al., 1999). effects were nausea and vertigo. A Cochrane review (Chen
680 Dementia

et al., 2013) of six RCTs with a total of 597 participants of medications such as pentoxifylline, ancrod (Sherman,
showed minimal MMSE and ADAS-cog improvements. 2002) or bezafibrate (Tanne et al., 2001) to lower fibrinogen
levels could be indicated.
60.6.15 SULODEXIDE Ancrod is a defibrinating enzyme (Sherman, 2002).
Ringelstein et al. (1988) used it in 10 patients with BD.
Sulodexide is a medium molecular weight glycosamino- Subcutaneous ancrod treatment decreased plasma levels
glycan with effects on plasma viscosity by lowering plasma of fibrinogen from 3.26 g/L (SD 1.3) pretreatment (normal
fibrinogen concentrations (Lunetta and Salanitri, 1992). 2.5–4.0 g/L) to 1.52 g/L (SD 0.53) after 1 month. Treatment
Sulodexide differs from other glycosaminoglycans, like improved retinal arteriovenous circulation time (abnor-
heparin, by having a longer half-life, reduced effect on sys- mally slow pretreatment) and increased CO2-induced cere-
temic clotting and bleeding and increased lipolytic activity. bral vasomotor response by transcranial Doppler. However,
Oral administration of sulodexide results in the release of clinical condition and neuropsychological tests were
tissue-type plasminogen activator (tPA) and an increase in unchanged, and there was no decrease in stroke recurrences
fibrinolytic activities. Sulodexide has been used in chronic during 6 months of observation.
venous disease (Andreozzi, 2014), peripheral occlusive arte- In 1968, Walsh observed improved dementia symp-
rial disease with claudication (Shustov, 1997) and diabetic toms with a treatment based on warfarin anticoagulation
nephropathy (Vilayur and Harris, 2009). Parnetti et al. (1997) (Walsh et al., 1978). Currently, this treatment is reserved for
conducted a trial of sulodexide in patients fulfilling NINDS- patients with nonvalvular atrial fibrillation (Hart, 2003),
AIREN criteria for probable VaD; 46 patients were included heart valve replacement or with anticardiolipin antibody
in the active treatment group, compared with 40 in the pent- syndrome. Direct thrombin inhibitors are equally effective;
oxifylline group. Larger reductions of plasma fibrinogen they inhibit fibrin-bound thrombin, produce a predictable
levels were seen with sulodexide, and both groups showed anticoagulant response and require no long-term monitor-
a slight reduction in activated factor VII levels. Dementia ing and no dose adjustment, but are irreversible in emer-
scores improved more in the sulodexide group. gencies. Stroke prevention in patients with atrial fibrillation
is complicated by dementia and cognitive decline (Flaker
60.6.16 THROMBIN INHIBITORS et al., 2010).
Since 1993, Walzl (1993, 2000) has developed a haemorhe-
There is evidence indicating that hypercoagulable states could ological treatment called Heparin-mediated Extracorporeal
result in increased dementia risk (Mari et al., 1996). Bots low-density lipoprotein (LDL)/fibrinogen Precipitation
et al. (1998) from the Dutch Vascular Factors in Dementia (HELP) to reduce elevated fibrinogen levels and increased
Study found that dementia, particularly post-stroke VaD, lipid fractions to control hyperviscosity of plasma and whole
was associated with increased thrombin generation. In this blood and to reduce aggregability or sludging of red blood
population, increased levels of thrombin–antithrombin cells. HELP was used in 141 patients with the MID type of
complex (TAT), cross-linked D-dimer and tPA activity were VaD. Laboratory and clinical evaluations were performed
associated with increased risk of dementia. In addition, before and after treatment. Each HELP treatment reduced
coagulation abnormalities have been described in patients whole blood and plasma viscosity and red cell transit time.
with BD. Schneider et al. (1987) found high fibrinogen and Total cholesterol, low-density lipoproteins, and triglycer-
hyperviscosity in patients with BD. Iwamoto et al. (1995) ides were reduced significantly. Neurological improvement
demonstrated increased platelet activation in BD manifested was confirmed by improved MMSE, Mathew Scale, and
by increased plasma β-thromboglobulin levels. Tomimoto ADL scores. The Italian HELP group continues to use this
et al. (1999, 2001) found coagulation activation leading to method (Stefanutti et al., 2013).
hypercoagulable state in Japanese patients with BD; levels of
fibrinogen, TAT complex, prothrombin fragment 1+2, and 60.6.17 HYPERBARIC OXYGEN
D-dimer were found to be significantly increased, particu- TREATMENT (HBOT)
larly in patients with recent aggravation of their deficits.
There are no RCTs in BD, but these results point toward Vila et al. (1999) reported on HBOT in four BD patients.
several potential therapeutic options (Román, 1999b). Patients received daily sessions of HBOT at 2.5 atmo-
Iwamoto et al. (1995) reported that the use of ticlopidine spheres absolute (ATA) for 45 minutes, for a total of 10 days.
hydrochloride in eight patients with BD resulted in lower Controls received room air at 1.1 ATA. The procedure was
levels of platelet activation, without major clinical change. well tolerated. After treatment, noticeable improvements in
There is a need for RCTs of ticlopidine and other antiplate- gait, urinary symptoms and cognitive tests were observed
let agents (e.g. ASA, dipyridamole and clopidogrel, alone or in all subjects, with increased independence. This improve-
combined) in BD. ment persisted for up to 5 months, after which the previ-
Hyperfibrinogenemia has deleterious effects on ous deficits reappeared but responded again to repeated
haemorheological conditions in the cerebral microcircula- HBOT treatment. Despite the method’s promise, there has
tion that result in hyperviscosity and slowing of blood flow been no independent confirmation of the benefits of HBOT
in deep border-zone territories in BD. Therefore, the use in BD (Román, 1999c). In China, Xiao et al. (2012) found a
Therapeutic strategies for vascular cognitive disorders 681

single study (Wang et al., 2009) involving 64 patients who of stroke and CVD appears to be mandatory for dementia
used HBOT as an adjuvant treatment to donepezil. After prevention (Lechner, 1998; Erkinjuntti and Gauthier, 2002;
12 weeks, patients on HBOT performed significantly better O’Brien et al., 2003; Barnes and Yaffe, 2011; Román et al.,
than controls on two cognitive tests: MMSE and Hasegawa’s 2012; Willis and Hakim, 2013; Román and Boller, 2014).
Dementia Rating Scale. The reader is referred to the Guidelines for Primary Stroke
Prevention of the American Heart Association/American
60.6.18 SPG STIMULATION Stroke Association (Meschia et al., 2014) for the prevention
of atherosclerotic cardiovascular disease (Gorelick et al.,
The SPG is the source of parasympathetic innervation to 2014).
the anterior cerebral circulation. Electrical stimulation of
SPG neurones leads to profound ipsilateral increase in CBF, 60.7.1 TREATMENT OF HYPERTENSION
augmenting tissue perfusion (Henninger and Fisher, 2007;
Khurana et al., 2009). This method was used in the treat- Treating hypertension protects against cognitive decline
ment of acute stroke in ImPACT-I and ImpACT-24 to evalu- even in the absence of stroke (Forette et al., 1998; Clarke,
ate the safety and effectiveness of SPG stimulation using the 1999; Forette et al., 2002). No deleterious effects on cogni-
Ischemic Stroke System (ISS), an implantable neurostimula- tion, mood and quality of life have been demonstrated with
tor (INS). The INS is implanted to all patients through the the treatment of hypertension in the elderly (Applegate et al.,
greater palatine canal, in a minimally invasive, oral proce- 1994; Starr et al., 1996; Prince et al., 1996; Prince, 1997). On
dure under local anaesthesia. Study patients are unilaterally the contrary, Forette et al. (2002) confirmed that treatment
implanted with the INS and treated daily for up to 90 days. of systolic hypertension with nitrendipine protects against
Treatment is expected to augment CBF, improve cerebral dementia in older patients. Compared with placebo, long-
perfusion, and halt cognitive deterioration. Preliminary term antihypertensive therapy reduced the risk of dementia
results in 10 patients (Alladi et al., 2009) confirmed signifi- by 55%, from 7.4 to 3.3 cases per 1000 patient-years. After
cant increase in CBF ipsilateral to the stimulated side and adjustment for sex, age, education, and entry BP, the relative
in the contralateral cerebellum by 18F-fluorodeoxyglucose hazard rate associated with the use of nitrendipine was 0.38
(FDG) positron emission tomography (PET) scan, along (95% CI 0.23–0.64, p < 0.001). Treatment of 1000 patients
with significant improvement in attention, orientation, for 5 years can prevent 20 cases of dementia (95% CI 7–33)
memory, ability to execute verbal commands, and executive (Forette et al., 1998, 2002). The results of the SPS3 study
function. SPG has also been used to treat cluster headaches. (Jacova et al., 2012, 2015) showed that adherence to the anti-
hypertensive treatment is more important than the level of
BP control achieved.
The SCOPE confirmed that the treatment of mild-to-
60.7 GENERAL MEDICAL
moderate hypertension in the elderly prevents stroke and
MANAGEMENT OF THE PATIENT
dementia (Lithell et al., 2003). SCOPE enrolled 4964 patients
WITH VaD aged 70–89 years, with systolic BP of 160–179 mm Hg and/
or diastolic BP of 90–99 mm Hg and an MMSE test score
Epidemiological studies confirm the increased risk of ≥24; patients were treated with candesartan, an angioten-
dementia associated with vascular risk factors, especially sin receptor blocker, or placebo, plus open-label active anti-
hypertension (Launer et al., 1995; Skoog et al., 1996; Kilander hypertensive therapy added as needed. BP reduction was
et al., 1998; Launer et al., 2000). In a French population- slightly better with candesartan, compared with control
based study, Tzourio et al. (1999) found that in 4 years, therapy; this was associated with a modest, non-significant,
untreated hypertension in subjects 59–71 years of age mul- reduction in major cardiovascular events and with a marked
tiplied by four the risk of cognitive decline (OR 4.3, 95% CI reduction in non-fatal stroke. Cognitive function was well
2.1–8.8) compared with 1.9 (95% CI 0.8–4.4) in those being maintained in both treatment groups in the presence of sub-
treated. stantial BP reductions. Current guidelines (JNC8) should
Other important risk factors include diabetes mellitus be consulted for the recommended antihypertensive treat-
(Ott et al., 1999; Stewart and Liolitsa, 1999;), smoking (Ott ments (James et al., 2014; Farooq and Ray, 2015).
et al., 1998), raised homocysteine (Seshadri et al., 2002), Finally, control of hypertension in patients with stroke
particularly with vitamin B12 deficiency (Hooshmand et al., (secondary prevention) also helps to prevent dementia. The
2010, 2012), and mutations of the MTHFR gene (Román, PROGRESS Collaborative Group (2001), an RCT of perin-
2015). These vascular risk factors increase the risk of both dopril, an angiotensin-converting enzyme (ACE) inhibitor
VaD and AD. Other than the APOE gene, other genetic and and a diuretic (indapamide) used in 6105 individuals with
racial factors are probably important. Zamrini et al. (2004a) previous stroke or TIA showed that after 3.9 years follow-
showed that blacks had higher rates of hypertension than up BP was reduced, lowering the risks of stroke and other
whites in Alabama, whereas whites had a higher incidence major vascular events. Dementia was decreased, with a RR
of atrial fibrillation, coronary artery disease, and higher reduction of 12% (Tzourio et al., 2003). Cognitive decline
cholesterol. Therefore, primary and secondary prevention occurred in 9.1% of the treated group and 11.0% of the
682 Dementia

placebo group (risk reduction, 19%). In summary, treat- decline, doubling the overall risk of dementia (Ott et al.,
ment resulted in reduced dementia risk and of the cognitive 1999; Stewart and Liolitsa, 1999).
decline associated with recurrent stroke. Diabetes increases blood viscosity from hyperglycaemia,
endothelial oxidative damage, loss of nitric oxide (NO)–
60.7.2 TREATMENT OF HYPERLIPIDAEMIA mediated endothelial functions and alterations of the BBB
WITH STATINS (Mooradian, 1997), as a result of excessive glycation as well
as from glycoxidized LDLs. The end result is impairment
The most recent recommendations for the management of of perfusion through the cerebral and retinal microcircula-
hyperlipidaemia suggest a reduction of LDL cholesterols to tion. In addition, stress-activated pathways such as the Jun-
below 100 mg/dL, and drug therapy for high-risk patients kinases play a major role in diabetic microangiopathy (Evans
whose LDL ranges from 100 to 129 mg/dL (Grundy et al., et al., 2002). The most current guidelines for the treatment
2004). High-risk patients have coronary heart disease or of type 2 diabetes are the American-European (Nathan et al.,
peripheral vascular disease in the extremities or the vessels 2009) and American Diabetes Association (2014) guidelines.
to the brain, or diabetes, or multiple (two or more) risk fac-
tors (e.g. smoking, hypertension) that give them a greater 60.7.4 CESSATION OF SMOKING
than 20% chance of having a heart attack within 10 years.
Very high-risk patients are those who have cardiovascular Smokers double their risk of coronary artery disease, con-
disease together with either multiple risk factors (especially gestive heart failure and peripheral vascular disease, and
diabetes) severe and poorly controlled risk factors (e.g. con- increase 1.5 times the risk of stroke and dementia (Ott et al.,
tinued smoking) or metabolic syndrome (a constellation of 1998). Smoke, in addition to nicotine and carbon monox-
risk factors associated with obesity including high triglyc- ide, contains a complex mixture of free radicals that cause
erides and low high-density lipoprotein [HDL]). Patients morphological irregularities of the endothelium, formation
hospitalized for acute coronary syndromes or strokes are at of blebs, leakage of macromolecules and increased endothe-
very high risk (Grundy et al., 2004). The most recent guide- lial cell death (Pittilo, 2000). Smoke reduces prostacyclin
lines recommend statin therapy for those with coronary release, enhances endothelium-derived vasodilatation, and
heart disease or other high-risk conditions such as diabetes decreases NO concentrations and cyclic guanosine mono-
mellitus or symptomatic carotid atherosclerosis. In these phosphate (cGMP) production, increasing aggregation of
groups, statin treatment is associated with 20% reduction in platelets and leucocytes. Smoking worsens atheromatous
the risk of a first stroke (Gorelick et al., 2014). plaque formation in carotid arteries and increases hyperten-
The use of statins may reduce the risk of dementia (Jick et al., sion, blood coagulability, serum viscosity and fibrinogen.
2000; Rockwood et al., 2002; Zamrini et al., 2004b). Jick et al. Smokers have worse cognitive performances than non-
(2000) studied 284 patients with dementia and 1080 controls smokers including reduced psychomotor speed and reduced
aged over 50 years; the adjusted RR for those prescribed statins cognitive flexibility. This effect is observed in subjects as
was 0.29 (95% CI 0.13–0.63, p = 0.002) indicating a substan- young as 45 years old (Kalmijn et al., 2002). A meta-analysis
tially lowered risk of developing dementia, independent of the of prospective cohort studies (Zhong et al., 2015) demon-
presence or absence of untreated hyperlipidaemia. Zamrini strated that smoking is associated with an increased risk
et al. (2004b) conducted between 1997 and 2001 a study of of dementia. For VaD, the RR was 1.38 (95% CI 1.15–1.66).
veterans in Birmingham, AL; patients with a new diagnosis of For all-cause dementia, the risk increased by 34% for every
AD (n = 309) were compared with age-matched non-AD con- 20 cigarettes per day (RR 1.34, 95% CI 1.25–1.43). Smoking
trols (n = 3088). In this group, statins users had a 39% lower cessation decreases the risk to that of never smokers.
risk of AD relative to nonstatin users (OR 0.61, 95% CI 0.42–
0.87). These results indicate a possible antidementia effect of 60.7.5 DIET
statins, perhaps related to anti-inflammatory effects. Positive
effects have been noted in patients with VaD (Giannopoulos Dietary change with reduced sodium intake is a crucial
et al., 2014). However, the use of statins in nondemented, non- component of the treatment of arterial hypertension. The
hyperlipidaemic patients cannot be recommended (Miller and Dietary Approaches to Stop Hypertension (DASH) diet
Chacko, 2004; McGuinness et al., 2014). (Sacks et al., 2001) is currently recommended by the US
Department of Health and Human Services. This diet is
60.7.3 CONTROL OF DIABETES rich in magnesium, potassium, calcium, protein and fibre
and low in saturated fat, cholesterol and total fat. There is
The abnormalities in carbohydrate, lipid and protein metab- emphasis on fruits, vegetables, low fat dairy foods, whole
olism resulting from diabetes mellitus injure blood vessels, grain products, fish, poultry and nuts. McGuire et al. (2004)
nerves and other tissues. Diabetes increases up to fourfold randomized hypertensive patients to advice-only group; to
the RR for cardiovascular and CVD due to the microvas- a group treated with weight loss, increased physical activity,
cular (retinopathy, nephropathy, neuropathy, lacunes) and and reduced sodium and alcohol intake and a third group
macrovascular (coronary heart disease, stroke, peripheral that included the latter plus the DASH diet. At 6 months,
arterial disease) complications. Diabetes produces cognitive compared with the advice-only group, the second group
Therapeutic strategies for vascular cognitive disorders 683

had a decline of mean systolic BP of 3.7 mm Hg (p < 0.001) Nonetheless, a diet rich in antioxidant phytophenols,
and 4.3 mm Hg for the DASH diet group (p < 0.001). The such as those found in fish, olive oil, red wine, and grape
study confirmed that hypertension control requires mul- juice, appears to inhibit endothelial adhesion molecule
tiple lifestyle changes including an appropriate eating plan. expression effectively, partly explaining the protection from
Epidemiological data suggested an association between atherosclerosis afforded by Mediterranean diets (Carluccio
dietary factors, particularly antioxidants, and cognition et al., 2003; Scarmeas et al., 2009a, 2009b; Lourida et al.,
(Deschamps et al., 2001). Similarities in the diets of patients 2013; Shen et al., 2015).
with AD and VaD have been reported in Japan (Otsuka et al.,
2002), with higher energy intake from fats, in particular poly- 60.7.6 CHRONIC INFLAMMATION
unsaturated fatty acids and decrease in antioxidant vitamins B,
C and carotene. However, in the Honolulu-Asia Aging Study, Markers of inflammation such as C-reactive protein (CRP)
midlife intakes of antioxidants, such as beta-carotene, flavo- are important predictors of atherosclerotic disease, par-
noids and vitamins E and C, did not modify late-life dementia ticularly in patients with diabetes (Rader, 2000). Chronic
risk, including AD and VaD (Laurin et al., 2004). Likewise, no infections, including periodontal disease and persistent
effect of fat intake in the development of dementia was found intracellular infection with Chlamydia pneumoniae have
in the Rotterdam study (Engelhart et al., 2002). been associated with increased risk of vascular events
Wald et al. (2002) concluded, based on the evidence (Kalayoglu et al., 2002).
from genetic and prospective studies, that the association
between increased homocysteine and cardiovascular dis-
ease is causal. Despite some disagreement (Homocysteine
Studies Collaboration, 2002), lowering homocysteine con- 60.8 PUBLIC HEALTH ASPECTS OF VaD
centrations by 3 µmol/L from current levels (achievable by
increasing folic acid intake) would reduce the risk of isch- Population ageing will lead to increasing incidence of stroke
aemic heart disease (IHD) by 16% (11%–20%), deep vein and heart disease. It is predicted that VaD will become the
thrombosis by 25% (8%–38%) and stroke by 24% (15%–33%). most common cause of senile dementia, both by itself and
Román (2015) observed a very high prevalence of MTHFR as a contributor to other degenerative dementias (Román,
mutations in a memory clinic population. DNA analyses of 2002e).
two MTHFR gene mutations (C667T and A1298C), includ- There is growing evidence that preventive measures to
ing homozygous and heterozygous polymorphisms, were decrease the vascular burden on the brain may also decrease
recorded in 92.5% of consecutive patients with late-onset VaD in the elderly. VaD and AD share with stroke a number
AD+CVD (Román, 2015). Elevation of total serum homo- of vascular risk factors. The most important of the modifi-
cysteine (tHcy) is considered a risk factor for dementia able factors is hypertension, which explains at least half of
(Hooshmand et al., 2010, 2012), particularly in association the attributable risk of stroke. As noted, three large RCTs
with low folate and vitamin B12 levels (Clarke et al., 1998). (Forette et al., 1998, 2002; Lithell et al., 2003; Tzourio et al.,
In the Framingham Study (Seshadri et al., 2002), elderly 2003) demonstrated that BP lowering has a significant effect
subjects (mean age, 76 years) with tHcy above 14 μmol/L in decreasing the risk of dementia, including VaD and
nearly doubled the risk of AD over a period of 8 years, even AD. Overall, these data suggest that interventions aiming
after adjustment for multiple factors. A review of cross- at reducing the level of vascular risk factors might prevent
sectional and prospective studies involving >46,000 sub- dementia. The expected benefit of these interventions could
jects (Smith, 2008) confirmed the association between high be estimated from data provided by epidemiological stud-
tHcy and cognitive deficit or dementia. However, elevation ies; however, there is a dearth of large population-based
of tHcy is affected by factors including age, diet, supple- controlled studies to demonstrate the efficacy of preven-
mentation of folic acid and B-group vitamins, smoking, tive interventions at Public Health level (Román, 2003b;
Helicobacter pylori infection and renal failure, among oth- Alperovitch et al., 2004; Williams, 2004). Prevention
ers. Nonetheless, combined treatment with folate and vita- appears to be the most promising avenue for decreasing the
mins B6 and B12 has been demonstrated effectively to halt incidence of the two most common forms of senile demen-
the conversion of MCI to AD (Smith et al., 2010; Douaud tia, AD and VaD dementia.
et al. 2013). These results disagree with a Cochrane review
(Malouf et al., 2003) that concluded that in older patients
with mild-to-moderate cognitive decline, supplementation
with 750 μg/day of folic acid, with or without B12, had no REFERENCES
beneficial effects on cognition or mood, although folic acid
plus vitamin B12 effectively reduced serum homocysteine Alexopoulos, G.S., Kiosses, D.N., Klimstra, S. et al. (2002).
concentrations. Along the same lines, a trial on the use of Clinical presentation of the depression-executive
vitamins to prevent recurrent stroke gave negative results dysfunction syndrome of late life. American Journal of
(Toole et al., 2004). Geriatric Psychiatry, 10: 98–106.
684 Dementia

Alladi, S., Panigrahi, M., Tripathi, M. et al.; for the Anstey, K.J., Cherbuin, N. and Herath, P.M. (2013).
Ischemic Stroke System (ISS) Trial. (2009). A Pilot Development of a new method for assessing global
Study Evaluating the Safety, Tolerability and risk of Alzheimer’s disease for use in population health
Effectiveness of the Ischemic Stroke System (ISS) approaches to prevention. Preventive Science, 14:
for Treatment of Subcortical Ischemic Vascular 411–421.
Disease. Barcelona, Spain: International Congress Applegate, W. B., Pressel, S., Wittes, J. et al. (1994).
on Vascular Dementia. Impact of the treatment of isolated systolic hyperten-
Almkvist, O., Darreh-Shori, T., Stefanova, E. et al. (2004). sion on behavioral variables. Results from the systolic
Preserved cognitive function after 12 months of treat- hypertension in the elderly program. Archives of
ment with rivastigmine in mild Alzheimer’s disease Internal Medicine, 154: 2154–2160.
in comparison with untreated AD and MCI patients. Auchus, A.P., Brashear, H.R., Salloway, S. et al. (2007).
European Journal of Neurology, 11: 253–261. Galantamine treatment of vascular dementia: A ran-
Alperovitch, A., Schwarzinger, M., Dufouil, C. et al. domized trial. GAL-INT-26 Study Group. Neurology,
(2004). Towards a prevention of dementia. Revue 69: 448–458.
Neurologique (Paris), 160: 256–260. Azuma, T., Nagai, Y., Saito, T. et al. (1999). The effect of
Álvarez-Sabín, J., Ortega, G., Jacas, C. et al. (2013). Long- dehydroepiandrosterone sulfate administration to
term treatment with citicoline may improve post- patients with multi-infarct dementia. Journal of the
stroke vascular cognitive impairment. Cerebrovascular Neurological Sciences, 162: 69–73.
Diseases, 35: 146–154. Babikian, V.L., Wolfe, N., Linn, R. et al. (1990). Cognitive
Álvarez-Sabín, J., Santamarina, E., Maisterra, O. et al. (2016). changes in patients with multiple cerebral infarcts.
Long-term treatment with citicoline prevents cogni- Stroke, 21: 1013–1018.
tive decline and predicts a better quality of life after a Ballard, C., Sauter, M., Scheltens, P. et al. (2008). Efficacy,
first ischemic stroke. International Journal of Molecular safety and tolerability of rivastigmine capsules in patients
Science, 16:17(3) pii: E390. with probable vascular dementia: The VantagE study.
Álvarez, X.A., Laredo, M., Corzo, D. et al. (1997). Citicoline Current Medical Research Opinion, 24: 2561–2574.
improves memory performances in elderly subjects. Barba, R., Castro, M.D., del Mar Morin, M. et al. (2001).
Methods and Findings in Experimental and Clinical Prestroke dementia. Cerebrovascular Diseases, 11:
Pharmacology, 19: 201–210. 216–224.
Álvarez-Sabín, J. and Román, G.C. (2011). Citicoline in Barba, R., Martinez-Espinosa, S., Rodriguez-Garcia, E.
vascular cognitive impairment and vascular dementia et al. (2000). Poststroke dementia: Clinical features
after stroke. Stroke, 42 (Suppl. 1): S40–S43. and risk factors. Stroke, 31: 1494–1501.
Álvarez-Sabín, J. and Román, G.C. (2013). The role of citi- Barnes, D.E. and Yaffe, K. (2011). The projected effect of
coline in neuroprotection and neurorepair in ischemic risk factor reduction on Alzheimer’s disease preva-
stroke. Brain Sciences, 3: 1395–1414. lence. The Lancet Neurology, 10: 819–828.
Amenta, F., Tomassoni, D., Traini, E. et al. (2008). Batysheva, T.T., Bagir, L.V., Kostenko, E.V. et al. (2009).
Nicardipine: A hypotensive dihydropyridine-type Experience of the out-patient use of memotropil
calcium antagonist with a peculiar cerebrovascular in the treatment of cognitive disorders in patients
profile. Clinical and Experimental Hypertension, 30: with chronic progressive cerebrovascular disorders.
808–826. Neuroscience and Behavioral Physiology, 39: 193–197.
Amenta, F., Lanari, A., Mignini, F., et al. (2009). Nicardipine Baumel, B., Eisner, L., Karukin, M. et al. (1989). Oxiracetam
use in cerebrovascular disease: A review of controlled in the treatment of multi-infarct dementia. Progress in
clinical studies. Journal of the Neurological Sciences, Neuropsychopharmacology and Biological Psychiatry,
283: 219–223. 13: 673–682.
American Diabetes Association (2014). Standards of Behl, P., Bocti, C., Swartz, R.H., Gao, F., Sahlas, D.J.
medical care in diabetes – 2014. Diabetes Care, 37: et al. (2007). Strategic subcortical hyperintensities in
S14–S80. cholinergic pathways and executive function decline
American Psychiatric Association (1994). Diagnostic and in treated Alzheimer patients. Archives of Neurology,
Statistical Manual of Mental Disorders, Fourth Edition 64: 266–272.
(DSM-IV). Washington, DC: American Psychiatric Benavente, O.R., Pearce, L.A., Bazan, C. et al.; for the SPS3
Association. Investigators. (2014). Clinical–MRI correlations in a
American Psychiatric Association. (2013). Diagnostic and multiethnic cohort with recent lacunar stroke: The SPS3
Statistical Manual of Mental Disorders, Fifth Edition trial. International Journal of Stroke, 9: 1057–1064.
(DSM-5). Washington, DC: American Psychiatric Benavente, O.R., White, C.L., Pearce, L., et al.; for the
Publishing. SPS3 Investigators. (2011). The secondary prevention
Andreozzi, G.M. (2014). Role of sulodexide in the treat- of small subcortical strokes (SPS3) study. International
ment of CVD. International Angiology, 33: 255–262. Journal of Stroke, 6: 164–175.
Therapeutic strategies for vascular cognitive disorders 685

Bennett, D.A., Wilson, R.S., Arvanitakis, Z. et al. (2013). Bousser, M.G. and Tournier-Lasserve, E. (1994). Summary
Selected findings from the Religious Orders Study and of the proceedings of the First International Workshop
Rush Memory and Aging Project. Journal of Alzheimer on CADASIL. Paris, May 19–21, 1993. Stroke, 25:
Disease, 33: S397–S403. 704–707.
Berg, L. (1988). Clinical dementia rating (CDR). Brainin, M., Tuomilehto, J., Heiss, W.D. et al.; for the Post
Psychopharmacology Bulletin, 24: 637–639. Stroke Cognition Study Group (2015). Post-stroke cog-
Bes, A., Orgogozo, J.M., Poncet, M. et al. (1999). A nitive decline: An update and perspectives for clinical
24-month, double-blind, placebo-controlled multicentre research. European Journal of Neurology, 22: 229–238.
pilot study of the efficacy and safety of nicergoline 60 Brookmeyer, R., Johnson, E., Ziegler-Graham, K. and
mg per day in elderly hypertensive patients with leuko- Arrighi, H.M. (2007). Forecasting the global burden of
araiosis. European Journal of Neurology, 6: 313–322. Alzheimer’s disease. Alzheimer’s & Dementia: Journal
Bicikova, M., Ripova, D., Hill, M. et al. (2004). Plasma levels of the Alzheimer’s Association, 3: 186–191.
of 7-hydroxylated dehydroepiandrosterone (DHEA) Brown, W.R., Moody, D.M., Thore, C.R. et al. (2009).
metabolites and selected amino-thiols as discriminatory Microvascular changes in the white mater in dementia.
tools of Alzheimer’s disease and vascular dementia. Journal of the Neurological Sciences, 283: 28–31.
Clinical Chemistry Laboratory Medicine, 42: 518–524. Brown, L.A., Riby, L.M. and Reay, J.L. (2010).
Biesold, D., Inanami, O., Sato, A. and Sato, Y. (1989). Supplementing cognitive aging: A selective review of
Stimulation of the nucleus basalis of Meynert increases the effects of Ginkgo biloba and a number of everyday
cerebral cortical blood flow in rats. Neuroscience nutritional substances. Experimental Aging Research,
Letters, 98: 39–44. 36: 105–122.
Binswanger, O. (1894). Die Abgrenzung des allgemeinen Brun, A. (1994). Pathology and pathophysiology of cere-
progressive paralyse. Berlin Klinische Wochenschrift, brovascular dementia: Pure subgroups of obstructive
31: 1103–1105, 1137–1139, 1180–1186. and hypoperfusive etiology. Dementia, 5: 145–147.
Birkenhäger, W.H. and Staessen, J.A. (2006). Cognitive Brun, A. and Englund, E. (1988). A white matter demen-
function in essential hypertension. Progress in tia of the Alzheimer type: A pathoanatomical study.
Cardiovascular Diseases, 49: 1–10. Annals of Neurology, 19: 253–262.
Birks, J., McGuinness, B. and Craig, D. (2013). Rivastigmine Bruno, R. de B., Marques, T.F., Batista, T.M. et al. (2009).
for vascular cognitive impairment. Cochrane Database Pentoxifylline treatment improves neurological and
of Systematic Reviews, (5): CD004744. neurochemical deficits in rats subjected to transient
Black, S.E. (2007). Therapeutic issues in vascular dementia: brain ischemia. Brain Research, 1260: 55–64.
Studies, designs and approaches. Canadian Journal of Bustamante, A., Giralt, D., García-Bonilla, L. et al. (2012).
Neurological Sciences, 34 (Suppl. 1): S125–S130. Citicoline in pre-clinical animal models of stroke: A
Black, R.S., Barclay, L.L., Nolan, K.A. et al. (1992). meta-analysis shows the optimal neuroprotective
Pentoxifylline in cerebrovascular dementia. Journal of profile and the missing steps for jumping into a stroke
the American Geriatrics Society, 40: 237–244. clinical trial. Journal of Neurochemistry, 123: 217–225.
Black, S., Gao, F.Q. and Bilbao, J. (2009). Understanding Cacabelos, R., Álvarez, X.A., Franco-Maside, A.
white matter disease: Imaging-pathological correla- et al. (1993). Effect of CDP-choline on cognition and
tions in vascular cognitive impairment. Stroke, 40: immune function in Alzheimer’s disease and multi-
S48–S52. infarct dementia. Annals of the New York Academy of
Black, S., Román, G.C., Geldmacher, D.S. et al.; for the Sciences, 695: 321–323.
Donepezil 307 Vascular Dementia Study Group (2003). Carluccio, M.A., Siculella, L., Ancora, M.A. et al. (2003).
Efficacy and tolerability of donepezil in vascular Olive oil and red wine antioxidant polyphenols inhibit
dementia. Positive results of a 24-week, multicenter, endothelial activation: Antiatherogenic properties of
international, randomized, placebo-controlled clinical Mediterranean diet phytochemicals. Arteriosclerosis,
trial. Stroke, 34: 2323–2332. Thrombosis, and Vascular Biology, 23: 622–629.
Bocti, C., Swartz, R.H., Gao, F.Q. et al. (2005). A new visual Chabriat, H. and Bousser, M.G. (2003). CADASIL. Cerebral
rating scale to assess strategic white matter hyper- autosomal dominant arteriopathy with subcorti-
intensities within cholinergic pathways in dementia. cal infarcts and leukoencephalopathy. Advances in
Stroke, 36: 2126–2131. Neurology, 92: 147–150.
Bosch, J., Yusuf, S., Pogue, J. et al.; for the HOPE Chabriat, H., Joutel, A., Dichgans, M. et al. (2009).
Investigators (2002). Heart outcomes prevention evalu- CADASIL. Lancet Neurology, 8: 643–653.
ation. Use of ramipril in preventing stroke: Double blind Chen, N., Yang, M., Guo, J. et al. (2013). Cerebrolysin for
randomised trial. British Medical Journal, 324: 699–702. vascular dementia. Cochrane Database of Systematic
Bots, M.L., Breteler, M.M., van Kooten, F. et al. (1998). Reviews, (1): CD008900.
Coagulation and fibrinolysis markers and risk of Chobanian, A.V., Bakris, G.L., Black, H.R. et al. for the
dementia. The Dutch Vascular Factors in Dementia Joint National Committee on Prevention, Detection,
Study. Haemostasis, 28: 216–222. Evaluation, and Treatment of High Blood Pressure.
686 Dementia

National Heart, Lung, and Blood Institute; National Court, J.A., Perry, E.K. and Kalaria, R.N. (2002).
High Blood Pressure Education Program Coordinating Neurotransmitter control of the cerebral vasculature
Committee (2003). Seventh report of the Joint and abnormalities in vascular dementia. In Erkinjuntti, T.
National Committee on Prevention, Detection, and Gauthier, S. (eds.), Vascular Cognitive Impairment.
Evaluation, and Treatment of High Blood Pressure. London, United Kingdom: Martin Duniz Ltd., pp. 167–185.
Hypertension, 42: 1206–1252. Craig, D. and Birks, J. (2006). Galantamine for vascular
Chui, H.C., Mack, W., Jackson, J.E. et al. (2000). Clinical cognitive impairment. Cochrane Database of
criteria for the diagnosis of vascular dementia: A multi- Systematic Reviews, (1): CD004746.
center study of comparability and interrater reliability. Cummings, J.L. (1993). Frontal-subcortical circuits and
Archives of Neurology, 57: 191–196. human behavior. Archives of Neurology, 50: 873–880.
Chui, H.C., Victoroff, J.I., Margolin, D. et al. (1992). Cummings, J.L., Mega, M., Gray, K. et al. (1994). The
Criteria for the diagnosis of ischemic vascular Neuropsychiatric Inventory. Comprehensive assess-
­dementia proposed by the State of California ment of psychopathology in dementia. Neurology, 44:
Alzheimer’s Disease Diagnostic and Treatment 2308–2314.
Centers. Neurology, 42: 473–480. Darreh-Shori, T., Almkvist, O., Guan, Z.Z. et al. (2002).
Chui, H.C., Zarow, C., Mack. W.J. et al. (2006). Cognitive Sustained cholinesterase inhibition in AD patients
impact of subcortical vascular and Alzheimer’s disease receiving rivastigmine for 12 months. Neurology, 59:
pathology. Annals of Neurology, 60: 677–687. 563–572.
Chukwudelunzu, F.E., Meschia, J.F., Graff-Radford, N.R. Dávalos, A., Álvarez-Sabín, J., Castillo, J. et al.; for the
and Lucas, J.A. (2001). Extensive metabolic and neu- International Citicoline Trial on acUte Stroke (ICTUS)
ropsychological abnormalities associated with discrete Trial Investigators (2012). Citicoline in the treatment of
infarction of the genu of the internal capsule. Journal of acute ischaemic stroke: An international, randomised,
Neurology, Neurosurgery, and Psychiatry, 71: 658–662. multicentre, placebo-controlled study (ICTUS trial).
Clarke, C.E. (1999). Does the treatment of isolated Lancet, 380: 349–357.
hypertension prevent dementia? Journal of Human Dávalos, A., Castillo, J, Alvarez-Sabin, J. et al. (2002).
Hypertension, 13: 357–358. Oral citicoline in acute ischemic stroke: An individual
Clarke, R., Smith, A.D., Jobst, K.A. et al. (1998). Folate, patient data pooling analysis of clinical trials. Stroke,
vitamin B12, and serum total homocysteine levels in 33: 2850–2857.
confirmed Alzheimer disease. Archives of Neurology, De Carli, C. (2012). Clinically asymptomatic vascular brain
55: 1449–1455. injury: A potent cause of cognitive impairment among
Cochrane, A.L. (1979). Concluding remarks. In Tognoni, older individuals. Journal of Alzheimer’s Disease, 33
G. and Garattini, S. (eds.), Treatment and Prevention (Suppl. 1): S417–S426.
in Cerebrovascular Disorders. Amsterdam, The de Jager, C.A., Oulhaj, A., Jacoby, R. et al. (2012).
Netherlands: Elsevier, pp. 453–455. Cognitive and clinical outcomes of homocysteine-
Cohen, R.A., Browndyke, J.N., Moser, D.J. et al. (2003). lowering B-vitamin treatment in mild cognitive impair-
Long-term citicoline (cytidine diphosphate choline) ment: A randomized controlled trial. International
use in patients with vascular dementia: Neuroimaging Journal of Geriatric Psychiatry, 27: 592–600.
and neuropsychological outcomes. Cerebrovascular De Jong, G.I., de Weerd, H., Schuurman, T. et al. (1990).
Diseases, 16: 199–204. Microvascular changes in aged rat forebrain. Effects of
Conant, R. and Schauss, A.G. (2004). Therapeutic applica- chronic nimodipine treatment. Neurobiology of Aging,
tions of citicoline for stroke and cognitive dysfunction 11: 381–389.
in the elderly: A review of the literature. Alternative Delay, J. and Brion, S. (1962). Les Démences Tardives.
Medicine Review, 9: 17–31. Paris, France: Masson et Cie.
Cook, P. and James, I. (1981). Drug therapy: Cerebral Deschamps, V., Barberger-Gateau, P., Peuchant, E.
vasodilators. New England Journal of Medicine, 305: and Orgogozo, J.M. (2001). Nutritional factors
1508–1513. in cerebral aging and dementia: Epidemiological
Cordonnier, C., Hénon, H., Derambure, P. et al. (2007). arguments for a role of oxidative stress.
Early epileptic seizures after stroke are associated Neuroepidemiology, 20: 7–15.
with increased risk of new-onset dementia. Journal of Desmond, D.W., Moroney, J.T., Bagiella, E. et al. (1998).
Neurology Neurosurgery and Psychiatry, 78: 514–516. Dementia as a predictor of adverse outcomes fol-
Corey-Bloom, J. (2003). Galantamine: A review of its lowing stroke. An evaluation of diagnostic methods.
use in Alzheimer’s disease and vascular dementia. Stroke, 29: 69–74.
International Journal of Clinical Practice, 57: 219–223. Dichgans, M. (2002). Cerebral autosomal dominant
Cotroneo, A.M., Castagna, A., Putignano, S. et al. (2013). arteriopathy with subcortical infarcts and leukoen-
Effectiveness and safety of citicoline in mild vascu- cephalopathy: Phenotypic and mutational spectrum.
lar cognitive impairment: The IDEALE study. Clinical Journal of the Neurological Sciences, 203–204:
Interventions in Aging, 8: 131–137. 77–80.
Therapeutic strategies for vascular cognitive disorders 687

Dichgans,-M., Markus,-H.S., Salloway,-S. et al. (2008). Erkinjuntti, T. and Sawada, T., (eds.) (1999). Summary of
Donepezil in patients with subcortical vascular cogni- the 1st International Conference on Development
tive impairment: A randomised double-blind trial in of Drug Treatment for Vascular Dementia, by the
CADASIL. Lancet Neurology, 7: 310–318. International Working Group on Harmonization of
Diener, H.C. (2009). Preventing stroke: The PRoFESS, Dementia Drug Guidelines, October 7–9, 1998, Osaka,
ONTARGET, and TRANSCEND trial programs. Journal Japan. Alzheimer Disease and Associated Disorders,
of Hypertension, 27 (Suppl. 5): S31–S36. 13 (Suppl. 3): S1–S212.
Diener, H.C., Sacco, R.L., Yusuf, S. et al.; for the Prevention Erkinjuntti, T., Sawada, T. and Whitehouse, P.J. (1999).
Regimen for Effectively Avoiding Second Strokes Osaka conference on vascular dementia 1998.
(PRoFESS) Study Group (2008). Effects of aspirin plus Alzheimer Disease and Associated Disorders, 13
extended-release dipyridamole versus clopidogrel (Suppl. 3): S1–S3.
and telmisartan on disability and cognitive function Erkinjuntti, T., Skoog, I., Lane, R. and Andrews, C. (2002b)
after recurrent stroke in patients with ischaemic stroke Rivastigmine in patients with Alzheimer’s disease
PRoFESS trial: A double-blind, active and placebo- and concurrent hypertension. International Journal of
controlled study. Lancet Neurology, 7: 875–884. Clinical Practice, 56: 791–796.
Douaud, G., Refsum, H., de Jager, C.A. et al. (2013). Erkinjuntti, T., Skoog, I., Lane, R. and Andrews, C. (2003b).
Preventing Alzheimer’s disease-related gray matter Potential long-term effects of rivastigmine on disease
atrophy by B-vitamin treatment. Proceedings of the progression may be linked to drug effects on vascular
National Academy of Sciences of the United States of changes in Alzheimer brains. International Journal of
America, 110: 9523–9528. Clinical Practice, 57: 756–760.
Dysken, M.W., Katz, R., Stallone, F. and Kuskowski, M. European pentoxifylline multi-infarct dementia study
(1989). Oxiracetam in the treatment of multi-infarct (1996). European Neurology, 36: 315–321.
dementia and primary degenerative dementia. Journal Evans, J.L.K., Goldfine, I.D., Maddux, B.A. and Grodsky,
of Neuropsychiatry and Clinical Neurosciences, 1 (3): G.M. (2002). Oxidative stress and stress-activated
249–252. signaling pathways: A unifying hypothesis of type 2
Easton, D. (2003). Evidence with antiplatelet therapy and diabetes. Endocrine Reviews, 23: 599–622.
ADP-receptor antagonists. Cerebrovascular Diseases, Farlow, M.R. (2004). NMDA receptor antagonists.
16 (Suppl. 1): 20–26. A new therapeutic approach for Alzheimer’s disease.
Engelhart, M.J., Geerlings, M.I., Ruitenberg, A. et al. Geriatrics, 59: 22–27.
(2002). Diet and risk of dementia: Does fat matter? The Farooq, U. and Ray, S.G. (2015). 2014 Guideline for the
Rotterdam Study. Neurology, 59: 1915–1921. management of high blood pressure (Eighth Joint
Englund, E., Brun, A. and Alling, C. (1988). White mat- National Committee): Take-home messages. Medical
ter changes in dementia of the Alzheimer’s type. Clinics of North America, 99: 733–738.
Biochemical and neuropathological correlates. Brain, Ferris, S. and Gauthier, S. (2002). Cognitive outcome mea-
111: 1425–1439. sures in vascular dementia. In Erkinjuntti, T and Gauthier,
Erkinjuntti, T. and Gauthier, S. (eds.) (2002). Vascular S. (eds.), Vascular Cognitive Impairment. London, United
Cognitive Impairment. London, United Kingdom: Kingdom: Martin Duniz Ltd., pp. 395–400.
Martin Duniz Ltd., pp. 395–400. Fioravanti, M., Nakashima, T., Xu, J. and Garg, A. (2014). A
Erkinjuntti, T., Gauthier, S., Bullock, R. et al. (2008). systematic review and meta-analysis assessing adverse
Galantamine treatment in Alzheimer’s disease with event profile and tolerability of nicergoline. British
cerebrovascular disease: Responder analyses from a Medical Journal Open, 4: e005090.
randomized, controlled trial (GAL-INT-6). Journal of Fioravanti, M. and Flicker, L. (2001). Efficacy of nicergoline
Psychopharmacology, 22: 761–768. in dementia and other age associated forms of cogni-
Erkinjuntti, T., Inzitari, D., Pantoni, L. et al. (2000). tive impairment. Cochrane Database of Systematic
Research criteria for subcortical vascular dementia Reviews, (4): CD003159.
in clinical trials. Journal of Neural Transmission, 59 Fioravanti, M. and Yanagi, M. (2005). Cytidine diphospho-
(Suppl. 1): 23–30. choline (CDP choline) for cognitive and behavioural
Erkinjuntti, T., Kurz, A., Gauthier, S. et al. (2002a). Efficacy disturbances associated with chronic cerebral disor-
of galantamine in probable vascular dementia and ders in the elderly. Cochrane Database of Systematic
Alzheimer’s disease combined with cerebrovascular Reviews, (2): CD000269.
disease: A randomized trial. Lancet, 359: 1283–1290. Fisher, C.M. (1965). Lacunes: Small deep cerebral infarcts.
Erkinjuntti, T., Kurz, A., Small, G.W. et al. (2003a). An Neurology, 15: 774–784.
open-label extension trial of galantamine in patients Flaker, G.C., Pogue, J., Yusuf, S. et al.; for the Atrial
with probable vascular dementia and mixed dementia. Fibrillation Clopidogrel Trial with Irbesartan for
Clinical Therapeutics, 25: 1765–1782. Prevention of Vascular Events (ACTIVE) Investigators
Erkinjuntti, T., Román, G., Gauthier, S. et al. (2004). (2010). Cognitive function and anticoagulation con-
Emerging therapies for vascular dementia and vascular trol in patients with atrial fibrillation. Circulation and
cognitive impairment. Stroke, 35: 1010–1017. Cardiovascular Quality Outcomes, 3: 277–283.
688 Dementia

Flicker, L. and Grimley Evans, G. (2001). Piracetam for Gold, G., Bouras, C., Canuto, A. et al. (2002).
dementia or cognitive impairment. Cochrane Database Clinicopathological validation study of four sets of
of Systematic Reviews, (2): CD001011. clinical criteria for vascular dementia. American Journal
Fogari, R., Mugellini, A., Zoppi, A. et al. (2006). Effect of of Psychiatry, 159: 82–87.
telmisartan/hydrochlorothiazide vs. lisinopril/hydro- Gold, G., Giannakopoulos, P., Montes-Paixao Junior, C.
chlorothiazide combination on ambulatory blood et al. (1997). Sensitivity and specificity of newly pro-
pressure and cognitive function in elderly hyperten- posed clinical criteria for possible vascular dementia.
sive patients. Journal of Human Hypertension, 20: Neurology, 49: 690–694.
177–185. Goldsmith, D.R. and Scott, L.J. (2003). Donepezil in vascu-
Folstein, M.F., Folstein, S.E. and McHugh, P.R. (1975). lar dementia. Drugs and Aging, 20: 1127–1136.
“Mini-mental state”: A practical method for grading Gómez-Isla T, Blesa R, Boada M, et al.; for the TRIMCI
the cognitive state of patients for the clinician. Journal Study Group (2008). A randomized, double-blind,
of Psychiatric Research, 12: 189–198. placebo controlled-trial of triflusal in mild cognitive
Fong, K.C. and Kinnear, P.E. (2003). Retrobulbar haemor- impairment: The TRIMCI study. Alzheimer Disease and
rhage associated with chronic Ginkgo biloba ingestion. Associated Disorders, 22: 21–29.
Postgraduate Medical Journal, 79: 531–532. González-González, J.A. and Lozano, R.A. (2000).
Forette, F., Seux, M.L., Staessen, J.A. et al.; for the Syst- Estudio sobre la tolerabilidad y efectividad del
Eur Investigators. (1998). Prevention of dementia in nimodipino retard en el deterioro cognitivo de ori-
randomised double-blind placebo-controlled Systolic gen vascular [Study of the tolerability and effective-
Hypertension in Europe (Syst-Eur) trial. Lancet, 352: ness of nicardipine retard in cognitive deterioration
1347–1351. of vascular origin]. Revista de Neurología,
Forette, F., Seux, M.L., Staessen, J.A. et al.; for the 30: 719–728.
­Syst-Eur investigators (2002). The prevention of Gorelick, P.B., Chatterjee, A., Patel, D. et al. (1992). Cranial
dementia with antihypertensive treatment: New computed tomographic observations in multi-infarct
evidence from the Systolic Hypertension in Europe dementia: A controlled study. Stroke, 23: 804–811.
(Syst-Eur) study. Archives of Internal Medicine, Gorelick, P.B., Goldstein, L.B., Ovbiagele, B. (2014). New
162: 2046–2052. guidelines to reduce risk of atherosclerotic cardio-
Fotuhi, M., Hachinski, V. and Whitehouse, P.J. (2009). vascular disease: Implications for stroke prevention in
Changing perspectives regarding late-life dementia. 2014. Stroke, 45: 945–947.
Nature Reviews Neurology, 5: 649–658. Gorelick, P.B., Scuteri, A., Black, S.E. et al.; for the
Frishman, W.H. (2002). Are antihypertensive agents American Heart Association Stroke Council, Council
protective against dementia? A review of clinical and on Epidemiology and Prevention, Council on
preclinical data. Heart Diseases, 4: 380–386. Cardiovascular Nursing, Council on Cardiovascular
Gauthier, S., Proaño, J.V., Jia, J. et al. (2015). Cerebrolysin Radiology and Intervention, and Council on
in mild-to-moderate Alzheimer’s disease: A meta- Cardiovascular Surgery and Anesthesia (2011).
analysis of randomized controlled clinical trials. Vascular contributions to cognitive impairment and
Dementia and Geriatric Cognitive Disorders, 39: dementia: A statement for healthcare professionals
332–347. from the American Heart Association/American Stroke
Gélinas, I., Gauthier, L., McIntyre, M. and Gauthier, S. Association. Stroke, 42: 2672–2713.
(1999). Development of a functional measure for per- Gottfries, C.G., Blennow, K., Karlsson, I. and Wallin, A.
sons with Alzheimer’s disease: The disability assess- (1994). The neurochemistry of vascular dementia.
ment for dementia. American Journal of Occupational Dementia, 5: 163–167.
Therapy, 53: 471–481. Gottfries, C.G., Bråne, G. and Steen, G. (1982). A new
Gelmers, H.J. and Hennerici, M. (1990). Effect of rating scale for dementia syndromes. Gerontology, 28
nimodipine on acute ischemic stroke. Pooled results (Suppl. 2): 20–31.
from five randomized trials. Stroke, 21 (Suppl. 12): Gray, F., Dubas, F., Roullet, E. and Escourolle, R. (1985).
IV81–IV84. Leukoencephalopathy in diffuse hemorrhagic cerebral
Gertz, H.J. and Kiefer, M. (2004). Review about Ginkgo amyloid angiopathy. Annals of Neurology, 18: 54–59.
biloba special extract EGb 761 (Ginkgo). Current Green, R.C., Goldstein, F.C., Auchus, A.P. et al. (1992).
Pharmaceutical Design, 10: 261–264. Treatment trial of oxiracetam in Alzheimer’s disease.
Giannopoulos, S., Katsanos, A.H., Kosmidou, M. and Archives of Neurology, 49: 1135–1136.
Tsivgoulis, G. (2014). Statins and vascular dementia: A Greenberg, J.H., Uematsu, D., Araki, N. et al. (1990).
review. Journal of Alzheimer’s Disease, 42 (Suppl. 3): Cytosolic free calcium during focal cerebral ischemia
S315–S320. and the effects of nimodipine on calcium and histo-
Giardino, L., Giuliani, A., Battaglia, A. et al. (2002). logical damage. Stroke, 21 (Suppl. 2): IV72–IV77.
Neuroprotection and aging of the cholinergic system: Grundy, S.M., Cleeman, J.I., Merz, C.N. et al.; for the
A role for the ergoline derivative nicergoline (Sermion). Coordinating Committee of the National Cholesterol
Neuroscience, 109: 487–497. Education Program (2004). Implications of recent
Therapeutic strategies for vascular cognitive disorders 689

clinical trials for the National Cholesterol Education Hooshmand, B., Solomon, A., Kåreholt, I. et al. (2010).
Program Adult Treatment Panel III guidelines. Homocysteine and holotranscobalamin and the risk of
Circulation, 110: 227–239. Alzheimer disease: A longitudinal study. Neurology,
Grupo Español de Estudio de Nicardipino en Demencia 75: 1408–1414.
Vascular (1999). Ensayo clínico experimental, doble- Hooshmand, B., Solomon, A., Kåreholt, I. et al. (2012).
mente ciego, aleatorizado, controlado con placebo, Associations between serum homocysteine, holotransco-
del efecto de nicardipino en la función cognitiva en balamin, folate and cognition in the elderly: A longitudi-
pacientes afectos de demencia vascular [Spanish group nal study. Journal of Internal Medicine, 271: 204–212.
of nicardipine study in vascular dementia. An experi- Huppert, F.A., Brayne, C., Gill, C. et al. (1995). CAMCOG
mental, randomized, double-blind, placebo-controlled – A concise neuropsychological test to assist demen-
clinical trial to investigate the effect of nicardipine on tia diagnosis: Sociodemographic characteristics in an
cognitive function in patients with vascular dementia.] elderly population sample. British Journal of Clinical
Revista de Neurología, 28: 835–845. Psychology, 34: 529–541.
Gutiérrez-Fernández, M., Rodríguez-Frutos, B., Fuentes, Hurtado, O., Cárdenas, A., Pradillo, J.M. et al. (2007).
B. et al. (2012). CDP-choline treatment induces brain A chronic treatment with CDP-choline improves
plasticity markers expression in experimental animal functional recovery and increases neuronal plasticity
stroke. Neurochemistry International, 60: 310–317. after experimental stroke. Neurobiology of Disease,
Hachinski, V., Iadecola, C., Petersen, R.C. et al. (2006). 26: 105–111.
National Institute of Neurological Disorders and Stroke– Hurtado, O., Hernández-Jiménez, M., Zarruk, J.G. et al.
Canadian Stroke Network vascular cognitive impair- (2013). Citicoline (CDP-choline) increases Sirtuin1 expres-
ment harmonization standards. Stroke, 37: 2220–2241. sion concomitant to neuroprotection in experimental
Hachinski, V.C., Iliff, L.D., Zilhka, E. et al. (1975). Cerebral stroke. Journal of Neurochemistry, 126: 819–826.
blood flow in dementia. Archives of Neurology, 32: Hurtado, O., Pradillo, J.M., Fernández-López, D. et al.
632–637. (2008). Delayed post-ischemic administration of CDP-
Hanon, O., Berrou, J.P., Negre-Pages, L. et al. (2008). choline increases EAAT2 association to lipid rafts
Effects of hypertension therapy based on eprosar- and affords neuroprotection in experimental stroke.
tan on systolic arterial blood pressure and cognitive Neurobiology of Disease, 29: 123–131.
function: Primary results of the observational study on Ingles, J.L., Wentzel, C., Fisk, J.D. and Rockwood, K.
cognitive function and systolic blood pressure reduc- (2002). Neuropsychological predictors of incident
tion open-label study. Journal of Hypertension, 26: dementia in patients with vascular cognitive impair-
1642–1650. ment, without dementia. Stroke, 33: 1999–2002.
Hart, R.G. (2003). Atrial fibrillation and stroke prevention. Itil, T.M., Menon, G.N., Songar, A. and Itil, K.Z. (1986).
New England Journal of Medicine, 349: 1015–1016. CNS pharmacology and clinical therapeutic effects of
Hennerici, M.G. (2009). What are the mechanisms of post- oxiracetam. Clinical Neuropharmacology, 9 (Suppl. 3):
stroke dementia? Lancet Neurology, 8: 973–975. S70–S72.
Henninger, N. and Fisher, M. (2007). Stimulating circle Itoh, T., Shimada, Y. and Terasawa, K. (1999). Efficacy of
of Willis nerve fibers preserves the diffusion-perfu- Choto-san on vascular dementia and the protective
sion mismatch in experimental stroke. Stroke, 38: effect of the hooks and stems of Uncaria sinensis on
2779–2786. glutamate-induced neuronal death. Mechanisms of
Hénon, H., Durieu, I., Guerouaou, D. et al. (2001). Ageing and Development, 111: 155–173.
Poststroke dementia: Incidence and relationship to Ivan, C.S., Seshadri, S., Beiser, A. et al. (2004). Dementia
prestroke cognitive decline. Neurology, 57: 1216–1222. after stroke: The Framingham Study. Stroke, 35:
Hénon, H., Pasquier, F., Durieu, I. et al. (1997). Preexisting 1264–1268.
dementia in stroke patients. Baseline frequency, asso- Iwamoto, T., Kubo, H. and Takasaki, M. (1995). Platelet
ciated factors, and outcome. Stroke, 28: 2429–2436. activation in the cerebral circulation in different sub-
Herrmann, W.M., Stephan, K., Gaede, K. and Apeceche, types of ischemic stroke and Binswanger’s disease.
M. (1997). A multicenter randomized double-blind Stroke, 26: 52–56.
study on the efficacy and safety of nicergoline in Jacova, C., Pearce, L.A., Costello, R. et al. (2012).
patients with multi-infarct dementia. Dementia and Cognitive impairment in lacunar strokes: The SPS3
Geriatric Cognitive Disorders, 8: 9–17. trial. Annals of Neurology, 72: 351–362.
Hitzenberger, G., Rameis, H. and Manigley, C. (1998). Jacova, C., Pearce, L.A., Roldan, A.M. et al. (2015).
Pharmacological properties of piracetam. Central Cognitive performance following lacunar stroke in
Nervous System Drugs, 9 (Suppl. 1): 19–27. Spanish-speaking patients: Results from the SPS3 trial.
Homocysteine Studies Collaboration (2002). International Journal of Stroke, 10: 519–528.
Homocysteine and risk of ischemic heart disease James, P.A., Oparil, S., Carter, B.L. et al. (2014). 2014
and stroke: A meta-analysis. JAMA: Journal of the Evidence-based guideline for the management of
American Medical Association, 288: 2015–2022. high blood pressure in adults. Report from the panel
690 Dementia

members appointed to the Eight Joint National Kamei, S., Oishi, M. and Takasu, T. (1996). Evaluation
Committee (JNC 8). JAMA: Journal of the American of fasudil hydrochloride treatment for wandering
Medical Association, 311: 507–520. ­symptoms in cerebrovascular dementia with
Jansen, I., Tfelt-Hansen, P. and Edvinsson, L. (1991). 31P-magnetic resonance spectroscopy and
Comparison of the calcium entry blockers nimodipine Xe-computed tomography. Clinical Neuropharmacology,
and flunarizine on human cerebral and temporal arter- 19: 428–438.
ies: Role in cerebral disorders. European Journal of Kanowski, S. and Hoerr, R. (2003). Ginkgo biloba extract
Clinical Pharmacology, 40: 7–15. EGb 761 in dementia: Intent-to-treat analyses of
January, C.T., Wann, L.S., Alpert, J.S. et al. (2014). AHA/ a 24-week, multi-center, double-blind, placebo-
ACC/HRS guideline for the management of patients controlled, randomized trial. Pharmacopsychiatry, 36:
with atrial fibrillation: A report of the American 297–303.
College of Cardiology/American Heart Association Karpe, F. and Frayn, K.N. (2004). The nicotinic acid recep-
Task Force on Practice Guidelines and the Heart tor – A new mechanism for an old drug. Lancet, 363:
Rhythm Society. Journal of the American College of 1892–1894.
Cardiology, 64: e1–e76. Kavirajan, H. and Schneider, L.S. (2007). Efficacy and
Jefferson, A.L., Beiser, A.S., Himali, J.J., et al. (2015a). Low adverse effects of cholinesterase inhibitors and
cardiac index is associated with incident dementia memantine in vascular dementia: A meta-analysis of
and Alzheimer disease: The Framingham Heart Study. randomized controlled trials. Lancet Neurology, 6:
Circulation, 131: 1333–1339. 782–792.
Jefferson, A.L., Hohman, T.J., Liu, D., et al. (2015b). Adverse Khachaturian, A.S., Zandi, P.P., Lyketsos, C.G. et al.
vascular risk is related to cognitive decline in older (2006). Antihypertensive medication use and incident
adults. Journal of Alzheimer’s Disease, 44: 1361–1373. Alzheimer disease: The Cache County Study. Archives
Jellinger, K.A. and Mitter-Ferstl, E. (2003). The impact of Neurology, 63: 686–692.
of cerebrovascular lesions in Alzheimer disease – A Khurana, D., Kaul, S. and Bornstein, N.M.; for the
comparative autopsy study. Journal of Neurology, 250: ImpACT-1 Study Group. (2009). Implant for augmenta-
1050–1055. tion of cerebral blood flow trial 1: A pilot study evalu-
Jick, H., Zornberg, G.L., Jick, S.S. et al. (2000). Statins and ating the safety and effectiveness of the Ischaemic
the risk of dementia. Lancet, 356: 1627–1631. Stroke System for treatment of acute ischaemic stroke.
Johnston, S.C., O’Meara, E.S., Manolio, T.A. et al. (2004). International Journal of Stroke, 4: 480–485.
Cognitive impairment and decline are associated with Kilander, L., Nyman, H., Boberg, M. et al. (1998).
carotid artery disease in patients without clinically Hypertension is related to cognitive impairment:
evident cerebrovascular disease. Annals of Internal A 20-year follow-up of 999 men. Hypertension, 31:
Medicine, 140: 237–247. 780–786.
Julius, S., Nesbitt, S.D., Egan, B.M. et al. (2006). Feasibility Kim, S.B., Hill, M., Kwak, Y.T. et al. (2003). Neurosteroids:
of treating prehypertension with an angiotensin- Cerebrospinal fluid levels for Alzheimer’s disease and
receptor blocker. The New England Journal of vascular dementia diagnostics. Journal of Clinical
Medicine, 354: 1685–1697. Endocrinology and Metabolism, 88: 5199–5206.
Kalaria, R.N. and Ballard, C. (1999). Overlap between Kittner, B. (1999). Clinical trials of propentofylline in vascu-
pathology of Alzheimer disease and vascular demen- lar dementia. The European/Canadian Propentofylline
tia. Alzheimer Disease and Associated Disorders, 13 Study Group. Alzheimer Disease and Associated
(Suppl. 3): S115–S123. Disorders, 13 (Suppl. 3): S166–S171.
Kalaria, R.N., Ballard, C.G., Ince, P.G. et al. (2001). Multiple Kittner, B., Rossner, M. and Rother, M. (1997). Clinical trials
substrates of late-onset dementia: Implications for in dementia with propentofylline. Annals of the New
brain protection. Novartis Foundation Symposium, York Academy of Sciences, 826: 307–316.
235: 49–60. Kleijnen, J. and Knipschild P. (1992). Ginkgo biloba.
Kalayoglu, M.V., Libby, P. and Byrne, G.I. (2002). Chlamydia Lancet, 340: 1136–1139.
pneumoniae as an emerging risk factor in cardiovas- Knopman, D. and Henderson, V.W. (2003). DHEA for
cular disease. JAMA: Journal of the American Medical Alzheimer’s disease: A modest showing by a superhor-
Association, 288: 2724–2731. mone. Neurology, 60: 1060–1061.
Kalimo, H., Ruchoux, M.-M., Viitanen, M. and Kalaria, Knopman, D.S., Rocca, W.A., Cha, R.H. et al. (2003).
R.N. (2002). CADASIL: A common form of hereditary Survival study of vascular dementia in Rochester,
arteriopathy causing brain infarcts and dementia. Brain Minnesota. Archives of Neurology, 60: 85–90.
Pathology, 12: 371–384. Krupinski, J., Abudawood, M., Matou-Nasri, M. et al.
Kalmijn, S., van Boxtel, M.P., Verschuren, M.W. et al. (2012). Citicoline induces angiogenesis improving
(2002). Cigarette smoking and alcohol consumption survival of vascular/human brain microvessel endothe-
in relation to cognitive performance in middle age. lial cells through pathways involving ERK1/2 and insulin
American Journal of Epidemiology, 156: 936–944. receptor substrate-1. Vascular Cell, 4: 20.
Therapeutic strategies for vascular cognitive disorders 691

Kumar, V., Anand, R., Messina, J. et al. (2000). An effi- Leys, D. and Pasquier, F. (2000). How can cerebral infarcts
cacy and safety analysis of Exelon in Alzheimer’s and hemorrhages lead to dementia? Journal of Neural
disease patients with concurrent vascular risk factors. Transmission, 59: 31–36.
European Journal of Neurology, 7: 159–169. Li, L., Sengupta, A., Haque, N. et al. (2004). Memantine
Kumari, M., Cover, P.O., Poyser, R.H. and Buckingham, inhibits and reverses the Alzheimer type abnormal
J.C. (1997). Stimulation of the hypothalamo- hyperphosphorylation of tau and associated neurode-
pituitary-adrenal axis in the rat by three selective generation. FEBS Letters, 566: 261–269.
type-4 phosphodiesterase inhibitors: In vitro and in Linden, T., Skoog, I., Fagerberg, B. et al. (2004). Cognitive
vivo studies. British Journal of Pharmacology, 121: impairment and dementia 20 months after stroke.
459–468. Neuroepidemiology, 23: 45–52.
Kurz, A. and Van Baelen, B. (2004). Ginkgo biloba com- Lithell, H., Hansson, L., Skoog, I., et al.; for the SCOPE
pared with cholinesterase inhibitors in the treatment Study Group (2003). The Study on Cognition and
of dementia: A review based on meta-analyses by Prognosis in the Elderly (SCOPE): Principal results of a
the Cochrane Collaboration. Dementia and Geriatric randomized double-blind intervention trial. Journal of
Cognitive Disorders, 18: 217–226. Hypertension, 21: 875–886.
Lacombe, P., Sercombe, R., Vaucher, E. and Seylaz, J. Liu, C.K., Miller, B.L., Cummings, J.L. et al. (1991). A quan-
(1997). Reduced cortical vasodilatory response to titative MRI study of vascular dementia. Neurology, 42:
stimulation of the nucleus basalis of Meynert in the 138–143.
aged rat and evidence for a control of the cerebral cir- Longstreth, W.T. Jr., Bernick, C., Manolio, T. et al. (1998).
culation. Annals of the New York Academy of Sciences, Lacunar infarcts defined by magnetic resonance imag-
826: 410–415. ing of 3660 elderly people. Archives of Neurology, 55:
Lammie, A. (2002). Hypertensive cerebral small vessel 1217–1225.
disease and stroke. Brain Pathology, 12: 358–370. Longstreth, W.T. Jr., Manolio, T.A., Arnold, A. et al.; for
Launer, L.J., Masaki, K., Petrovitch, H. et al. (1995). The the Cardiovascular Health Study (1996). Clinical cor-
association between midlife blood pressure levels relates of white matter findings on cranial magnetic
and late-life cognitive function. The Honolulu-Asia resonance imaging of 3301 elderly people. Stroke, 27:
Aging Study. JAMA: Journal of the American Medical 1274–1282.
Association, 274: 1846–1851. López-Arrieta, J.M. and Birks, J. (2002). Nimodipine for
Launer, L.J., Ross, G.W., Petrovik, H. et al. (2000). Midlife primary degenerative, mixed and vascular demen-
blood pressure and dementia: The Honolulu-Asia tia. Cochrane Database of Systematic Reviews, (3):
aging study. Neurobiology of Aging, 21: 49–55. CD000147.
Laurin, D., Masaki, K.H., Foley, D.J. et al. (2004). Midlife López-Pousa, S., Mercadal-Dalmau, J., Marti-Cuadros,
dietary intake of antioxidants and risk of late-life A.M. et al. (1997). Triflusal in the prevention of vascu-
incident dementia: The Honolulu-Asia Aging Study. lar dementia. Revista de Neurología (Barcelona), 25:
American Journal of Epidemiology, 159: 959–967. 1525–1528.
Le Bars, P.L., Katz, M.M., Berman, N., et al.; for the Lourida, I., Soni, M., Thompson-Coon, J. et al. (2013).
North American EGb Study Group. (1997). A pla- Mediterranean diet, cognitive function, and dementia:
cebo-controlled, double-blind, randomized trial of A systematic review. Epidemiology, 24: 479–489.
an extract of Ginkgo biloba for dementia. JAMA: Lunetta, M. and Salanitri, T. (1992). Lowering of plasma vis-
Journal of the American Medical Association, 278: cosity by the oral administration of the glycosaminoglycan
1327–1332. sulodexide in patients with peripheral vascular disease.
Leblanc, G.G., Meschia, J.F., Stuss, D.T. and Hachinski, V. Journal of International Medical Research, 20: 45–53.
(2006). Genetics of vascular cognitive impairment: The Maina, G., Fiori, L., Torta, R. et al. (1989). Oxiracetam in
opportunity and the challenges. Stroke, 37: 248–255. the treatment of primary degenerative and multi-
Lechner, H. (1998). Status of treatment of vascular demen- infarct dementia: A double-blind, placebo-controlled
tia. Neuroepidemiology, 17: 10–13. study. Neuropsychobiology, 21: 141–145.
Lee, H.J., Kang, J.S. and Kim, Y.I. (2009). Citicoline protects Mak, I.T., Boehme, P. and Weglicki, W.B. (1995).
against cognitive impairment in a rat model of chronic Antioxidant effects of calcium channel blockers
cerebral hypoperfusion. Journal of Clinical Neurology, against free radical-impaired endothelial cell prolifera-
5: 33–38. tion. Biochemical Pharmacology, 50: 1531–1534.
Lefebvre, C., Deplanque, D., Hénon, H. et al.; for the SAFE II Malouf, R. and Birks, J. (2004). Donepezil for vascular cog-
Investigators. (2006). Influence of dementia on antithrom- nitive impairment. Cochrane Database of Systematic
botic therapy prescribed before stroke in patients with Reviews, (1): CD004395.
atrial fibrillation. Cerebrovascular Disease, 21: 401–407. Malouf, M., Grimley, E.J. and Areosa, S.A. (2003). Folic
Leys, D., Hénon, H., Mackowiak-Cordoliani, M.A. and acid with or without vitamin B12 for cognition and
Pasquier. F. (2005). Poststroke dementia. Lancet dementia. Cochrane Database of Systematic Reviews,
Neurology, 4: 752–759. (4): CD004514.
692 Dementia

Mani, R.B. (2004). The evaluation of disease modifying Mendez, M.F. and Cummings, J.L. (2003). Dementia. A
therapies in Alzheimer’s disease: A regulatory view- Clinical Approach. Philadelphia, PA: Butterworth-
point. Statistics in Medicine, 23: 305–314. Heinemann (Elsevier Science), pp. 142–146.
Mari, D., Parnetti, L., Coppola, R. et al. (1996). Hemostasis Meschia, J.F., Bushnell, C., Boden-Albala, B. et al. (2014).
abnormalities in patients with vascular dementia and Guidelines for the primary prevention of stroke:
Alzheimer’s disease. Thrombosis and Haemostasis, 75: A statement for healthcare professionals from
216–218. the American Heart Association/American Stroke
Marie, P. (1901). Des foyers lacunaires de désintégration et Association. Stroke, 45: 3754–3832.
de différents autres états cavitaires du cerveau. Revue Messerli, F.H., Williams, B. and Ritz. E. (2007). Essential
de Médecine (Paris), 21: 281–298. hypertension. Lancet, 370: 591–603.
Mark, I.T., Boehme, P., Weqlicki, W.B. (1992). Antioxidant Mesulam, M., Siddique, T. and Cohen, B. (2003).
effects of calcium channel blockers against free radical Cholinergic denervation in a pure multi-infarct state:
injury in endothelial cells. Correlation of protection Observations on CADASIL. Neurology, 60: 1183–1185.
with preservation of glutathione levels. Circulation Mielke, R., Möller, H.-J., Erkinjunti, T. et al. (1998).
Research, 70: 1099–1103. Propentofylline in the treatment of vascular demen-
Marta, M., Zanchetti, A., Wong, N.D. et al. (2014). Patients tia and Alzheimer-type dementia: Overview of phase
with prehypertension – Do we have enough evidence to I and phase II clinical trials. Alzheimer Disease and
treat them? Current Vascular Pharmacology, 12: 586–597. Associated Disorders, 12 (Suppl. 2): 29–35.
Martí-Massó, J.F. and Lozano, R. (1990). Nicardipine in the Mikol, J. (1968). Binswanger’s disease and related forms.
prevention of cerebral infarction. Clinical Therapeutics, Contribution to the study of arteriosclerotic leuko-
12: 344–351. encephalopathies. Revue Neurologique (Paris), 118:
Matthews, F.E., Brayne, C., Lowe, J. et al. (2009). 111–132.
Epidemiological pathology of dementia: Attributable- Mikol, J. and Vallat-Decouvelaere, A.V. (2000). Vascular
risks at death in the Medical Research Council dementia. Annales de Pathologie, 20: 470–478.
Cognitive Function and Ageing Study. PLoS Medicine, Miller, L.J. and Chacko, R. (2004). The role of choles-
6 (11): e1000180. terol and statins in Alzheimer’s disease. Annals of
McCurry, S.M., Edland, S.D., Teri, L. et al. (1999). The cog- Pharmacotherapy, 38: 91–98.
nitive abilities screening instrument (CASI): Data from a Mills, E.J. and Chow, T.W. (2003). Randomized controlled
cohort of 2524 cognitively intact elderly. International trials in long-term care residents with dementia: A
Journal of Geriatric Psychiatry, 14: 882–888. systematic review. Journal of the American Medical
McGuinness, B., Craig, D., Bullock, R. et al. (2014). Statins Directors Association, 4: 302–307.
for the treatment of dementia. Cochrane Database of Mirra, S.S. (1997). Neuropathological assessment of
Systematic Review, (7): CD007514. Alzheimer’s disease: The experience of the consor-
McGuinness, B., Todd, S., Passmore, P. and Bullock, R. tium to establish a registry for Alzheimer’s
(2009). Blood pressure lowering in patients without disease. International Psychogeriatrics, 9 (Suppl. 1):
prior cerebrovascular disease for prevention of cogni- 263–268.
tive impairment and dementia. Cochrane Database of Möbius, H. J. (1999). Pharmacologic rationale for meman-
Systematic Review, (4): CD004034. tine in chronic cerebral hypoperfusion, especially
McGuire, H.L., Svetkey, L.P., Harsha, D.W. et al. (2004). vascular dementia. Alzheimer Disease and Associated
Comprehensive lifestyle modification and blood Disorders, 13 (Suppl. 3): S172–S178.
pressure control: A review of the PREMIER trial. Möbius, H. J. (2003). Memantine: Update on the current
Journal of Clinical Hypertension (Greenwich), evidence. International Journal of Geriatric Psychiatry,
6: 383–390. 18 (Suppl. 1): S47–S54.
McKhann, G., Drachman, D., Folstein, M. et al. (1984). Möbius, H. J. and Stoffler, A. (2002). New approaches to
Clinical diagnosis of AD: Report of the NINCDS- clinical trials in vascular dementia: Memantine in small
ADRDA Work Group under the auspices of vessel disease. Cerebrovascular Diseases, 13 (Suppl. 2):
Department of Health and Human Services Task Force 61–66.
on AD. Neurology, 34: 939–944. Möbius, H. J. and Stoffler, A. (2003). Memantine in
McNeely, W. and Goa, K.L. (1998). Triflusal. Drugs, 55: vascular dementia. International Psychogeriatrics, 15
823–833. (Suppl. 1): 207–213.
Mead, S., James-Galton, M, Revesz, T. et al. (2000). Mohs, R.C., Doody, R.S., Morris, J.C. et al. (2001). A 1-year
Familial British dementia with amyloid angiopathy: placebo-controlled preservation of function survival
Early clinical, neuropsychological and imaging find- study of donepezil in AD patients. Neurology, 57:
ings. Brain, 123: 975–991. 481–488.
Meisel, C., Johne, A. and Roots, I. (2003). Fatal intracere- Mondadori, C. (1993). The pharmacology of the noot-
bral mass bleeding associated with Ginkgo biloba and ropics: New insights and new questions. Behavioural
ibuprofen. Atherosclerosis, 167: 367. Brain Research, 59: 1–9.
Therapeutic strategies for vascular cognitive disorders 693

Moody, D.M., Brown, W.R., Challa, V.R. and Anderson, R.L. people (FINGER): A randomised controlled trial. The
(1995). Periventricular venous collagenosis: Association Lancet, 385: 2255–2263. http://dx.doi.org/10.1016/
with leukoaraiosis. Radiology, 194: 469–476. S0140-6736(15)60461-5.
Mooradian, A.D. (1997). Central nervous system compli- Norton, S., Matthews, F.E., Barnes, D.E. et al. (2014).
cations of diabetes mellitus – A perspective from the Potential for primary prevention of Alzheimer’s
blood-brain barrier. Brain Research and Brain Research disease: An analysis of population-based data. The
Reviews, 23: 210–218. Lancet Neurology, 13: 788–794.
Moretti, R., Torre, P., Antonello, R.M. and Cazzato, G. (2001). Nyenhuis, D.L. and Gorelick, P.B. (2007). Diagnosis and
Rivastigmine in subcortical vascular dementia: A com- management of vascular cognitive impairment. Current
parison trial on efficacy and tolerability for 12 months Atherosclerosis Reports, 9: 326–332.
follow-up. European Journal of Neurology, 8: 361–362. O’Brien, J.T., Erkinjuntti, T., Reisberg, B. et al. (2003).
Moretti, R., Torre, P., Antonello, R.M. et al. (2002). Vascular cognitive impairment. Lancet Neurology, 2:
Rivastigmine in subcortical vascular dementia: An 89–98.
open 22-month study. Journal of the Neurological Obulesu, M. and Rao, D.M. (2011). Effect of plant extracts
Sciences, 203: 141–146. on Alzheimer’s disease: An insight into therapeutic
Moretti, R., Torre, P., Antonello, R.M. et al. (2004). avenues. Journal of Neurosciences in Rural Practice, 2:
Rivastigmine in vascular dementia. Expert Opinion in 56–61.
Pharmacotherapy, 5: 1399–1410. Ohman, J., Servo, A. and Heiskanen, O. (1991). Long-term
Moretti, R., Torre, P., Antonello, R.M. et al. (2008). effects of nimodipine on cerebral infarcts and out-
Different responses to rivastigmine in subcortical vas- come after aneurysmal subarachnoid hemorrhage and
cular dementia and multi-infarct dementia. American surgery. Journal of Neurosurgery, 74: 8–13.
Journal of Alzheimer’s Disease and Other Dementias, Olin, J., Schneider, l., Novit, A. and Luczak, S. (2001).
23: 167–176. Hydergine for dementia. Cochrane Database of
Moriau, M., Crasborn, L., Lavenne-Pardonge, E. et al. (1993). Systematic Reviews, (2): CD000359.
Platelet antiaggregant and rheological properties of Orgogozo, J.-M., Rigaud, A.S., Stoffler, A. et al. (2002).
piracetam. A pharmacodynamic study in normal subjects. Efficacy and safety of memantine in patients with
Arzneimittel-Forschung/Drug Research, 43: 110–118. mild to moderate vascular dementia: A random-
Moroney, J.T., Bagiella, E., Desmond, D.W. et al. (1996). ized, placebo-controlled trial (MMM 300). Stroke, 33:
Risk factors for incident dementia after stroke. Role of 1834–1839.
hypoxic ischemic disorders. Stroke, 27: 1283–1289. Otsuka, M., Yamaguchi, K. and Ueki, A. (2002). Similarities
Nagai, M., Hoshide, S., Ishikawa, J. et al. (2008). and differences between Alzheimer’s disease and vas-
Ambulatory blood pressure as an independent cular dementia from the viewpoint of nutrition. Annals
determinant of brain atrophy and cognitive function of the New York Academy of Sciences, 977: 155–161.
in elderly hypertension. Journal of Hypertension, 26: Ott, A., Slooter, A.J., Hofman, A. et al. (1998). Smoking
1636–1641. and risk of dementia and Alzheimer’s disease in a
Nagai, M., Hoshide, S., Ishikawa, J. et al. (2012). Visit- population-based cohort study: The Rotterdam Study.
to-visit blood pressure variations: New independent Lancet, 351: 1840–1843.
determinants for cognitive function in the elderly Ott, A., Stolk, R.P., van Harskamp, F. et al. (1999). Diabetes
at high risk of cardiovascular disease. Journal of mellitus and the risk of dementia: The Rotterdam
Hypertension, 30: 1556–1563. Study. Neurology, 53: 1937–1942.
Nagai, M., Hoshide, S. and Kario, K. (2010). Hypertension Pantoni, L., Bianchi, C., Beneke, M. et al. (2000). The
and dementia. American Journal of Hypertension, 23: Scandinavian Multi-Infarct Dementia Trial: A double-
116–124. blind, placebo-controlled trial on nimodipine in multi-
Nasreddine, Z.S., Phillips, N.A., Bedirian, V. et al. (2005). infarct dementia. Journal of the Neurological Sciences,
The Montreal cognitive assessment, MoCA: A brief 175: 116–123.
screening tool for mild cognitive impairment. Journal Pantoni, L., Carosi, M., Amigoni, S. et al. (1996). A pre-
of the American Geriatric Society, 53: 695–699. liminary open trial with nimodipine in patients with
Nathan, D.M., Buse, J.B., Davidson, M.B. et al. (2009). cognitive impairment and leukoaraiosis. Clinical
Medical management of hyperglycemia in type 2 Neuropharmacology, 19: 497–506.
diabetes: A consensus algorithm for the initiation and Pantoni, L., del Ser, T., Soglian, A.G. et al. (2005). Efficacy
adjustment of therapy: A consensus statement of the and safety of nimodipine in subcortical vascular
American Diabetes Association and the European dementia: A randomized placebo-controlled trial.
Association for the Study of Diabetes. Diabetes Care, Stroke, 36: 619–624.
32: 193–203. Pantoni, L., Rossi, R., Inzitari, D. et al. (2000). Efficacy and
Ngandu, T., Lehtisalo, J. and Solomon, A. et al. (2015). safety of nimodipine in subcortical vascular dementia:
A 2 year multidomain intervention of diet, exercise, A subgroup analysis of the Scandinavian multi-infarct
cognitive training, and vascular risk monitoring versus dementia trial. Journal of the Neurological Sciences,
control to prevent cognitive decline in at-risk elderly 175: 124–134.
694 Dementia

Parnetti, L. (1999). Pathophysiology of vascular dementia Petrovitch, H., Ross, G.W., Steinhorn, S.C. et al. (2005). AD
and white matter changes. Revue Neurologique (Paris), lesions and infarcts in demented and non-demented
155: 754–758. Japanese-American men. Annals of Neurology, 57:
Parnetti, L., Ambrosoli, L., Abate, G. et al. (1995). 98–103.
Posatirelin for the treatment of late-onset Alzheimer’s Pfeiffer, E. (1975). A Short Portable Mental Status
disease: A double-blind multicentre study vs citicoline Questionnaire for the assessment of organic brain
and ascorbic acid. Acta Neurologica Scandinavica, 92: deficit in elderly patients. Journal of the American
135–140. Geriatrics Society, 23: 433–441.
Parnetti, L., Ambrosoli, L., Agliati, G. et al. (1996). Pioro, E.P., Brooks, B.R., Cummings, J. et al. (2010).
Posatirelin in the treatment of vascular dementia: Dextromethorphan plus ultra low-dose quinidine
A double-blind multicentre study vs placebo. Acta reduces pseudobulbar affect. Annals of Neurology, 68:
Neurologica Scandinavica, 93: 456–463. 693–702.
Parnetti, L., Mari, D., Abate, G. et al. (1997). Vascular Pischel, T. (1998). Long-term-efficacy and safety of pro-
dementia Italian sulodexide study (VA.D.I.S.S.). Clinical pentofylline in patients with vascular dementia. Results
and biological results. Thrombosis Research, 87: of a 12 months placebo-controlled trial. Neurobiology
225–233. of Aging, 19 (Suppl. 4): S182.
Parnetti, L., Mignini, F., Tomassoni, D. et al. (2007). Pittilo, R.M. (2000). Cigarette smoking, endothelial injury
Cholinergic precursors in the treatment of cognitive and cardiovascular disease. International Journal of
impairment of vascular origin: Ineffective approaches Experimental Pathology, 81: 219–230.
or need for re-evaluation? Journal of the Neurological Plosker, G.L. and Gauthier, S. (2009). Cerebrolysin: A
Sciences, 257: 264–269. review of its use in dementia. Drugs in Aging, 26:
Pasquier, F., Boulogne, A., Leys, D. and Fontaine, P. 893–915.
(2006). Diabetes mellitus and dementia. Diabetes and Pohjasvaara, T., Erkinjuntti, T., Vataja, R. and Kaste, M.
Metabolism, 32: 403–414. (1997). Dementia three months after stroke. Baseline
Pearce, J.D., Li, J., Edwards, M.S. et al. (2004). Differential frequency and effect of different definitions of demen-
effects of Rho-kinase inhibition on artery wall mass tia in the Helsinki Stroke Aging Memory Study (SAM)
and remodeling. Journal of Vascular Surgery, 39: cohort. Stroke, 28: 785–792.
223–228. Pohjasvaara, T., Leppavouri, A., Siira, I. et al. (1998).
Pearce, L.A., McClure, L.A., Anderson, D.C. et al.; for the Frequency and clinical determinants of post-stroke
SPS3 Investigators (2014). Effects of long-term blood depression. Stroke, 29: 2311–2317.
pressure lowering and dual antiplatelet treatment Pohjasvaara, T., Leskela, M., Vataja, R. et al. (2002). Post-
on cognitive function in patients with recent lacunar stroke depression, executive dysfunction and func-
stroke: A secondary analysis from the SPS3 ran- tional outcome. European Journal of Neurology, 9:
domised trial. Lancet Neurology, 13: 1177–1185. 269–275.
Pendlebury, S.T. and Rothwell, P.M. (2009). Prevalence, Pohjasvaara, T., Mantyla, R., Ylikoski, R. et al. (2000).
incidence, and factors associated with pre-stroke and Comparison of different clinical criteria (DSMIII,
post-stroke dementia: A systematic review and meta- ADDTC, ICD-10, NINDS-AIREN, DSM-IV) for the diag-
analysis. Lancet Neurology, 8: 1006–1018. nosis of vascular dementia. Stroke, 31: 2952–2957.
Pepine, C.J. and Lambert, C.R. (1990). Cardiovascular Polidori, M.C., Pientka, L. and Mecocci, P. (2012). A review
effects of nicardipine. Angiology, 41: 978–986. of the major vascular risk factors related to Alzheimer’s
Perry, E.K., Gibson, P.H., Blessed, G. et al. (1977). disease. Journal of Alzheimer’s Disease, 32: 521–530.
Neurotransmitter enzyme abnormalities in senile Polinsky, R.J. (1998). Clinical pharmacology of rivastig-
dementia. Choline acetyltransferase and glutamic mine: A new generation acetylcholinesterase inhibi-
acid decarboxylase activities in necropsy brain tor for the treatment of Alzheimer’s disease. Clinical
tissue. Journal of the Neurological Sciences, 34: Therapeutics, 20: 634–647.
247–265. Pratt, R.D. (2002). Patient populations in clinical trials of
Peters, R., Beckett, N., Forette, F. et al.; for the HYVET the efficacy and tolerability of donepezil in patients
Investigators (2008). Incident dementia and blood with vascular dementia. Journal of the Neurological
pressure lowering in the Hypertension in the Sciences, 203–204: 57–65.
Very Elderly Trial cognitive function assessment Prince, M.J. (1997). The treatment of hypertension in
(HYVETCOG): A double-blind, placebo controlled trial. older people and its effect on cognitive function.
Lancet Neurology, 7: 683–689. Biomedicine and Pharmacotherapy, 51: 208–212.
Petersen, R.C. (2004). Mild cognitive impairment as a Prince, M.J., Bird, A.S., Blizard, R.A. and Mann, A. H.
diagnostic entity. Journal of Internal Medicine, 256: (1996). Is the cognitive function of older patients
183–194. affected by antihypertensive treatment? Results from
Petersen, R.C., Smith, G.E., Waring, S.C. et al. (1999). Mild 54 months of the Medical Research Council’s trial of
cognitive impairment: Clinical characterization and hypertension in older adults. British Medical Journal,
outcome. Archives of Neurology, 56: 303–308. 312: 801–805.
Therapeutic strategies for vascular cognitive disorders 695

PROGRESS Collaborative Group (2001). Randomised trial Román, G.C. (1999c). Hyperbaric oxygenation: A promis-
of a perindopril-based blood-pressure-lowering regi- ing treatment for Binswanger’s disease. Revista de
men among 6,105 individuals with previous stroke or Neurología (Barcelona), 28: 707.
transient ischaemic attack. Lancet, 358: 1033–1041. Román, G.C. (2000). Perspectives in the treatment of vas-
Quinn, J., Moore, M., Benson, D.F. et al. (2000). A vid- cular dementia. Drugs of Today, 36: 641–653.
eotaped CIBIC for dementia patients: Validity and Román, G.C. (2002a). Vascular dementia revisited:
reliability in a simulated clinical trial. Neurology, 58: Diagnosis, pathogenesis, treatment, and prevention.
433–437. Medical Clinics of North America, 86: 477–499.
Rainer, M., Brunnbauer, M., Dunky, A. et al. (1997). Román, G.C. (2002b). Defining dementia: Clinical cri-
Therapeutic results with Cerebrolysin in the treatment teria for the diagnosis of vascular dementia. Acta
of dementia. Wiener Medizinische Wochenschrift, 147: Neurologica Scandinavica, 178: 6–9.
426–431. Román, G.C. (2002c). On the history of lacunes, état
Reisberg, B., Doody, R., Stoffler, A. et al.; for the criblé, and the white matter lesions of vascular demen-
Memantine Study Group. (2003). Memantine in tia. Cerebrovascular Diseases, 13 (Suppl. 2): 1–6.
moderate-to-severe Alzheimer’s disease. New England Román, G.C. (2002d). Historical evolution of the concept
Journal of Medicine, 348: 1333–1341. of dementia: A systematic review from 2000 BC to
Richard, E., Gouw, A.A., Scheltens, P. and van Gool, W.A. AD 2000. In N. Qizilbash et al. (eds.), Evidence-based
(2010). Vascular care in patients with Alzheimer disease Dementia Practice. Oxford, United Kingdom: Blackwell
with cerebrovascular lesions slows progression of Science, pp. 199–227.
white matter lesions on MRI. The evaluation of vascular Román, G.C. (2002e). Vascular dementia may be the most
care in Alzheimer’s disease (EVA) study. Stroke, 41 (3): common form of dementia in the elderly. Journal of
554–556. the Neurological Sciences, 203–204: 7–10.
Ringelstein, E.B., Mauckner, A., Schneider, R. et al. (1988). Román, G.C. (2003a). Vascular dementia: Distinguishing
Effects of enzymatic blood defibrination in subcortical characteristics, treatment, and prevention. Journal
arteriosclerotic encephalopathy. Journal of Neurology, of the Americana Geriatrics Society, 51 (Suppl. 5):
Neurosurgery and Psychiatry, 51: 1051–1057. S296–S304.
Rockenstein, E., Adame, A., Mante, M. et al. (2003). The Román, G.C. (2003b). Stroke, cognitive decline and vascu-
neuroprotective effects of Cerebrolysin in a transgenic lar dementia: The silent epidemic of the 21st century.
model of Alzheimer’s disease are associated with Neuroepidemiology, 22: 161–164.
improved behavioral performance. Journal of Neural Román, G.C. (2004). Brain hypoperfusion: A critical fac-
Transmission, 110: 1313–1327. tor in vascular dementia. Neurological Research, 26:
Rockwood, K., Kirkland, S. and Hogan, D.B. (2002). Use 454–458.
of lipid-lowering agents, indication bias, and the risk Román, G.C. (2008a). Alzheimer disease research:
of dementia in community-dwelling elderly people. Have we forgotten the cerebrovascular circulation?
Archives of Neurology, 59: 223–227. Alzheimer Disease and Associated Disorders, 22: 1–3.
Rockwood, K., Wentzel, C., Hachinski, V. et al. (2000). Román, G.C. (2008b). The epidemiology of vascular
Prevalence and outcomes of vascular cognitive impair- dementia. In Duyckaerts, C. and Litvan, I. (eds.),
ment. Vascular cognitive impairment investigators of Handbook of Clinical Neurology (Vol. 89, 3rd Series,
the Canadian study on health and aging. Neurology, Chapter 59, pp. 639–658). Aminoff, M.J., Boller, F. and
54: 447–451. Swaab, D.F. (Series Editors), Dementias. Amsterdam,
Román, G.C. (1985a). The identity of lacunar dementia and The Netherlands: Elsevier.
Binswanger disease. Medical Hypotheses, 16: 389–391. Román, G.C. (2014). Vascular neurocognitive disorders. In
Román, G.C. (1985b). Lacunar dementia. In Hutton, Gabbard’s Treatments of Psychiatric Disorders (DSM-5
J.T. and Kenny, A.D. (eds.), Senile Dementia of the ed., Chapter 66, pp. 977–985).
Alzheimer Type. New York, NY: Alan R. Liss, Inc., Román, G.C. (2015). MTHFR gene mutations: A potential
pp. 131–151. marker of late-onset Alzheimer’s disease? Journal of
Román, G.C. (1987). Senile dementia of the Binswanger Alzheimer’s Disease, 47 (2): 323–327.
type: A vascular form of dementia in the elderly. Román, G.C. and Boller, F. (2014). Vascular factors in neu-
Journal of the American Medical Association, 258: rodegenerative diseases: A path towards treatment
1782–1788. and prevention. Functional Neurology, 29: 85–86.
Román, G.C. (1991). White matter lesions and normal-pres- Román, G.C., Erkinjuntti, T., Wallin, A. et al. (2002).
sure hydrocephalus: Binswanger disease or Hakim syn- Subcortical ischaemic vascular dementia. Lancet
drome? American Journal of Neuroradiology, 12: 40–41. Neurology, 1: 426–436.
Román, G.C. (1999a). Vascular dementia today. Revue Román, G.C. and Kalaria, R.N. (2006). Vascular deter-
Neurologique (Paris), 155 (Suppl. 4): S64–S72. minants of cholinergic deficits in Alzheimer disease
Román, G.C. (1999b). Editorial: New insight into Binswanger and vascular dementia. Neurobiology of Aging, 27:
disease. Archives of Neurology, 56: 1061–1062. 1769–1785.
696 Dementia

Román, G.C., Nash, D.T. and Fillit, H. (2012). Translating Ruchoux, M.M., Guerouaou, D., Vandenhaute, B. et al.
current knowledge into dementia prevention. (1995). Systemic vascular smooth muscle cell impair-
Alzheimer Disease and Associated Disorders, 26: ment in cerebral autosomal dominant arteriopathy
295–299. with subcortical infarcts and leukoencephalopathy.
Román, G.C. and Pascual, B. (2012). Contribution of neu- Acta Neuropathologica, 89: 500–512.
roimaging to the diagnosis of Alzheimer’s disease and Sabbatini, M., Coppi, G. and Maggioni, A. et al. (1998).
vascular dementia. Archives of Medical Research, 43: Effect of lesions of the nucleus basalis magnocellu-
671–676. laris and of treatment with posatirelin on cholinergic
Román, G.C. and Rogers, S.J. (2004). Donepezil: A clinical neurotransmission enzymes in the rat cerebral cor-
review of current and emerging indications. Expert tex. Mechanisms of Ageing and Development, 104:
Opinion in Pharmacotherapy, 5: 161–180. 183–194.
Román, G.C. and Royall, D.R. (1999). Executive control Sachdev, P. (1999). Vascular cognitive disorder. International
function: A rational basis for the diagnosis of vas- Journal of Geriatric Psychiatry, 14: 402–403.
cular dementia. Alzheimer Disease and Associated Sachdev, P., Kalaria, R. and O’Brien, J. et al. (2014).
Disorders, 13 (Suppl. 3): S69–S80. Diagnostic criteria for vascular cognitive disorders:
Román, G.C. and Royall, D.R. (2004). A diagnostic dilemma: A VASCOG statement. Alzheimer Disease and
Is Alzheimer’s dementia Alzheimer’s disease, vascular Associated Disorders, 28: 206–218.
dementia, or both? The Lancet Neurology, 3: 141. Sacks, F.M., Svetkey, L.P., Vollmer, W.M. et al.; for the DASH-
Román, G.C., Sachdev, P., Royall, D.R. et al. (2004). Vascular Sodium Collaborative Research Group (2001). Effects
cognitive disorder: A new diagnostic category updating on blood pressure of reduced dietary sodium and the
vascular cognitive impairment and vascular dementia. Dietary Approaches to Stop Hypertension (DASH) diet.
Journal of the Neurological Sciences, 226: 81–87. New England Journal of Medicine, 344: 3–10.
Román, G.C., Salloway, S., Black, S.E. et al. (2010). Sahlas, D.J., Bilbao, J.M., Swartz, R.H. and Black, S.E.
Randomized, placebo-controlled, clinical trial of (2002). Clasmatodendrosis correlating with periven-
donepezil in vascular dementia: Differential effects by tricular white matter signal hyperintensity in a case of
hippocampal size. Stroke, 41: 1213–1221. mixed dementia. Annals of Neurology, 52: 378–81.
Román, G.C., Tatemichi, T.K., Erkinjuntti, T. et al. (1993). Sakakura, K., Ishikawa, J., Okuno, M. et al. (2007).
Vascular dementia: Diagnostic criteria for research Exaggerated ambulatory blood pressure variability
studies. Report of the NINDS-AIREN International is associated with cognitive dysfunction in the very
Workshop. Neurology, 43: 250–260. elderly and quality of life in the younger elderly.
Román, G.C., Wilkinson, D.G., Doody, R.S. et al. (2005). American Journal of Hypertension, 20: 720–727.
Donepezil in vascular dementia: Combined analysis of Sakurada, T., Alufuzoff, I., Winblad, B. and Nordberg, A.
two large-scale clinical trials. Dementia and Geriatric (1990). Substance P-like immunoreactivity, choline
Cognitive Disorders, 20: 338–344. acetyltransferase activity and cholinergic muscarinic
Rosen, W.G., Mohs, R.C. and Davis, K.L. (1984). A new rat- receptors in Alzheimer’s disease and multi-infarct
ing scale for Alzheimer’s disease. American Journal of dementia. Brain Research, 521: 329–332.
Psychiatry, 14: 1356–1364. Salloway, S., Pratt, R.D., Perdomo, C.A.; for the Donepezil
Rosen, W.G., Terry, R.D., Fuld, P.A. et al. (1980). 307 and 308 Study Groups (2003). A comparison of
Pathological verification of ischemic score in differen- the cognitive benefits of donepezil in patients with
tiation of dementias. Annals of Neurology, 7: 486–488. cortical versus subcortical vascular dementia: A sub-
Roth, M., Tym, E., Mountjoy, C.Q. et al. (1986). CAMDEX. analysis of two 24-week, randomized, double-blind,
A standardised instrument for the diagnosis of placebo-controlled trials. Neurology, 60 (Suppl. 1):
mental disorder in the elderly with special reference A141–A142.
to the early detection of dementia. British Journal of Samlaska, C.P. and Winfield, E.A. (1994). Clinical review:
Psychiatry, 149: 698–709. Pentoxifylline. Journal of the American Academy of
Rother, M., Erkinjuntti, T., Roessner, M. et al. (1998). Dermatology, 30: 603–621.
Propentofylline in the treatment of Alzheimer’s disease Sarazin, M., Amarenco, P., Mikol, J. et al. (2002). Reversible
and vascular dementia: A review of phase III trials. leukoencephalopathy in cerebral amyloid angiopathy
Dementia and Geriatric Cognitive Disorders, 9 (Suppl. presenting as subacute dementia. European Journal of
1): 36–43. Neurology, 9: 353–358.
Royall, D.R., Cordes, J.A. and Polk, M. (1998). CLOX: An Sato, A., Sato, Y. and Uchida, S. (2001). Regulation of
executive clock drawing task. Journal of Neurology, regional cerebral blood flow by cholinergic fibers origi-
Neurosurgery and Psychiatry, 64: 588–594. nating in the basal forebrain. International Journal of
Royall, D.R., Mahurin, R.K. and Gray, K.F. (1992). Bedside Developmental Neuroscience, 19: 327–337.
assessment of executive cognitive impairment: The Saver, J.L. (2008). Citicoline: Update on a promising and
executive interview. Journal of the American Geriatric widely available agent for neuroprotection and neuro-
Society, 40: 1221–1226. repair. Reviews in Neurological Diseases, 5: 167–177.
Therapeutic strategies for vascular cognitive disorders 697

Savva, G.M. and Stephan, B.C.M.; for the Alzheimer’s Society Semplicini, A., Maresca, A., Simonella, C., et al. (2000).
Vascular Dementia Systematic Review Group (2010). Cerebral perfusion in hypertensives with carotid artery
Epidemiological studies of the effect of stroke on incident stenosis: A comparative study of lacidipine and hydro-
dementia: A systematic review. Stroke, 41: e41–e46. chlorothiazide. Blood Pressure, 9: 34–39.
Scarmeas, N., Luchsinger, J.A., Schupf, N. et al. (2009b). Seshadri, S., Beiser, A., Selhub, J. et al. (2002). Plasma
Physical activity, diet, and risk of Alzheimer disease. homocysteine as a risk factor for dementia and
JAMA: Journal of the American Medical Association, Alzheimer’s disease. New England Journal of
302: 627–637. Medicine, 346: 476–483.
Scarmeas, N., Stern, Y., Mayeux, R. et al. (2009a). Sha, M.C. and Callahan, C.M. (2003). The efficacy of
Mediterranean diet and mild cognitive impairment. pentoxifylline in the treatment of vascular dementia: A
Archives of Neurology, 66: 216–25. systematic review. Alzheimer Disease and Associates
Schneider, J.A., Aggarwal, N.T., Barnes, L. et al. (2009). Disorders, 17: 46–54.
The neuropathology of older persons with and with- Shader, R.I., Harmatz, J.S. and Salzman, C. (1974). A new
out dementia from community versus clinic cohorts. scale for clinical assessment in geriatric populations:
Journal of Alzheimer’s Disease, 18: 691–701. Sandoz Clinical Assessment – Geriatric (SCAG). Journal
Schneider, J.A., Arvanitakis, Z., Bang, W. and Bennett, of the American Geriatric Society, 22: 107–113.
D.A. (2007). Mixed brain pathologies account for most Shah, K., Qureshi, S.U., Johnson, M. et al. (2009). Does
dementia cases in community-dwelling older persons. use of antihypertensive drugs affect the incidence
Neurology, 69: 2197–2204. or progression of dementia? A systematic review.
Schneider, L.S. and Olin, J.T. (1994). Overview of clinical American Journal of Geriatric Pharmacotherapy, 7:
trials of Hydergine in dementia. Archives of Neurology, 250–261.
51: 787–798. Shen, J., Wilmot, K.A., Ghasemzadeh, N. et al. (2015).
Schneider, L.S., Olin, J.T., Doody, R.S. et al. (1997). Validity Mediterranean dietary patterns and cardiovascular
and reliability of the Alzheimer’s Disease Cooperative health. Annual Review of Nutrition, 35: 11.1–11.25.
Study – Clinical Global Impression of Change. Alzheimer Sherman, D.G. (2002). Ancrod. Current Medical Research
Disease and Associated Disorders, 11: S22–S32. and Opinion, 18 (Suppl. 2): S48–S52.
Schneider, R., Ringelstein, E.B., Kiesewetter, H. and Jung, Shustov, S.B. (1997). Controlled clinical trial on the effi-
F. (1987). The role of plasma hyperviscosity in subcor- cacy and safety of oral sulodexide in patients with
tical arteriosclerotic encephalopathy (Binswanger’s peripheral occlusive arterial disease. Current Medical
disease). Journal of Neurology, 234: 67–73. Research and Opinion, 13: 573–582.
Schunkert, H. (2006). Pharmacotherapy for prehyperten- Silveri, M.M., Dikan, J., Ross, A.J. et al. (2008). Citicoline
sion—Mission accomplished? New England Journal of enhances frontal lobe bioenergetics as measured by
Medicine, 354: 1742–1744. phosphorus magnetic resonance spectroscopy. NMR
Secades, J.J., Álvarez-Sabín, J., Castillo, J. et al. (2016). in Biomedicine, 21: 1066–1075.
Citicoline for acute ischemic stroke: A system- Skoog, I., Lernfelt, B. and Landahl, S. (1996). 15-year longi-
atic review and formal meta-analysis of random- tudinal study of blood pressure and dementia. Lancet,
ized, double-blind, and placebo-controlled trials. 347: 1141–1145.
Journal of Stroke and Cerebrovascular Disease, pii: Smith, A.D. (2008). The worldwide challenge of the
S1052-3057(16)30019-2. dementias: A role for B vitamins and homocysteine?
Secades, J.J. (2002). CDP-choline: Update and review Food and Nutrition Bulletin, 29: S143–S172.
of its pharmacology and clinical use. Methods and Smith, A.D., Smith, S.M., de Jager, C.A. et al. (2010).
Findings in Experimental and Clinical Pharmacology, 24 Homocysteine-lowering by B vitamins slows the rate
(Suppl. B): 1–53. of accelerated brain atrophy in mild cognitive impair-
Secades, J.J. (2011). Citicoline: Pharmacological and clini- ment: A randomized controlled trial. PLoS ONE, 5:
cal review, 2010 update. Revista de Neurología, 52 e12244.
(Suppl. 2): S1–S62. Snitz, B.E., O’Meara, E.S., Carlson, M.C. et al.; for
Secades, J.J. and Frontera, G. (1995). CDP-choline: the Ginkgo Evaluation of Memory (GEM) Study
Pharmacological and clinical review. Methods and Investigators (2009). Ginkgo biloba for preventing
Findings in Experimental and Clinical Pharmacology, 17 cognitive decline in older adults: A randomized trial.
(Suppl. B): 1–54. JAMA: Journal of the American Medical Association,
Secades, J.J. and Lorenzo, J.L. (2006). Citicoline: 302: 2663–2670.
Pharmacological and clinical review, 2006 update. Snowdon, D., Greiner, L.H., Mortimer, J.A. et al. (1997).
Methods and Findings in Experimental and Clinical Brain infarction and the clinical expression of
Pharmacology, 28 (Suppl. B): 1–56. Alzheimer disease: The Nun Study. JAMA: Journal of
Selden, N.R., Gitelman, D.R., Salamon-Murayama, N. et al. the American Medical Association, 277: 813–817.
(1998). Trajectories of cholinergic pathways within the Solerte, S.B., Ceresini, G., Ferrari, E. and Fioravanti, M.
cerebral hemispheres of the human brain. Brain, 121: (2000). Hemorreological changes and overproduction
2249–2257. of cytokines from immune cells in mild to moderate
698 Dementia

dementia of the Alzheimer’s type: Adverse effects on Tanne, D., Benderly, M., Goldbourt, U. et al.; for the
cerebromicrovascular system. Neurobiology of Aging, Bezafibrate Infarction Prevention Study Group (2001).
21: 271–281. A prospective study of plasma fibrinogen levels and
Sonninen, V. and Savontaus, M.L. (1987). Hereditary the risk of stroke among participants in the bezafi-
multi-infarct dementia. European Neurology, 27: brate infarction prevention study. American Journal of
209–215. Medicine, 111: 457–463.
Sourander, P. and Walinder, J. (1977). Hereditary multi-infarct Tariot, P.N., Farlow, M.R., Grossberg, G.T. et al.; for the
dementia. Morphological and clinical studies of a new Memantine Study Group (2004). Memantine treatment
disease. Acta Neuropathologica (Berlin), 39: 247–254. in patients with moderate to severe Alzheimer disease
Spiegel, R., Brunner, C., Ermini-Funfschilling, D. et al. already receiving donepezil: A randomized con-
(1991). A new behavioral assessment scale for geri- trolled trial. JAMA: Journal of the American Medical
atric out- and in-patients: The NOSGER (Nurses’ Association, 291: 317–324.
Observation Scale for Geriatric Patients). Journal of Tatemichi, T.K., Desmont, D.W., Paik, M. et al. (1993).
the American Geriatrics Society, 39: 339–347. Clinical determinants of dementia related to stroke.
The SPS3 Investigators (2012). Effects of clopidogrel Annals of Neurology, 33: 568–575.
added to aspirin in patients with recent lacunar stroke. Tatemichi, T.K., Desmont, D.W., Prohovnik, I. et al. (1992).
New England Journal of Medicine, 367: 817–825. Confusion and memory loss from capsular genu
The SPS3 Study Group (2013). Blood-pressure targets in infarction: A thalamocortical disconnection syndrome?
patients with recent lacunar stroke: The SPS3 ran- Neurology, 42: 1966–1979.
domised trial. Lancet, 382: 507–515. Tatemichi, T.K., Desmont, D.W., Prohovnik, I. et al. (1995).
Staekenborg, S.S., Koedam, E.L., Henneman, W.J.P. et al. Strategic infarcts in vascular dementia. A clinical and
(2009). Progression of mild cognitive impairment to imaging experience. Arzneimittel-Forschung/Drug
dementia: Contribution of cerebrovascular disease Research, 45: 371–385.
compared with medial temporal lobe atrophy. Stroke, Tatemichi, T.K., Foulkes, M.A., Mohr, J.P. et al. (1990).
40: 1269–1274. Dementia in stroke survivors in the Stroke Data Bank
Starr, J.M., Whalley, L.J. and Deary, I.J. (1996). The effects cohort: Prevalence, incidence, risk factors, and com-
of antihypertensive treatment on cognitive function: puted tomographic findings. Stroke, 21: 858–866.
Results from the HOPE study. Journal of the American Teng, E.L., Hasegawa, K., Homma, A. et al. (1994). The
Geriatrics Society, 44: 411–415. cognitive abilities screening instrument (CASI):
Stefanutti, C., Morozzi, C., Di Giacomo, S.; for the Italian A practical test for cross-cultural epidemiological
Multicenter Study on Low-Density Lipoprotein studies of dementia. International Psychogeriatrics,
Apheresis Working Group (2013). Italian multicenter 6: 45–58.
study on low-density lipoprotein apheresis Working Thavichachart, N., Phanthumchinda, K., Chankrachang, S.
Group 2009 survey. Therapeutic Apheresis and et al. (2006). Efficacy study of galantamine in possible
Dialysis, 17: 169–178. Alzheimer’s disease with or without cerebrovascular
Stewart, R. and Liolitsa, D. (1999). Type 2 diabetes mel- disease and vascular dementia in Thai patients: A
litus, cognitive impairment and dementia. Diabetic slow-titration regimen. International Journal of Clinical
Medicine, 16: 93–112. Practice, 60: 533–540.
Sturmer, T., Glynn, R.J., Field, T.S. et al. (1996). Aspirin use Thomas, S.J. and Grossberg, G.T. (2009). Memantine: A
and cognitive function in the elderly. American Journal review of studies into its safety and efficacy in treat-
of Epidemiology, 143: 683–691. ing Alzheimer’s disease and other dementias. Clinical
Suzuki, Y., Shibuya, M., Satoh, S. et al. (2008). Safety and effi- Interventions on Aging, 4: 367–377.
cacy of fasudil monotherapy and fasudil-ozagrel combina- Tohgi, H., Abe, T., Kimura, M. et al. (1996). Cerebrospinal
tion therapy in patients with subarachnoid hemorrhage: fluid acetylcholine and choline in vascular dementia
Sub-analysis of the post-marketing surveillance study. of Binswanger and multiple small infarct types as
Neurologia Medico-Chirurgica (Tokyo), 48: 241–247. compared with Alzheimer-type dementia. Journal of
Swartz, R.H., Sahlas, D.J., Black, S.E. et al. (2003). Neural Transmission, 103: 1211–1220.
Strategic involvement of cholinergic pathways and Toledo, J.B., Arnold, S.E., Raible, K. et al. (2013).
executive dysfunction: Does location of white matter Contribution of cerebrovascular disease in autopsy con-
signal hyperintensity matter? Journal of Stroke and firmed neurodegenerative disease cases in the National
Cerebrovascular Diseases, 12: 29–36. Alzheimer’s Coordinating Centre. Brain, 136: 2697–2706.
Szatmari, S.Z. and Whitehouse, P.J. (2003). Vinpocetine Tomimoto, H., Akiguchi, I., Ohtani, R. et al. (2001). The
for cognitive impairment and dementia. Cochrane coagulation-fibrinolysis system in patients with
Database of Systematic Reviews, (1): CD003119. leukoaraiosis and Binswanger disease. Archives of
Szilagyi, A.K., Nemeth, A., Martini, E. et al. (1987). Serum Neurology, 58: 1620–1625.
and CSF cholinesterase activity in various kinds Tomimoto, H., Akiguchi, I., Wakita, H. et al. (1999).
of dementia. European Archives of Psychiatry and Coagulation activation in patients with Binswanger
Neurological Sciences, 236: 309–311. disease. Archives of Neurology, 56: 1104–1108.
Therapeutic strategies for vascular cognitive disorders 699

Toole, J.F., Malinow, M.R., Chambless, L.E. et al. (2004). Vermeer, S.E., Den Heijer, T., KJoudstaal, P.J. et al. (2003).
Lowering homocysteine in patients with ischemic stroke Silent brain infarcts and the risk of dementia and cog-
to prevent recurrent stroke, myocardial infarction, and nitive decline. New England Journal of Medicine, 348:
death: The Vitamin Intervention for Stroke Prevention 1215–1222.
(VISP) randomized controlled trial. JAMA: Journal of Vernon, M.W. and Sorkin, E. M. (1991). Piracetam: An over-
the American Medical Association, 291: 565–575. view of its pharmacological properties and a review of
Traber, W.H. and Gibsen, J. (1989). Nimodipine and its therapeutic use in senile cognitive disorders. Drugs
Central Nervous System Function: New Vistas. and Aging, 1: 17–35.
Stuttgart, Germany: Schattauaer-Verlag. Vila, J.F., Balcarce, P.E., Abiusi, G.R. et al. (1999).
Treves, T.A. and Korczyn, A.D. (1999). Denbufylline in [Hyperbaric oxygenation in subcortical frontal syn-
dementia: A double-blind controlled study. Dementia drome due to small artery disorders with leukoara-
and Geriatric Cognitive Disorders, 10: 505–510. iosis]. Revista de Neurología (Barcelona), 28: 655–660.
Tzourio, C., Anderson, C., Chapman, N. et al.; for the Vilayur, E. and Harris, D.C. (2009). Emerging therapies
PROGRESS Collaborative Group (2003). Effects of for chronic kidney disease: What is their role? Nature
blood pressure lowering with perindopril and inda- Reviews, Nephrology, 5: 375–383.
pamide therapy on dementia and cognitive decline Vinters, H.V., Ellis, W.G., Zarow, C. et al. (2000).
in patients with cerebrovascular disease. Archives of Neuropathological substrate of ischemic vas-
Internal Medicine, 163: 1069–1075. cular dementia. Journal of Neuropathology and
Tzourio, C., Dufouil, C., Ducimetiere, P. and Alperovitch, Experimental Neurology, 59: 931–945.
A. (1999). Cognitive decline in individuals with high Wadworth, A.N. and Chrisp, P. (1992). Co-dergocrine
blood pressure: A longitudinal study in the elderly. mesylate. A review of its pharmacodynamic and
EVA Study Group. Epidemiology of Vascular Aging. pharmacokinetic properties and therapeutic use in
Neurology, 53: 1948–1952. age-related cognitive decline. Drugs and Aging, 2:
Uchida, S., Suzuki, A., Kagitani, F. and Hotta, H. (2000). 153–173.
Effects of age on cholinergic vasodilation of cortical Waegemans, T., Wilsher, C.R., Danniau, A. et al. (2002).
cerebral blood vessels in rats. Neuroscience Letters, Clinical efficacy of piracetam in cognitive impairment:
294: 109–112. A meta-analysis. Dementia and Geriatric Cognitive
Uebelhack, R., Vohs, K., Zytowski, M. et al. (2003). Effect of Disorders, 13: 217–224.
piracetam on cognitive performance in patients under- Wald, D.S., Law, M. and Morris, J.K. (2002). Homocysteine
going bypass surgery. Pharmacopsychiatry, 36: 89–93. and cardiovascular disease: Evidence on causality from
Ulus, I.H., Wurtman, R.J., Mauron, C. and Blusztajn, J.K. a meta-analysis. British Medical Journal, 325: 1202.
(1989). Choline increases acetylcholine release and Wallin, A., Blennow, K. and Gottfries, C.G. (1989).
protects against the stimulation-induced decrease in Neurochemical abnormalities in vascular dementia.
phosphatide levels within membranes of rat corpus Dementia, 1: 120–130.
striatum. Brain Research, 484: 217–227. Wallin, A. and Gottfries, C.G. (1990). Biochemical sub-
Umegaki, H. (2010). Pathophysiology of cognitive dysfunc- strates in normal aging and Alzheimer’s disease.
tion in older people with type 2 diabetes: Vascular Pharmacopsychiatry, 23: 37–43.
changes or neurodegeneration? Age and Ageing, 39: Walsh, A.C. (1968). Anticoagulant therapy as potentially
8–10. effective method for the prevention of presenile
Vale, S. (1998). Subarachnoid haemorrhage associated dementia: Two case reports. Journal of the American
with Ginkgo biloba. Lancet, 352: 36. Geriatrics Society, 16: 472–481.
van Bogaert, L. (1955). Encéphalopathie sous-corticale Walsh, A.C., Walsh, B.H. and Melaney, C. (1978). Senile-
progressive (Binswanger) à évolution rapide chez deux presenile dementia: Follow-up data on an effective
sœurs. Médecine Hellénique, 24: 961–972. psychoterapy-anticoagulant regimen. Journal of the
van Dongen, M., van Rossum, E., Kessels, A. et al. (2003). American Geriatrics Society, 26: 467–470.
Ginkgo for elderly people with dementia and age- Walzl, M. (1993). Effect of heparin-induced extracorporeal
associated memory impairment: A randomized clinical low-density lipoprotein precipitation and bezafibrate
trial. Journal of Clinical Epidemiology, 56: 367–376. on hemorheology and clinical symptoms in cerebral
Vataja, R., Pohjasvaara, T., Matyla, R. et al. (2003). MRI multiinfarct disease. Haemostasis, 23: 192–202.
correlates of executive dysfunction in patients with Walzl, M. (2000). A promising approach to the treatment
ischaemic stroke. European Journal of Neurology, 10: of multi-infarct dementia. Neurobiology of Aging, 21:
625–631. 283–287.
Verhey, F.R., Lodder, J., Rosendaal, N. and Jolles, J. (1996). Wang, S.P., Tao, Z., Ding, S.J. et al. (2009). Hyperbaric
Comparison of seven sets of criteria used for the oxygen combined with donepezil in the treatment
diagnosis of vascular dementia. Neuroepidemiology, of vascular dementia. Chinese Journal of Physical
15: 166–172. Medicine and Rehabilitation, 31: 478–480.
700 Dementia

Watanabe, H., Zhao, Q., Matsumoto, K. et al. (2003). Williams, P.S., Rands, G., Orrel, M. and Spector, A. (2000).
Pharmacological evidence for antidementia effect of Aspirin for vascular dementia. Cochrane Database of
Choto-san (Gouteng-san), a traditional Kampo medicine. Systematic Reviews, (4): CD001296.
Pharmacology Biochemistry and Behavior, 75: 635–643. Willis, K.J. and Hakim, A.M. (2013). Stroke prevention and
Weil, C. (1988). Hydergine: Pharmacologic and Clinical cognitive reserve: Emerging approaches to modify-
Facts. Berlin, Germany: Springer-Verlag. ing risk and delaying onset of dementia. Frontiers in
Weinstein, D.B. and Heider, J.G. (1989). Protective action Neurology, 4: 13.
of calcium channel antagonists in atherogenesis and Winblad, B. and Machado, J.C. (2008). Use of rivastigmine
experimental vascular injury. American Journal of transdermal patch in the treatment of Alzheimer’s dis-
Hypertension, 2.3 (Pt 1): 205–212. ease. Expert Opinion on Drug Delivery, 5: 1377–1386.
Wetterling, T., Kanitz, R.D. and Borgis, K.J. (1996). Winblad, B. and Poritis, N. (1999). Memantine in severe
Comparison of different diagnostic criteria for vascular dementia: Results of the 9M-Best Study (Benefit
dementia (ADDTC, DSM-IV, ICD-10, NINDS-AIREN). and efficacy in severely demented patients during
Stroke, 27: 30–36. treatment with memantine). International Journal of
Wharton, S.B., Brayne, C., Savva, G.M. et al.; for the Geriatric Psychiatry, 14: 135–146.
Medical Research Council Cognitive Function and Winblad, B., Fioravanti, M., Dolezal, T. et al. (2008).
Aging Study (2011). Epidemiological neuropathol- Therapeutic use of nicergoline. Clinical Drug
ogy: The MRC Cognitive Function and Aging Study Investigation, 28: 533–552.
experience. Journal of Alzheimers Disease, 25: Winblad, B., Palmer, K., Kivipelto, M. et al. (2004). Mild
359–372. cognitive impairment: Beyond controversies, towards a
White, C.L., Szychowski, J.M., Roldan, A. et al. (2013). consensus. Report of the international working group
Clinical features and racial/ethnic differences among on mild cognitive impairment. Journal of International
the 3020 participants in the Secondary Prevention of Medicine, 256: 240–246.
Small Subcortical Strokes (SPS3) trial. Journal of Stroke Windisch, M., Gschanes, A. and Hutter-Paier, B. (1998).
and Cerebrovascular Disease, 22: 764–774. Neurotrophic activities and therapeutic experience
Whitehouse, P.J., Kittner, B., Roessner, M. et al. (1998). with a brain derived peptide preparation. Journal of
Clinical trial designs for demonstrating disease- Neural Transmission, 53: 289–298.
course-altering effects in dementia. Alzheimer Disease Wolkowitz, O.M., Kramer, J.H., Reus, V.I. et al.; for the
and Associated Disorders, 12: 281–94. DHEA-Alzheimer’s Disease Collaborative Research
Wiederkehr, S., Simard, M., Fortin, C. and van Reekum, R. (2003). DHEA treatment of Alzheimer’s disease: A
(2008a). Comparability of the clinical diagnostic criteria randomized, double-blind, placebo-controlled study.
for vascular dementia: A critical review. Part I. Journal Neurology, 60: 1071–1076.
of Neuropsychiatry and Clinical Neurosciences, 20: Wong, C.L., Bansback, N., Lee, P.E. and Anis, A.H. (2009).
150–161. Cost-effectiveness: Cholinesterase inhibitors and
Wiederkehr, S., Simard, M., Fortin, C. and van Reekum, R. memantine in vascular dementia. Canadian Journal of
(2008b). Validity of the clinical diagnostic criteria for Neurological Sciences, 36: 735–739.
vascular dementia: A critical review. Part II. Journal World Health Organization (WHO) (2008). ICD-10
of Neuropsychiatry and Clinical Neurosciences, 20: International Statistical Classification of Diseases and
162–177. Related Health Problems, 10th Revision. Geneva,
Wilcock, G., Möbius, H.J., Stoffler, A. and the MMM 500 Switzerland: World Health Organization.
Group (2002). A double-blind, placebo-controlled Xiao, Y., Wang, J., Jiang, S. and Luo, H. (2012). Hyperbaric
multicentre study of memantine in mild to moderate oxygen therapy for vascular dementia. Cochrane
vascular dementia (MMM 500). International Clinical Database of Systematic Reviews, (7): CD009425.
Psychopharmacology, 17: 297–305. Yamada, Y., Furui, H., Furumichi, T. et al. (1990). Inhibitory
Wilkinson, D., Doody, R., Helme, R. et al. (2003). Donepezil effects of endothelial cells and calcium channel block-
in vascular dementia. A randomized, placebo-con- ers on platelet aggregation. Japanese Heart Journal,
trolled study. Neurology, 61: 479–486. 31: 201–205.
Willette, R.N., Shiloh, A.O., Sauermelch, C.F. et al. Yamaguchi, S., Matsubara, M. and Kobayashi, S. (2004).
(1997). Identification, characterization, and functional Event-related brain potential changes after Choto-san
role of phosphodiesterase type IV in cerebral vessels: administration in stroke patients with mild cogni-
Effects of selective phosphodiesterase inhibitors. tive impairments. Psychopharmacology (Berlin), 171:
Journal of Cerebral Blood Flow and Metabolism, 17: 241–249.
210–219. Yarchoan, M., Xie, S.X., Kling, M.A. et al. (2012).
Williams, B. (2004). Protection against stroke and demen- Cerebrovascular atherosclerosis correlates with
tia: An update on the latest clinical trial evidence. Alzheimer pathology in neurodegenerative dementias.
Current Hypertension Reports, 6: 307–313. Brain, 135: 3749–3756.
Therapeutic strategies for vascular cognitive disorders 701

Yates, P.A., Desmond, P.M., Phal, P.M. et al. for the AIBL Zekry, D., Duyckaerts, C., Moulias, R. et al. (2002c).
Research Group (2014). Incidence of cerebral micro- Degenerative and vascular lesions of the brain have
bleeds in preclinical Alzheimer disease. Neurology, synergistic effects in dementia of the elderly. Acta
82:1266-1273. Neuropathologica (Berlin), 103: 481–487.
Yesavage, J.A., Tinkleberg, J.R., Hollister, L.E. and Berger, Zekry, D. and Gold, G. (2010). Management of mixed
P.A. (1979). Vasodilators in senile dementia: A review dementia. Drugs and Aging, 27: 715–728.
of the literature. Archives of General Psychiatry, 36: Zekry, D., Hauw, J.-J. and Gold, G. (2002a). Mixed
220–223. dementia: Epidemiology, diagnosis, and treatment.
Zamrini, E., McGwin, G. and Roseman, J.M. (2004b). Journal of the American Geriatrics Society,
Association between statin use and Alzheimer’s dis- 50: 1431–1438.
ease. Neuroepidemiology, 23: 94–98. Zhang, B.L., Wang, Y.Y. and Chen, R.X. (2002). Clinical
Zamrini, E., Parrish, J.A., Parsons, D. and Harrell, L.E. randomized double-blinded study on treatment
(2004a). Medical comorbidity in black and white of vascular dementia by jiannao yizhi granule.
patients with Alzheimer’s disease. Southern Medical Zhongguo Zhong xi yi jie he za zhi Zhungguo
Journal, 97: 2–6. Zhongguo Zhongxiyi jiehe zazhi, 22 (8):
Zanchetti, A., Bond, M.G., Hennig, M. et al.; for the ELSA 577–580.
Investigators. (2004). Absolute and relative changes Zhong, G., Wang, Y., Zhang, Y. et al. (2015). Smoking is
in carotid intima-media thickness and atherosclerotic associated with an increased risk of dementia: A meta-
plaques during long-term antihypertensive treatment: analysis of prospective cohort studies with investiga-
Further results of the European Lacidipine Study on tion of potential effect modifiers. PLoS One, 10 (3):
Atherosclerosis (ELSA). Journal of Hypertension, 22: e0118333
1201–1212. Zimmermann, M., Colciaghi, F., Cattabeni, F. and Di Luca,
Zekry, D., Duyckaerts, C., Belmin, J. et al. (2002b). M. (2002). Ginkgo biloba extract: From molecular
Alzheimer’s disease and brain infarcts in the mechanisms to the treatment of Alzheimer’s dis-
elderly. Agreement with neuropathology. Journal of ease. Cellular and Molecular Biology (Noisy-le-grand,
Neurology, 249: 1529–1534. France), 48: 613–623.
6
Part    

Dementia with Lewy Bodies and


Parkinson’s Disease

61 Dementia with Lewy bodies: A clinical overview 703


Ian G. McKeith
62 Pathology of dementia with Lewy bodies 714
Glenda Halliday, Simon Lewis and Paul Ince
63 The treatment of dementia with Lewy bodies 730
John-Paul Taylor
64 Cognitive impairment and dementia in Parkinson’s disease 740
Dag Aarsland, Kolbjørn Brønnick, Milica G. Kramberger and Joana B. Pereira
61
Dementia with Lewy bodies: A clinical overview

IAN G. MCKEITH

61.1.2 SCALE OF THE PROBLEM


61.1 INTRODUCTION
Several studies in a range of settings have suggested that
61.1.1 DEMENTIA WITH LEWY BODIES: DLB accounts for just 20% below of all cases of dementia
WHAT’S IN A NAME? referred for neuropathological autopsy (McKeith et al.,
2005). Community-based estimates are much more in vari-
Dementia with Lewy bodies (DLB) is now the preferred able ranging from 0% to 30.5% (Zaccai et al., 2005). A com-
term (McKeith et al., 1996) for a variety of clinical diagno- munity study of 85+ year olds found 5.0% met diagnostic
ses including diffuse Lewy body disease (DLBD) (Kosaka criteria for DLB representing 22% of all demented cases
et al., 1984; Dickson et al., 1987; Lennox et al., 1989), (Rahkonen et al., 2003), similar to other clinical estimates
Lewy body dementia (LBD) (Gibb et al., 1987), demen- (Stevens et al., 2002; Aarsland et al., 2008). A systematic
tia associated with cortical Lewy bodies (DCLB) (Byrne review reported that DLB accounted for 4.2% of all diag-
et al., 1991), the Lewy body variant of Alzheimer’s disease nosed dementias in the community, increasing to 7.5% in
(LBVAD) (Hansen et al., 1990; Förstl et al., 1993) and senile secondary care. The incidence of DLB was 3.8% of new
dementia of Lewy body type (SDLT) (Perry et al.,1990). dementia cases. Vann Jones et al. (2014) estimated the inci-
Figure 61.1 provides a simple schematic of the relationship dence of DLB as 3.5 per 100,000 person-years overall (Savica
between the various Lewy body (LB) terminologies rec- et al., 2013). In clinical practice, therefore, it is likely that
ommended currently, and consistency of usage is essential DLB is significantly under-diagnosed compared to autopsy
for a full understanding of the disorder. This chapter con- findings. Survival in DLB is significantly reduced compared
siders the evolution of diagnostic concepts and methods; to AD, but studies have not confirmed a general tendency
the pathology and management of DLB are considered in to a more rapid cognitive decline. Risk factors for DLB are
subsequent chapters. unclear as prevalence studies have been too limited.
DLB is important in clinical practice because of the fol-
lowing reasons:

●● It usually has a clinical presentation and course, which 61.2 DEVELOPMENT OF CLINICAL


differs from Alzheimer’s disease (AD) and other non- DIAGNOSTIC CRITERIA FOR DLB
AD dementias.
●● Functional disability and impairment in quality of life 61.2.1 EARLY CASE REPORTS
(QoL) are greater in DLB than AD and generate signifi-
cantly higher costs of care. When Friedrich Lewy first described the eosinophilic,
●● The management of psychosis and behavioural distur- neuronal inclusions that we now call Lewy bodies, in
bances, which are common in DLB, is complicated by the brain stem of patients with paralysis agitans, he did
sensitivity to neuroleptic medication. not associate the inclusions with cognitive impairment
●● It may be particularly amenable to cholinesterase and psychiatric disorder (Forster and Lewy, 1912). This
inhibitor treatment. was despite half of his sample being clinically demented

703
704 Dementia

Lewy body disease (LBD) any elderly patient who presents with a rapidly progressive
dementia, followed in short order by rigidity and other par-
kinsonian features. Myoclonus may be present.’
Parkinson’s Lewy body Crystal et al. (1990) in a paper entitled ‘Antemortem diag-
disease dementias nosis of diffuse Lewy body disease’ criticized this approach
(PD) (LBDs)
on the grounds that ‘extrapyramidal features occur in many
patients with severe AD and since dementia occurs in many
subjects with PD, the clinical criteria for the diagnosis of
DLBD remain unclear’. The authors proposed alternative
criteria of ‘progressive dementia with gait disorder, psychiat-
PD dementia Dementia with Lewy ric symptoms and a burst pattern on electroencephalogram
(PDD) bodies (DLB) (EEG) at the time of moderate dementia’. No particular char-
acteristics of the pattern of cognitive impairment were noted.
50–60 Increasing age 70–80 Although, these early clinical definitions were important
in drawing attention to the existence of DLB and describing
some of its salient characteristics, neither could be regarded
Figure 61.1 Schematic of the relationship between the as satisfactory for clinical diagnostic purposes, since they
various Lewy body terminologies.
lacked detail and were not operationalized in a way which
would allow acceptable inter-rater reliability (Hansen and
and a quarter having mood disorders, hallucinations and Galasko, 1992).
paranoid delusions (Förstl and Levy, 1991). The first case
reports specifically describing patients with dementia and 61.2.3 THE NOTTINGHAM GROUP
LBs did not appear until 1961 when Okazaki et al. (1961) FOR THE STUDY OF
published two cases, both elderly men presenting cogni- NEURODEGENERATIVE DISORDERS
tive decline and subsequently developing severe demen-
tia. Over the next 20 years, 34 similar cases were reported. The Nottingham group reported the clinical characteristics of
Kosaka et al. (1984) noted a 3:1 male predominance, with 15 new UK cases in considerable detail, the largest individual
memory disturbance as the presenting feature in 67%, series published at that time (Byrne et al., 1989) leading to the
psychotic states (sic) in 17% and dizziness due to ortho- first formal proposal of operational criteria for DCLB (Byrne
static hypotension in 17%. Progressive dementia with et al., 1991). Seven were men in this sample study, the mean
muscular rigidity occurred eventually in 80% although age of onset was 72 years and the mean duration of illness
only 25% of cases were diagnosed with parkinsonism. The was 5.5 years. Forty per cent presented symptoms and signs of
first substantial listing of these Japanese cases in western idiopathic PD with cognitive impairment occurring 1–4 years
literature was done in 1987 by Gibb et al. who added four later. A further 20% had parkinsonism and mild cognitive
new UK cases. impairment (MCI) during presentation and the remaining
40% showed motor features later in their illnesses, gait distur-
61.2.2 EARLY DIAGNOSTIC CRITERIA bance and postural abnormalities being commonest. These
latter cases presented with neuropsychiatric features, only
The following year, Burkhardt et al. (1988) listed 34 US cases in various combinations of cognitive impairment, paranoid
and carried out a simple meta-analysis. The most common delusions and visual or auditory hallucinations. Fourteen of
presentation was a ‘neurobehavioural syndrome’; memory the 15 people were demented before death, the exception pre-
impairment and other cognitive deficits were typical, all but senting itself with classical PD and later becoming depressed,
one eventually becoming demented. Psychotic features such irritable and mildly forgetful with frequent falls. Fluctuating
as depression, hallucinations and paranoia were seen in 10 cognition with episodic confusion for which no adequate
patients (29%), 2 patients being psychotic for many years underlying cause could be found, was observed in 80% of the
before developing other symptoms. Parkinsonian features, Nottingham cases. Byrne also drew attention to the frequent
the most common of which was rigidity, were usually over- occurrence of depression (20%) and psychosis (33%).
shadowed by dementia; in only five cases (15%), there were
no extrapyramidal features present. Duration of illness was
variable (1–20 years) with an end state of severe dementia,
rigidity, akinetic mutism, quadriparesis in flexion and emaci- 61.2.4 THE NEWCASTLE STUDIES
ation. The most common reported cause of death was aspira- At around the same time, the Newcastle upon Tyne group
tion pneumonia. Based upon these observations, Burkhardt identified 14 UK cases with the neuropathological features
et al. (1988) were the first to attempt a general description of of ‘SDLT’ (Perry et al., 1990) accounting for 15% of a series
the clinical syndrome associated with DLBD, distinguishing of dementia autopsies. These SDLT cases had been regarded,
it as separate from Parkinson’s disease (PD) with dementia when patients were alive, as clinically atypical ‘caus-
(PDD). They concluded that ‘DLBD should be suspected in ing much diagnostic perplexity’. Acute onset, fluctuating
Dementia with Lewy bodies: A clinical overview 705

course, more rapid deterioration, early and prominent fixed flexion deformities of the neck and trunk and
hallucinatory and behavioural disturbances and associ- severe gait ­impairment (McKeith et al., 1992a).
ated mild parkinsonian features were present. Two further
Newcastle series were reported soon after (McKeith et al.,
1992a, 1992b) and depressive symptoms, unexplained falls, 61.2.5 INTERNATIONAL CONSENSUS
observed disturbances of consciousness and excessive sen- CRITERIA
sitivity to side effects of neuroleptic medication were added
to the list of clinical characteristics with potential to distin- 61.2.5.1 First report of the DLB consortium
guish DLB pathology cases from AD. By the early 1990s, it was becoming apparent that DLB was a
Based upon these observations, new clinical diagnos- relatively common cause of dementia in old age and that the
tic criteria was proposed with an emphasis upon cognitive several research groups investigating it were adopting differ-
dysfunction and neuropsychiatric features, which could ent terminologies. The Consortium on DLB met in October
occur in the absence of extrapyramidal motor signs and an 1995 to agree on common clinical and pathological methods
attempt was made to describe the typical course of illness. and nomenclature. The consensus criteria (McKeith et al.,
1996) described the particular characteristics of the cogni-
The first stage is often recognized only in retro- tive impairments of DLB in some detail as differing from
spect and may extend back to 1–3 years’ pre- the dementia syndrome of AD, in which memory deficits
presentation with occasional minor episodes of predominate. In DLB, attentional deficits and prominent
forgetfulness, sometimes described as lapses of visuospatial dysfunction are the main features (Salmon and
concentration or ‘switching off’. A brief period Galasko, 1996). Probable DLB could be diagnosed if any two
of delirium is sometimes noted for the first time, of the three key symptoms are present, namely fluctuation,
often associated with genuine physical illness visual hallucinations or spontaneous motor features of par-
and/or surgical procedures. Disturbed sleep, kinsonism and possible DLB if only one is present.
nightmares and daytime drowsiness often per- Several retrospective and two prospective studies have
sist after recovery. examined the predictive accuracy of these original consen-
Progression to the second stage frequently sus clinical criteria for probable DLB (McKeith et al., 2000;
prompts psychiatric or medical referral. A more Litvan et al., 2003). They suggest that sensitivity of case
sustained cognitive impairment is established, detection is generally low at around 40% whereas, specific-
albeit, with marked fluctuations in severity. ity is much higher, typically around 90%, suggesting that
Recurrent episodes of confusion are accompa- the criteria are most useful for confirmation of diagnosis
nied by vivid hallucinatory experiences, visual (low false positive rate) but less helpful for case finding.
misidentification syndromes and topographi-
cal disorientations. Extensive medical screen-
ing is usually negative. Attentional deficits are 61.2.5.2 Second report of the DLB
apparent as apathy and daytime somnolence consortium
and sleep behaviour disorder may be severe.
Gait disorder and bradykinesia are often over- The Second International DLB Workshop met in July 1998
looked, particularly in elderly subjects. Frequent (McKeith et al., 1999). The objectives were to review devel-
falls occur either due to postural instability or opments since publication of the consensus guidelines and
due to syncope. to determine whether these were required to be modified
The third and final stage often begins with a further. It was recommended that the clinical consensus
sudden increase in behavioural disturbance lead- criteria should continue to be used in their current for-
ing to requests for sedation or hospital admission mat with the addition of two new supportive features,
by perplexed and exhausted carers. The natural namely, rapid eye movement (REM), REM behaviour dis-
course from this point is variable and obscured by order (RBD) (Section 61.3.1.3.3) and depression (Section
the high incidence of adverse reactions to neu- 61.3.1.3.4).
roleptic medication. For patients not receiving
or tolerating neuroleptics, a progressive decline
into severe dementia with dysphasia and dys- 61.2.5.3 Third report of the DLB consortium
praxia occurs over months or years, with death
usually due to cardiac or pulmonary disease. In order to address the perceived shortcomings of the origi-
During this terminal phase, patients show con- nal diagnostic criteria, the DLB Consortium met again in
tinuing behavioural disturbances including vocal 2003 to resolve improved methods of identifying cases ante-
and motor responses to hallucinatory phenom- mortem. No major amendments to the three core features
ena. Lucid intervals with some retention of recent of DLB were proposed, but better methods for their clinical
memory function and insight may still be appar- assessment were recommended. A new category of features
ent. Neurological disability is often profound with ‘suggestive’ of DLB was described comprising RBD, severe
706 Dementia

Table 61.1 Criteria for the clinical diagnosis of dementia with Lewy bodies (DLB)
1. Central feature (essential for a diagnosis of possible or probable DLB)
Dementia is defined as progressive cognitive decline of sufficient magnitude to interfere with normal, social or
occupational function. Prominent or persistent memory impairment may not necessarily occur in the early stages but is
usually evident with progression of the disease. Deficits on tests of attention, executive function and visuospatial ability
may be especially prominent.
2. Core features (two core features are sufficient for a diagnosis of probable DLB, one for possible DLB)
Fluctuating cognition with pronounced variations in attention and alertness.
Recurrent visual hallucinations that is typically well formed and detailed.
Spontaneous features of parkinsonism.
3. Suggestive features (if one or more of these is present in the presence of one or more core features, a diagnosis of
probable DLB can be made. In the absence of any core features, one or more suggestive features are sufficient for
possible DLB. Probable DLB should not be diagnosed on the basis of suggestive features alone).
Rapid eye movement (REM) sleep behaviour disorder.
Severe neuroleptic sensitivity.
Low dopamine transporter uptake in basal ganglia demonstrated by single photon emission computed tomography
(SPECT) or positron emission tomography (PET) imaging.
4. Supportive features (commonly present but not proven to have diagnostic specificity)
Repeated falls and syncope.
Transient, unexplained loss of consciousness.
Severe autonomic dysfunction, e.g. orthostatic hypotension, urinary incontinence.
Hallucinations in other modalities.
Systematized delusions.
Depression.
Relative preservation of medial temporal lobe structures on computerized tomography/magnetic resonance imaging
(CT/MRI) scan.
Generalized low uptake on SPECT/PET perfusion scan with reduced occipital activity.
Abnormal (low uptake) meta-iodobenzylguanidine (MIBG) myocardial scintigraphy.
Prominent slow wave activity on electroencephalogram (EEG) with temporal lobe transient sharp waves.
5. A diagnosis of DLB is less likely in the following cases:
In the presence of cerebrovascular disease evident as focal neurological signs or on brain imaging.
In the presence of any other physical illness or brain disorder sufficient to account in part or in total for the clinical
picture.
If parkinsonism only appears for the first time at a stage of severe dementia.
6. Temporal sequence of symptoms
DLB should be diagnosed when dementia occurs before or concurrently with parkinsonism (if it is present). The term
Parkinson’s disease (PD) with dementia (PDD) should be used to describe dementia that occurs in the context of
well-established Parkinson’s disease. In practice, setting the term that is most appropriate to the clinical situation
should be used and generic terms such as Lewy body (LB) disease are often helpful. In research studies that require
distinction between DLB and PDD, the existing 1-year rule between the onset of dementia and parkinsonism, DLB
continues to be recommended. Adoption of other time periods will simply confound data pooling or comparison
between studies. In other research settings, that may include clinico-pathologic studies and clinical trials, both clinical
phenotypes may be considered collectively under categories such as LB disease or alpha-synucleinopathy (AS).

neuroleptic sensitivity and abnormal dopamine transporter to an autopsy verified sample reported that cases with Braak
(DAT) neuroimaging. If one or more of these suggestive fea- stage 5 and 6 Alzheimer pathology were still unlikely to be
tures is present, in addition to one or more core features, detected ante-mortem. Such cases tend to lack core or sugges-
a diagnosis of probable DLB can be made – see Table 61.1. tive DLB features and clinically resemble AD (Weisman et al.,
Possible DLB can be diagnosed if one or more suggestive 2007). It has also been shown that the DLB clinical syndrome is
features are present in a patient with dementia even in the modified by the severity of Alzheimer neuritic pathology, while
absence of any core features. The revised criteria are also β-amyloid load has no effect (Tiraboschi et al., 2015) on it. This
explicit about the importance to be attached to clinical and is consistent with the proposal made by the third Consortium
radiological evidence of cerebrovascular disease. group that the likelihood of a patient having the typical DLB
These improved criteria have been suggested to detect 25% clinical syndrome is ‘directly related to the severity of Lewy-
more DLB cases than the previous version (Aarsland et al., related pathology and inversely related to the severity of con-
2008) although, a retrospective application of the new criteria current AD-type pathology’. Given that AD-type pathology is
Dementia with Lewy bodies: A clinical overview 707

frequently present in DLB, there will, therefore, be a significant self-contained part of the CAMDEX (CAMCOG) scores
number of patients who will always present symptoms difficult were identical in DLB and AD, significant differences were
to identify solely on clinical grounds alone. This group, who are found in mean scores for the delayed recall subtest (DLB
best described pathologically as AD+LB or as LBVAD (Hansen performing better) and the visuospatial praxis subtest (DLB
et al., 1990) can probably be reliably identified by additional performing worse) (Walker et al., 1996). With the progres-
investigations and biomarkers only – see Section 61.4. sion of dementia, this selective pattern of cognitive deficits
may be lost, making differential diagnosis based on neuro-
61.2.5.4 Future reports of the DLB psychological examination difficult during the later stages.
consortium DLB is particularly characterized by increased vari-
ability in performance on cognitive tasks, within and
The DLB Consortium met again in December 2015 and is between patients and when compared to age-matched
likely to publish an updated report of the proceedings in 2016. controls and patients with AD. This variability is most
evident in executive and attentional tasks. Wide test –
retest variation in performance of timed computer-based
tasks (Walker et al., 2000a) have been proposed as a reli-
61.3 DIFFERENTIAL DIAGNOSIS OF able and widely applicable quantified measure, which is
DLB IN CLINICAL PRACTICE well correlated with clinical expert estimates of fluctuat-
ing performance.
61.3.1 CLINICAL PRESENTATIONS OF DLB
61.3.3 CORE FEATURES
DLB patients may present psychiatric disorders (cognitive
impairment, psychosis or behavioural disturbance), inter- Review of nine studies that report clinical details on 190
nal medicine (acute state of confusion or syncope) or neu- autopsies confirmed DLB cases and compared them to
rology (movement disorder or disturbed consciousness). 261 AD cases, providing a useful source of frequency rates
The details of clinical assessment and differential diagnoses of the core features of DLB (Byrne et al., 1989; McKeith
to a large extent are shaped by these symptoms and specialty et al., 1992a, 1992b; Förstl et al., 1993; Galasko et al., 1996;
biases (McKeith et al., 1995). In all cases, a detailed history Hely et al., 1996; Klatka et al., 1996; Weiner et al., 1996;
from patient and reliable informants should document the Ala et al., 1997). The wide ranges reported most likely
time of onset of relevant key symptoms, the nature of their reflect sampling biases and different methods of symptom
progression and their effects on social, occupational and ascertainment.
personal function. Functional disability in DLB is generally
greater than in AD patients with similar severity of cogni-
61.3.3.1 Fluctuation
tive impairment. Most of the extra burden is due to motor
impairments that cause difficulty with mobility, feeding Fluctuations in cognitive function, which may vary over
and toileting (McKeith et al., 2006). Given the additional minutes, hours or days, occur in 58% of DLB cases at the
disabilities associated with DLB, it is not surprising that time of presentation (range 8%–85%) and are observed at
care costs have been estimated twice compared to those for some point during the course of the illness in 75% (45%–
patients with AD (Boström et al., 2007a) and that carers rate 90%). Substantial changes in mental state and behaviour
DLB patients’ Qol as significantly worse than that of AD may be seen within the duration of a single interview or
(Boström et al., 2007b) patients. between consecutive examinations. Fluctuation includes
and indeed may be based on pronounced variations in
61.3.2 COGNITIVE IMPAIRMENT attention and alertness. Excessive daytime drowsiness
with transient confusion on waking commonly occurs.
Although, brief bedside tests of mental status, e.g. Mini- Episodes may last from a few seconds to several hours and
Mental State Examination (MMSE) may confirm the pres- can be ascertained by caregiver report, observer rating
ence of cognitive impairment in DLB, total scores are usually (Walker et al., 2000b) or using computer-based measures
equivalent to AD and unhelpful in differential diagnosis. of variation in attentional performance (Walker et al.,
More detailed psychometric examination is usually required 2000a). Questions such as ‘are there episodes when his/her
to reveal a profile of deficits that helps to identify DLB from thinking seems quite clear and then becomes muddled?’
AD or other dementias (Ferman et al., 2006). Prominent may be useful opening probes although, up to 75% of car-
deficits on tests of executive function and problem-solving, ers will respond in the affirmative regardless of AD/DLB
such as the Trail Making Test (TMT) and verbal fluency for diagnosis (Bradshaw et al., 2004; Ferman et al., 2004).
categories and letters, may be useful clinical discrimina- Objective questions about daytime drowsiness and leth-
tors (Salmon and Galasko, 1996), as may disproportionate argy, daytime sleep of 2 or more hours, staring into space
impairment on tests of visuospatial performance, present for long periods and episodes of disorganized speech are
in up to three quarters of DLB patients at first evaluation more informative. The presence of three or four features of
(Tiraboschi et al., 2006). Although, total cognitive and this composite occurred in 63% of DLB patients compared
708 Dementia

to 12% of AD patients and 0.5% of normal elderly persons 61.3.4 SUGGESTIVE FEATURES


(Ferman et al., 2004).
61.3.4.1 REM sleep behaviour disorder
61.3.3.2 Visual hallucinations RBD is a parasomnia characterized by loss of normal skeletal
Visual hallucinations are present in 33% of DLB cases at muscle atonia during REM sleep with resultant motor activ-
the time symptoms present themselves (range 11%–64%) ity and ‘acting out of dreams’. Patients demonstrate a variety
and occur at some point during the course of the illness of movements ranging from verbal outbursts to pugilistic
in 46% (13%–80%). Most patients experience complex movements and even more dramatic motor activity. Injuries
hallucinations daily, normally lasting for at least several to patients and bed partners are common. Estimates from
minutes. They commonly see people or animals that do prospective studies in specialist centres place the frequency
not exist in reality and the experiences are usually per- of RBD at 50%–80% in DLB (Boeve et al., 2004) and it is
ceived as unpleasant. Simple hallucinations are rare. uncommon in patients with AD. The diagnosis is made on
Neuropsychiatric symptoms commonly coexisting with clinical grounds and careful questioning of bed partners is
hallucinations are apathy, sleep disturbance and anxiety therefore vital. Use of a validated instrument such as the
(Mosimann et al., 2006). Carers are more likely to give Mayo Sleep Questionnaire (MSQ), is recommended since
‘don’t know’ answers to mild or moderately demented techniques such as video polysomnography (PSG), utiliz-
patients about visual hallucinations and the use of a semi- ing audiovisual footage to confirm the physical features of
structured interview can increase both the quantity and RBD in addition to electromyography, electro-oculography,
quality of information about a symptom, which is present EEG and oxygen saturation recordings during sleep are
in the early stages and strongly predictive of a DLB diag- generally unavailable for most patients. The history of RBD
nosis (Tiraboschi et al., 2006; Mosimann et al., 2008). It is may precede the diagnosis of dementia by several years and
the persistence of visual hallucinations in DLB (McShane may either persist or diminish following the onset of cogni-
et al., 1995) that helps distinguish them from the episodic tive decline. REM sleep-wakefulness dissociations charac-
perceptual disturbances that occur transiently in demen- teristic of narcolepsy may explain several features of DLB
tias of other aetiology or during a delirium provoked by an including daytime hypersomnolence, visual hallucinations
external cause. It has been suggested that they arise from and cataplexy. Sleep disorders may contribute to fluctua-
a combination of faulty perceptual processing of environ- tions typical of DLB and their treatment may improve fluc-
mental stimuli and less detailed recollection of experience tuations and QoL.
combined with intact image generation (Collerton et al.,
2005). Visual hallucinations in DLB are associated with
greater deficits in cortical acetylcholine (Perry et al.,1991) 61.3.4.2 Severe neuroleptic sensitivity
and predict better response to cholinesterase inhibitors The hypothesis of an abnormal sensitivity to adverse effects
(McKeith et al., 2004b). of neuroleptic medication was based upon the observation
that 57% of a series of DLB patients deteriorated rapidly
61.3.3.3 Motor parkinsonism after either receiving neuroleptics for the first time or fol-
lowing an increase in dosage (McKeith et al., 1992a, 1992b).
Extrapyramidal signs (EPS) are reported in 25%–50% of
Mortality risk, estimated by survival analysis was increased
DLB cases at diagnosis and the majority develop some EPS
by a factor of 2.7. A severe adverse reaction to neuroleptic
during the natural course. Up to 25% of autopsy confirmed
medication may be an important diagnostic indicator of
cases may, however, have no record of EPS indicating that
underlying LB disorder but tolerance of neuroleptics does
parkinsonism is not necessary for a clinical diagnosis of
not rule out DLB. Neuroleptic challenge is never justified
DLB. Initial suggestions that parkinsonism in DLB is mild
as a diagnostic test and prescribing neuroleptic is routinely
has not been supported by studies finding equal severity
and desirably avoided in patients suspected of having DLB.
with non-demented PD patients (Louis et al., 1997; Aarsland
Newer atypical antipsychotics used at low dose may be safer
et al., 2001) with similar annual progression rates in motor
in this regard but sensitivity reactions have been docu-
Unified Parkinson’s Disease Rating Scale (UPDRS) scores
mented with most and they should be used with great cau-
(Ballard et al., 2000). The pattern of EPS in DLB shows an
tion (McKeith et al., 2004a; Aarsland et al., 2005).
axial bias with greater postural instability and facial impas-
sivity and a tendency towards fewer tremors consistent
with greater ‘non-dopaminergic’ motor involvement (Burn 61.3.4.3 Low DAT uptake in basal ganglia
et al., 2003). The degree of cognitive impairment appears
to have no influence on the occurrence of EPS, although, Functional imaging of the DAT defines integrity of the
more demented patients may have difficulty in following nigrostriatal dopaminergic system and was originally
instructions on formal motor tests. A modified version developed to assist diagnosis of patients with tremor of
of the UPDRS, which allows for this instruction has been uncertain aetiology (Marshall and Grosset, 2003). Positron
developed (Ballard et al., 1997). emission tomography (PET) and single photon emission
Dementia with Lewy bodies: A clinical overview 709

computed tomography (SPECT) ligands are available for diarrhoea (Del-Ser et al., 1996). Orthostatic hypotension
this purpose and both have subsequently been useful in the and carotid sinus sensitivity appear to occur more com-
discrimination of DLB from AD. 123I-radiolabelled 2 beta-­ monly in DLB than AD or age-matched controls (Kenny
carbomethoxy-3 beta-(4-iodophenyl)-N-(3-fluoropropyl) et al., 2004; Allan et al., 2007) and probably contribute to the
nortropane (FP-CIT) has been the most investigated and a high rates of syncope and falls that occur in this population.
sensitivity of 78% for detecting probable DLB with a speci-
ficity of 85% for AD and 94% for controls, has been reported 61.3.5.3 Systematized delusions and
(O’Brien et al., 2004). A multicentre clinical trial subse-
hallucinations in other modalities
quently demonstrated similar results across a wide variety
of settings (McKeith et al., 2007) and regulatory approval for Delusions are common in DLB, occurring in about half of
distinguishing probable DLB from AD with FP-CIT imaging the patients. They are usually based on recollections of hal-
now exists in Europe. The most useful clinical application lucinations and perceptual disturbances and consequently
of the test however, is probably in cases of possible DLB or have a fixed, complex and bizarre content that often contrasts
where diagnostic uncertainty is high. A longitudinal study with the mundane and poorly formed persecutory ideas
found that of 12/19 people (63%) who were diagnosed of encountered in AD, which are based on forgetfulness and
possible DLB accompanied by an abnormal DAT scan were confabulation. Auditory, olfactory and tactile hallucinations
re-diagnosed with probable DLB at 1-year follow-up and occur lesser often but can present striking features in some
subsequent clinical follow-ups. By contrast, 100% of cases DLB cases leading to initial diagnoses of late onset psycho-
initially diagnosed as possible DLB but reclassified as AD a sis (Birkett et al., 1992) and temporal lobe epilepsy (McKeith
year later had normal DAT scans at first evaluation (O’Brien et al., 1992a). In such circumstances, severe neuroleptic sen-
et al., 2009). sitivity reactions may indicate the true underlying cause.

61.3.5 SUPPORTIVE FEATURES 61.3.5.4 Depression


A detailed discussion of every feature identified as support- Depressive symptoms are frequent in most neuropsychi-
ive of a diagnosis of DLB is beyond the scope of this chap- atric syndromes and often of little differential diagnostic
ter. Clinical symptoms are summarized briefly below and value. However, depression was diagnosed at first consul-
investigations including neuroimaging are detailed out in tation in 38% of DLB patients compared to 16% with AD
Section 61.4. (p < 0.05) and was the primary reason for referral in 60% of
the patients in whom it was present (McKeith et al., 1992a).
61.3.5.1 Repeated falls, syncope and Klatka et al. (1996) found depression in 14/28 (50%) DLB,
transient losses of consciousness 8/58 (14%) AD and 15/26 (58%) PD cases with autopsy con-
firmation. No particular characteristics of depression have
Dementia of any aetiology is probably a risk factor for all been described in DLB but apathy and psychomotor retar-
three of these clinical features and it can be difficult to dation are to be expected in this population and may con-
clearly distinguish between them. Repeated falls may be due found the assessment of affective state.
to posture, gait and balance difficulties in DLB and are a
significant cause of hospitalization contributing to reduced 61.3.5.5 Differential diagnosis
survival (Hanyu et al., 2009). Syncopal attacks in DLB with
complete loss of consciousness and muscle tone and tran- There are four main categories of disorder that should be
sient episodes of unresponsiveness without loss of muscle considered in the differential diagnosis of DLB. These are
tone are probably related phenomena representing one as follows:
extreme of fluctuating attention and cognition. Episodes of
each type occur in about 20% of DLB cases at primal stages 1. Other dementia syndromes
and up to a half during the whole course of illness (McKeith 2. Other causes of delirium
et al., 1992a, 1992b). Such episodes frequently lead to a mis- 3. Other neurological syndromes including PD
diagnosis of vascular dementia because of the suspicion of 4. Other psychiatric disorders
transient ischaemic attack.

61.3.5.2 Autonomic dysfunction 61.3.5.6 Other dementia syndromes


Autonomic dysfunction that affects both sympathetic and Sixty-five per cent of autopsy confirmed DLB cases meet
parasympathetic systems is a significant, common and the National Institute of Neurological and Communicative
often early feature of DLB presenting itself as orthostatic Diseases and Stroke–Alzheimer’s Disease and Related
hypotension, carotid sinus hypersensitivity, reduced secre- Disorders Association (NINCDS-ADRDA) clinical criteria
tions (lacrimal, mouth, sweat), urinary retention, erectile for probable or possible AD (McKeith et al., 1994) and this is
dysfunction and bowel problems, both constipation and the most frequent clinical misdiagnosis of DLB. Conversely,
710 Dementia

12%–36% of cases meeting NINCDS criteria for a clinical decide which term is the most appropriate for each indi-
diagnosis of AD are unexpectedly found to have LB pathol- vidual patient and carefully explain the terminology to the
ogy at autopsy (Burns et al., 1990; Hansen et al., 1990). patient and his or her caregivers.
There is a large ‘overlap’ group of patients who have both LB
and Alzheimer pathologies and who cannot easily be clas- 61.3.5.9 Other neurological syndromes
sified by clinical criteria alone. Up to a third of DLB cases
can be misclassified as having vascular dementia by virtue Other atypical parkinsonian syndromes associated with
of symptoms such as fluctuating nature and course of ill- poor levodopa response, cognitive impairment and postural
ness. Pyramidal and focal neurological signs are however, instability include progressive supranuclear palsy (Fearnley
usually absent. et al., 1991), multisystem atrophy and corticobasal degen-
eration. The development of myoclonus in patients with a
rapidly progressive form of DLB may lead the clinician to
61.3.5.7 Other causes of delirium suspect Creutzfeldt–Jacob disease (CJD). Syncopal epi-
In patients with intermittent delirium, appropriate exami- sodes in DLB are often incorrectly attributed to transient
nation and laboratory tests should be performed during the ischaemic attacks, despite an absence of focal neurological
acute phase to maximize the chances of detecting infec- signs. Recurrent disturbances in consciousness accompa-
tive, metabolic, inflammatory or other aetiological factors. nied by complex visual hallucinations may suggest complex
Pharmacological causes are particularly common in elderly partial seizures (temporal lobe epilepsy) and vivid dream-
patients. Although, the presence of any of these features ing with violent movements during sleep and may meet cri-
makes a diagnosis of DLB less likely, comorbidity is not teria for RBD.
unusual in elderly patients and the diagnosis should not be
excluded simply on this basis. 61.3.5.10 Other psychiatric disorders
If a patient spontaneously develops parkinsonian features
61.3.5.8 Patients with a previous diagnosis or cognitive decline or shows excessive sensitivity to neu-
of PD roleptic medication, in the course of late onset delusional
disorder, depressive psychosis or hypomania (Birkett et al.,
Dementia in PD (discussed fully in Chapter 64) is often 1992; Mullan et al., 1996), DLB should be considered.
similar to DLB with respect to fluctuating neuropsychologi-
cal function and neuropsychiatric features. Such patients
who develop the typical features of DLB only after many
years of motor disability present some problems of classi-
fication. Anti-parkinsonian medications are usually held 61.4 NEW AND FUTURE CLINICAL
responsible for hallucinations and confusion in PD, but DEVELOPMENTS
research findings do not fully support this clinical impres-
sion. There has been considerable debate as to whether PD, 61.4.1 RECOGNITION OF DLB AS A
PDD and DLB are simply different clinical presentations of CLINICAL DIAGNOSIS IN FORMAL
the same underlying biological process, i.e. LB disease and CLASSIFICATIONS
indeed, whether LB disease might be a better clinical term
to use for the whole spectrum of such phenotypes (McKeith, Although, the DLB consensus criteria was constructed for
2009). The current recommendation is to apply an arbi- both research and general clinical use, it is only recently
trary 1-year rule that proposes that the onset of dementia that they have been formally incorporated into the official
within 12 months of the onset of parkinsonism qualifies as diagnostic classification systems. Diagnostic and Statistical
DLB and more than 12 months of parkinsonism before the Manual of Mental Disorders, Fifth Edition (DSM-5)
onset of dementia qualifies as PDD (McKeith et al., 2005). (American Psychiatric Association, 2013) contains catego-
Using diagnostic labels such as PDD and DLB that describe ries to identify major and mild neurocognitive disorder with
the order of onset of symptoms is generally helpful in the LBs that are consistent with the 2006 consensus criteria,
diagnosis and management of most clinical cases and the both in the diagnostic features listed and the possible/prob-
majority of patients and carers (and clinicians) are able to able distinction. DSM-5 also contains a separate category of
understand that the diagnosis is predictable, e.g. DLB is major and mild neurocognitive disorder due to PD, which
‘caused by’ LB disease. In some patients, particularly those uses the previously stated 1-year rule to distinguish from
with an admixture of neurological and psychiatric symp- DLB. It is anticipated that the International Classification of
toms occurring in relatively short order, a single label of LB Disease, Eleventh Revision (ICD-11) will similarly incor-
disease may serve as the best primary diagnosis, qualified porate DLB into the formal dementia subtype classifica-
by a secondary descriptive epithet, for example, LB disease tion and together these changes should assist case detection
with parkinsonism or LB disease with dementia or LB dis- rates, clinical service development and reimbursement and
ease with parkinsonism and dementia. Clinicians need to research funding.
Dementia with Lewy bodies: A clinical overview 711

61.4.2 PRE-DEMENTIA AND PRODROMAL Allan, L.M., Ballard, C.G., Allen, J. et al. (2007). Autonomic
DIAGNOSIS OF DLB dysfunction in dementia. Journal of Neurology,
Neurosurgery, and Psychiatry, 78: 671–677.
Possibly the most significant clinical development in the American Psychiatric Association (2013). Diagnostic
whole dementia field over the last decade has been a shift and Statistical Manual of Mental Disorders: DSM-5,
away from diagnosing at a mild/moderate symptomatic Fifth Edition. Washington, DC: American Psychiatric
stage to prodromal or even pre-symptomatic diagnosis. Association.
The main justification for this shift to earlier diagnosis is Ballard, C.G., McKeith, I., Burn, D. et al. (1997). The UPDRS
that preventative treatment will only be effective if given scale as a means of identifying extrapyramidal signs
before extensive neurochemical and anatomic pathological in patients suffering from dementia with Lewy bodies.
changes have occurred in the brain. It follows that trials of Acta Neurolgica Scandinavica, 96: 366–371.
potential preventative agents will only be possible once reli- Ballard, C., O’Brien, J., Swann, A. et al. (2000). One year
able methods of prodromal diagnosis have been established. follow-up of parkinsonism in dementia with Lewy bodies.
Efforts to start developing diagnostic methods and cri- Dementia and Geriatric Cognitive Disorders, 11: 219–222.
teria for LB disease are already under way (Donaghy and Birkett, D.P., Desouky, A., Han, L. and Kaufman, M. (1992).
McKeith, 2014). Since we do not yet know why some cases Lewy bodies in psychiatric patients. International
progress to PD and others to DLB, very early identification Journal of Geriatric Psychiatry, 7: 235–240.
of LB disease would identify individuals at risk of an uncer- Boeve, B.F., Silber, M.H. and Ferman, T.J. (2004). REM
tain future clinical course and progression. Criteria for the sleep behavior disorder in Parkinson’s disease and
identification of early cases at high risk of progression to dementia with Lewy bodies. Journal of Geriatric
dementia syndromes of the DLB type might be made at a Psychiatry and Neurology, 17 (3): 146–157.
stage when an individual has at least one clinical feature Boström, F., Jonsson, L., Minthon, L., Londos, E. (2007a).
suggestive of prodromal DLB, e.g. cognitive or neuropsy- Patients with Lewy body dementia use more resources
chiatric features and at least one biomarker supportive of than those with Alzheimer’s disease. International
LB disease. Given the large number of potential prodromal Journal of Geriatric Psychiatry, 22(8): 713–719.
symptoms and existence of several biomarker candidates, Boström, F., Jonsson, L., Minthon, L., Londos, E. (2007b).
it is anticipated that formal criteria for prodromal DLB are Patients with dementia with Lewy bodies have more
still distant. The MCI stage of dementia with Lewy bodies impaired quality of life than patients with Alzheimer
(MCI-DLB) category is however, already being used infor- disease. Alzheimer Disease & Associated Disorders,
mally in some memory clinic settings with evidence that 21(2): 150–154.
non-amnestic MCI subjects are more likely to progress to Bradshaw, J., Saling, M., Hopwood, M. et al. (2004).
DLB rather than AD, particularly, if they have additional Fluctuating cognition in dementia with Lewy bodies and
DLB core or suggestive features (Ferman et al., 2013). DSM-5 Alzheimer’s disease is qualitatively distinct. Journal of
also contains criteria for ‘mild neurocognitive disorder with Neurology, Neurosurgery, and Psychiatry, 75: 382–387.
LB’ although, these have not yet been validated. Burkhardt, C.R., Filley, C.M., Kleinschmidt-DeMasters, B.K.
et al. (1988). Diffuse Lewy body disease and progres-
REFERENCES sive dementia. Neurology, 38: 1520–1528.
Burn, D.J., Rowan, E.N., Minett, T. et al. (2003).
Aarsland, D., Ballard, C., McKeith, I. et al. (2001). Extrapyramidal features in Parkinson’s disease with
Comparison of extrapyramidal signs in dementia and without dementia and dementia with Lewy bod-
with Lewy bodies and Parkinson’s disease. Journal ies: A cross-sectional comparative study. Movement
of Neuropsychiatry and Clinical Neurosciences, 13: Disorders, 18 (8): 884–889.
374–379. Burns, A., Luthert, P., Levy, R. et al. (1990). Accuracy
Aarsland, D., Ballard, C., Larsen, J.P. et al. (2005). of clinical diagnosis of Alzheimer’s disease. British
Neuroleptic sensitivity in Parkinson’s disease and par- Medical Journal, 301: 1026.
kinsonian dementias. The Journal of Clinical Psychiatry, Byrne, E.J., Lennox, G., Godwin-Austen, R.B. et al. (1991).
66 (5): 633–637. Dementia associated with cortical Lewy bodies.
Aarsland, D., Rongve, A., Nore, S.P. et al. (2008). Proposed diagnostic criteria. Dementia, 2: 283–284.
Frequency and case identification of dementia with Byrne, E.J., Lennox, G., Lowe, J. and Godwin-Austen, R.B.
Lewy bodies using the revised consensus criteria. (1989). Diffuse Lewy body disease: Clinical features
Dementia and Geriatric Cognitive Disorders, 26 (5): in 15 cases. Journal of Neurology, Neurosurgery, and
445–452. Psychiatry, 52: 709–717.
Ala, T.A., Yang, K.-H., Sung, J.H. and Frey, W.H.I. (1997). Collerton, D., Perry, E. and McKeith, I. (2005). Why people
Hallucinations and signs of parkinsonism help distin- see things that are not there: A novel perception and
guish patients with dementia and cortical Lewy bodies attention deficit model for recurrent complex visual hal-
from patients with Alzheimer’s disease. Journal of lucinations. The Behavioral and Brain Sciences, 28 (6):
Neurology, Neurosurgery, and Psychiatry, 62: 16–21. 737–757.
712 Dementia

Crystal, H.A., Dickson, D.W., Lizardi, J.E. et al. (1990). Kenny, R.A., Shaw, F.E., O’Brien, J.T. et al. (2004). Carotid
Antemortem diagnosis of diffuse Lewy body disease. sinus syndrome is common in dementia with Lewy
Neurology, 40: 1523–1528. bodies and correlates with deep white matter lesions.
Del-Ser, T., Munoz, D.G. and Hachinski, V. (1996). Temporal Journal of Neurology, Neurosurgery, and Psychiatry,
pattern of cognitive decline and incontinence is dif- 75: 966–971.
ferent in Alzheimer’s disease and diffuse Lewy body Klatka, L.A., Louis, E.D. and Schiffer, R.B. (1996).
disease. Neurology, 46: 682–686. Psychiatric features in diffuse Lewy body disease:
Dickson, D.W., Davies, P., Mayeux, R. et al. (1987). Diffuse Findings in 28 pathologically diagnosed cases.
Lewy body disease. Neuropathological and biochemical Neurology, 47: 1148–1152.
studies of six patients. Acta Neuropathologica, 75: 8–15. Kosaka, K., Yoshimura, M., Ikeda, K. and Budka, H. (1984).
Donaghy, P.C. and McKeith, I.G. (2014). The clinical char- Diffuse type of Lewy body disease: Progressive
acteristics of dementia with Lewy bodies and a consid- dementia with abundant cortical Lewy bodies and
eration of prodromal diagnosis. Alzheimer’s Research senile changes of varying degree – A new disease?
and Therapy, 6 (4): 46. Clinical Neuropathology, 3: 185–192.
Fearnley, J.M., Revesz, T., Brooks, D.J. et al. (1991). Diffuse Lennox, G., Lowe, J., Byrne, E.J. et al. (1989). Diffuse Lewy
Lewy body disease presenting with a supranuclear body disease. Lancet, 8633 (1): 323–324.
gaze palsy. Journal of Neurology, Neurosurgery, and Litvan, I., Bhatia, K.P., Burn, D.J. et al. (2003). SIC Task
Psychiatry, 54 (2): 159–161. Force Appraisal of clinical diagnostic criteria for
Ferman, T., Smith, G.E., Boeve, B.F. et al. (2004). DLB parkinsonian disorders. Movement Disorders, 18 (5):
fluctuations: Specific features that reliably differentiate 467–486.
from AD and normal aging. Neurology, 62 (2): 181–187. Louis, E., Klatka, L., Lui, Y. et al. (1997). Comparison of
Ferman, T.J., Smith, G.E., Boeve, B.F. et al. (2006). extrapyramidal features in 31 pathologically con-
Neuropsychological differentiation of dementia with firmed cases of diffuse Lewy body disease and 34
Lewy bodies from normal aging and Alzheimer’s dis- pathologically confirmed cases of Parkinson’s disease.
ease. The Clinical Neuropsychologist, 20 (4): 623–636. Neurology, 48: 376–380.
Ferman, T.J., Smith, G.E., Kantarci, K. et al. (2013). Marshall, V. and Grosset, D. (2003). Role of dopamine
Nonamnestic mild cognitive impairment progresses transporter imaging in routine clinical practice.
to dementia with Lewy bodies. Neurology, 81 (23): Movement Disorders, 18: 1415–1423.
2032–2038. McKeith, I., Fairbairn, A., Perry, R. et al. (1992b). Neuroleptic
Forster, E. and Lewy, F.H. (1912). Paralysis agitans. I. sensitivity in patients with senile dementia of Lewy body
Pathologische anatomie. In Lewandowsky, M. (ed.), type. British Medical Journal, 305: 673–678.
Handbuch der Neurologie. Berlin, Germany: Springer, McKeith, I.G. (2009). DLB and PDD: The same or differ-
pp. 920–933. ent? Is there a debate? International Psychogeriatrics,
Förstl, H., Burns, A., Luthert, P. et al. (1993). The Lewy-body 21: 220–224.
variant of Alzheimer’s disease. Clinical and pathological McKeith, I.G., Ballard, C.G., Perry, R.H. et al. (2000).
findings. British Journal of Psychiatry, 162: 385–392. Prospective validation of consensus criteria for the
Förstl, H., Levy, R. (1991). FH Lewy on Lewy bodies, diagnosis of dementia with Lewy bodies. Neurology,
Parkinsonism and dementia. International Journal of 54: 1050–1058.
Geriatric Psychiatry, 6: 757–766. McKeith, I.G., Dickson, D.W., Lowe, J. et al. (2005).
Galasko, D., Katzman, R., Salmon, D.P. et al. (1996). Clinical Diagnosis and management of dementia with
and neuropathological findings in Lewy body demen- Lewy bodies: Third report of the DLB consortium.
tias. Brain and Cognition, 31: 166–175. Neurology, 65 (12): 1863–1872.
Gibb, W.R.G., Esiri, M.M. and Lees, A.J. (1987). Clinical McKeith, I.G., Fairbairn, A.F., Perry, R.H. and Thompson, P.
and pathological features of diffuse cortical Lewy (1994). The clinical diagnosis and misdiagnosis of senile
body disease (Lewy body dementia). Brain: A Journal dementia of Lewy body type (SDLT). British Journal of
of Neurology, 110 (Pt. 5): 1131–1153. Psychiatry, 165: 324–332.
Hansen, L., Salmon, D., Galasko, D. et al. (1990). The Lewy McKeith, I.G., Galasko, D., Kosaka, K. et al. (1996).
body variant of Alzheimer’s disease: A clinical and Consensus guidelines for the clinical and pathologic
pathologic entity. Neurology, 40: 1–8. diagnosis of dementia with Lewy bodies (DLB): Report
Hanyu, H., Sato, T., Hirao, K. et al. (2009). Differences in of the consortium on DLB international workshop.
clinical course between dementia with Lewy bod- Neurology, 47: 1113–1124.
ies and Alzheimer’s disease. European Journal of McKeith, I.G., Galasko, D., Wilcock, G.K. and Byrne, E.J.
Neurology, 16 (2): 212–217. (1995). Lewy body dementia – Diagnosis and treat-
Hely, M.A., Reid, W.G.J., Halliday, G.M. et al. (1996). ment. British Journal of Psychiatry, 167: 709–717.
Diffuse Lewy body disease: Clinical features in nine McKeith, I., Mintzer, J., Aarsland, D. et al. (2004a).
cases without coexistent Alzheimer’s disease. Journal of Dementia with Lewy bodies. The Lancet Neurology, 3:
Neurology, Neurosurgery, and Psychiatry, 60: 531–538. 19–28.
Dementia with Lewy bodies: A clinical overview 713

McKeith, I., O’Brien, J., Walker, Z. et al. (2007). Sensitivity Perry, R.H., Irving, D., Blessed, G. et al. (1990). Senile
and specificity of dopamine transporter imaging with dementia of Lewy body type. A clinically and neuro-
(123) I-FP-CIT SPECT in dementia with Lewy bodies: A pathologically distinct form of Lewy body dementia
phase III, multicentre study. The Lancet Neurology, 6 in the elderly. Journal of Neurological Sciences, 95:
(4): 305–313. 119–139.
McKeith, I.G., Perry, R.H., Fairbairn, A.F. et al. (1992a). Rahkonen, T., Eloniemi-Sulkava, U., Rissanen, S. et al.
Operational criteria for senile dementia of Lewy body (2003). Dementia with Lewy bodies according to the
type (SDLT). Psychological Medicine, 22: 911–922. consensus criteria in a general population aged 75
McKeith, I.G., Perry, E.K. and Perry, R.H. (1999). Report years or older. Journal of Neurology, Neurosurgery,
of the second dementia with Lewy body international and Psychiatry, 74 (6): 720–724.
workshop. Diagnosis and treatment. Neurology, 53 (5): Salmon, D. and Galasko, D. (1996). Neuropsychological
902–905. aspects of Lewy body dementia. In Perry, R.,
McKeith, I.G., Rowan, E., Askew, K. et al. (2006). More McKeith, I. and Perry, E. (eds.), Dementia with Lewy
severe functional impairment in dementia with Lewy Bodies. New York, NY: Cambridge University Press,
bodies than Alzheimer disease is related to extrapyra- pp. 99–114.
midal motor dysfunction. American Journal of Geriatric Savica, R., Brandon, R., Grossardt, M.S. et al. (2013).
Psychiatry, 14 (7): 582–588. Incidence of dementia with Lewy bodies and
McKeith, I.G., Wesnes, K.A., Perry, E. and Ferrara, R. Parkinson disease dementia. JAMA Neurology, 70 (11):
(2004b). Hallucinations predict attentional improve- 1396–1402.
ments with rivastigmine in dementia with Lewy bod- Stevens, T., Livingston, G., Kitchen, G. et al. (2002).
ies. Dementia and Geriatric Cognitive Disorders, 18: Islington study of dementia subtypes in the commu-
94–100. nity. British Journal of Psychiatry, 180: 270–276.
McShane, R., Gedling, K., Reading, M. et al. (1995). Tiraboschi, P., Attems, J., Thomas, A. et al. (2015). Clinicians’
Prospective study of relations between corti- ability to diagnose dementia with Lewy bodies is not
cal Lewy bodies, poor eyesight, and hallucina- affected by β-amyloid load. Neurology, 84: 1–4.
tions in Alzheimer’s disease. Journal of Neurology, Tiraboschi, P., Salmon, D.P., Hansen, L.A. et al. (2006).
Neurosurgery, and Psychiatry, 59: 185–188. What best differentiates Lewy body from Alzheimer’s
Mosimann, U.P., Collerton, D., Dudley, R. et al. (2008). A disease in early-stage dementia? Brain: A Journal of
semi-structured interview to assess visual hallucina- Neurology, 129: 729–735.
tions in older people. International Journal of Geriatric Vann Jones, S.A. and O’Brien, J.T. (2014). The preva-
Psychiatry, 23 (7): 712–718. lence and incidence of dementia with Lewy bodies: A
Mosimann, U.P., Rowan, E.N., Partington, C.E. et al. (2006). systematic review of population and clinical studies.
Characteristics of visual hallucinations in Parkinson Psychological Medicine, 44(4): 673–683.
disease dementia and dementia with Lewy bod- Walker, M.P., Ayre, G.A., Cummings, J.L. et al. (2000a).
ies. American Journal of Geriatric Psychiatry, 14 (2): Quantifying fluctuation in dementia with Lewy bod-
153–160. ies, Alzheimer’s disease, and vascular dementia.
Mullan, E., Cooney, C. and Jones, E. (1996). Mania and Neurology, 54: 1616–1624.
cortical Lewy body dementia. International Journal of Walker, M.P., Ayre, G.A., Cummings, J.L. et al. (2000b).
Geriatric Psychiatry, 11: 837–839. The Clinician Assessment of Fluctuation and the One
O’Brien, J.T., Colloby, S.J., Fenwick, J. et al. (2004). Day Fluctuation Assessment Scale. Two methods
Dopamine transporter loss visualised with FP-CIT to assess fluctuating confusion in dementia. British
SPECT in Dementia with Lewy bodies. Archives of Journal of Psychiatry, 177: 252–256.
Neurology, 61 (6): 919–925. Walker, Z., Allen, R.L., Shergill, S. and Katona, C. (1996).
O’Brien, J.T., McKeith, I.G., Walker, Z. et al. (2009). Neuropsychological performance in Lewy body
Diagnostic accuracy of 123I-FP-CIT SPECT in pos- dementia and Alzheimer’s disease. British Journal of
sible dementia with Lewy bodies. British Journal of Psychiatry, 170: 156–158.
Psychiatry, 194 (1): 34–39. Weiner, M.F., Risser. R.C., Cullum, C.M. et al. (1996).
Okazaki, H., Lipton, L.S. and Aronson, S.M. (1961). Diffuse Alzheimer’s disease and its Lewy body variant: A
intracytoplasmic ganglionic inclusions (Lewy type) clinical analysis of postmortem verified cases. The
associated with progressive dementia and quadrapesis American Journal of Psychiatry, 153 (10): 1269–1273.
in flexion. Journal of Neurology, Neurosurgery, and Weisman, D., Cho, M., Taylor, C. et al. (2007). In dementia
Psychiatry, 20: 237–244. with Lewy bodies, Braak stage determines phenotype,
Perry, E.K., McKeith, I., Thompson, P. et al. (1991). not Lewy body distribution. Neurology, 69 (4): 356–359.
Topography, extent, and clinical relevance of neu- Zaccai, J., McCracken, C. and Brayne, C. (2005). A sys-
rochemical deficits in dementia of Lewy body type, tematic review of prevalence and incidence studies of
Parkinson's disease and Alzheimer’s disease. Annals of dementia with Lewy bodies. Age and Ageing, 34 (6):
the New York Academy of Sciences, 640: 197–202. 561–566.
62
Pathology of dementia with Lewy bodies

GLENDA HALLIDAY, SIMON LEWIS AND PAUL INCE

deposition of α-synuclein has also been identified in DLB


62.1 INTRODUCTION (Kramer and Schulz-Schaeffer, 2007). While this has not been
independently replicated, it is likely that synaptic changes in
Dementia with Lewy bodies (DLB) is a primary degenera- α-synuclein occur in DLB, and many new techniques are see-
tive dementia sharing features of Parkinson’s disease (PD) ing dot-like inclusions in LB disorders consistent with syn-
and most often also of Alzheimer’s disease (AD). The diag- aptic aggregations, although not as extensively (Kovacs et al.,
nosis of DLB implies an underlying pathology, the Lewy 2014; Roberts et al., 2015). In addition to neuronal inclu-
body (LB), and all patients with DLB have these inclusions. sions and aggregates (LBs and Lewy neurites), astrocytes also
LBs are intraneuronal insoluble fibrillar aggregations of the aggregate non-fibrillar α-synuclein in brain regions contain-
synaptic protein α-synuclein (Figure 62.1). Causal mutations ing LBs (Braak et al., 2007; Song et al., 2009; Kovacs et al.,
in the gene encoding α-synuclein associate with familial PD 2014) (Figure 62.1). Astrocytes are specialized glial cells that
(Polymeropoulos et al., 1997), the other major LB disease. outnumber neurones by over fivefold and are the main sup-
LBs were originally described by Friedrich Lewy over a cen- port cells of the brain, modulating neuronal activity and form-
tury ago (Lewy, 1913) but the major constituent protein was ing the blood–brain barrier (Sofroniew and Vinters, 2010).
only identified as α-synuclein in 1997 (Spillantini et al., 1997). The abnormal increase in α-synuclein in astrocytes appears
Lewy described these neuronal inclusions as intracytoplas- to compromise their function. A recent study has also high-
mic, spherical or elongated, eosinophilic masses possessing lighted oligodendroglial α-synuclein pathologies in brain-
a dense core and a peripheral halo, with neurones able to stem regions in DLB (Seidel et al., 2015). Oligodendroglia
contain multiple inclusions (Figure 62.1). Patients with a pri- wraps all neuronal membranes and myelinate axons.
mary degenerative dementia and these inclusions have DLB. Oligodendroglial α-synuclein inclusions have been observed
As described in the preceding chapter, they often also have in PD cases with longer disease durations and more marked
clinical features that differentiate them from other demen- neuronal loss (Wakabayashi et al., 2000) and in the DLB cases
tia syndromes. While the definition of DLB in itself seems with similar inclusions (Terada et al., 2000), marked to severe
simple, very few patients with DLB have only LB pathology, neuronal degeneration in restricted brainstem nuclei was
and the variety of other pathologies, neurochemical abnor- observed (Seidel et al., 2015). It may be that oligodendroglial
malities and patterns of neuronal loss continue to be assessed α-synuclein inclusions are a sign of advanced LB disease.
and defined, with multiple phenotypes now acknowledged.
LBs develop in many grey matter regions, and LBs are
62.2 CURRENT DIAGNOSTIC CRITERIA
not the only α-synuclein pathology observed. The majority
of patients with DLB have extensive LBs in limbic and less
FOR/AND THE PATHOLOGY OF
so in association cortical regions (Mckeith et al., 2005). LBs DLB
are predominantly found in deeper cortical layers (4, 5 and
6) within small- and medium-sized neurones (Kosaka et al., The DLB Consortium pathological guidelines have evolved
1984). In addition to classical LBs, there is an extensive neu- over more than a decade and require the presence of LBs in a
ritic component and diffuse cytoplasmic granular staining primary dementia syndrome (Mckeith et al., 2005) and were
(Duda et al., 2002). In fact, neuritic α-synucleinopathy is originally based on the description of LB stages identified
now considered the most consistent feature of LB diseases in DLB patients by Kosaka et al. (1988) who identified three
(Klucken et al., 2003; Duda, 2004). Considerable synaptic types (brainstem, transitional and diffuse). This DLB staging

714
Pathology of dementia with Lewy bodies 715

(a) (b) (d) (e)

50 μm 50 μm 20 μm

(c) (f )

50 μm 20 μm

100 μm 50 μm

Figure 62.1 (See colour insert.) Lewy bodies and Lewy pathologies in Parkinson’s disease. (a) and (b) Haematoxylin and
eosin stained classic Lewy bodies in pigmented dopaminergic neurones of the substantia nigra showing the typical core
and halo structure. Scale in (b) is equivalent for (a). (c)–(f). α-Synuclein immunoreactive pathologies showing (c) and (e)
mature Lewy bodies, (c) and (d) pale bodies and neuronal aggregates and (f) astrocytes containing aggregates.

scheme is virtually identical to that subsequently described of LB pathologies (Alafuzoff et al., 2009). Note that current
by Braak et al. (2003) for PD, but it differs from a more recent probability criteria for the pathological diagnosis of AD have
staging scheme suggested for LB disorders by Beach et al. recently changed to formally incorporate β-amyloid immu-
(2009). In 2005, the DLB pathological scheme changed from nohistochemistry (Montine et al., 2012). The pathological cri-
a staging scheme to give a likelihood ratio that the pathologi- teria of DLB now need to be formally updated in line with the
cal findings were associated with a clinical DLB syndrome, AD criteria and to determine which standardized protocol
taking into account both the extent of LB pathologies and should be formally adopted.
AD-related pathologies, as AD pathologies are very com- As indicated by the diagnostic criteria, apart from LBs,
mon in DLB (Mckeith et al., 2005). The practical implemen- AD-related pathologies are common in DLB. While the
tation of the criteria requires LB pathologies to be identified degree of neocortical plaque formation is equivalent to that
with immunohistochemistry and semiquantitatively graded found in AD, the presence of neocortical neurofibrillary
(Table 62.1), and National Institute on Aging (NIA)–Reagan tangles is variable and may be infrequent (Table 62.2), with
diagnostic criteria for AD-related pathologies to be applied this variability contributing to variable dementia pheno-
(National Institute on Aging and Reagan Institute Working types (Weisman et al., 2007; Tiraboschi et al., 2015). If neu-
Group, 1997). The probability of pathological DLB is then rofibrillary tangles and LB pathologies are both present, the
identified using this information with the higher LB staging likelihood of clinical expression of DLB is greater at lower
in the presence of less AD pathology more likely to be clini- Braak tangle stages (Ballard et al., 2004; Fujishiro et al., 2008;
cally significant (Table 62.2). While the pathological criteria Tiraboschi et al., 2015). The current diagnostic criteria for
have been validated in small samples (Fujishiro et al., 2008), AD with its focus on biological markers of β-amyloid deposi-
in large dementia populations a number of cases do not fit tion subsume DLB as a variant of AD (Dubois et al., 2014).
the LB staging scheme (Fujimi et al., 2008; Leverenz et al., However, the pathological distinctions between AD and
2008; Zaccai et al., 2008) and modifications have been sug- DLB are clear and include:
gested, including the incorporation of an amygdala only stage ●● There are rare cases of DLB with no significant

(Uchikado et al., 2006; Leverenz et al., 2008; Zaccai et al., AD-related pathologies.
2008). All modifications suggest fewer regions are required ●● Quantitative and qualitative analysis of the AD-related

for the assessment of LB pathologies without compromising pathologies shows differences between DLB and AD.
accuracy (Harding and Halliday, 1998; Leverenz et al., 2008; ●● While ß-amyloid deposition is similar in both AD and

Alafuzoff et al., 2009) with the latest modification showing DLB, they can be distinguished by their neuronal inclu-
better inter-observer agreement without scoring the severity sion pathology.
716 Dementia

Table 62.1 Methods for the pathological diagnosis of DLB


Assignment of Lewy body type based upon pattern of Lewy-related pathology in brainstem, limbic and neocortical
regions

Brainstem regions Basal forebrain/limbic regions Neocortical regions


IX–X LC SN nbM Amygdala Transentorhinal Cingulate Temporal Frontal Parietal
Lewy body Brainstem 1–3 1–3 1–3 0–2 0–2 0–1 0–1 0 0 0
type predominant
Limbic 1–3 1–3 1–3 2–3 2–3 1–3 1–3 0–2 0–1 0
(transitional)
Diffuse neocortical 1–3 1–3 1–3 2–3 3–4 2–4 2–4 2–3 1–3 0–2
Notes: Brain regions are as defined anatomically in the original consensus report.
Abbreviations: DLB, dementia with Lewy bodies; IX, ninth cranial nerve nucleus; X, tenth cranial nerve nucleus; LC, locus ceruleus; SN,
substantia nigra; nbM, nucleus basalis of Meynert.

Table 62.2 Methods for the pathological diagnosis of DLB


Assessment of the likelihood that the pathologic findings are associated with a DLB clinical syndrome

Alzheimer type pathology


NIA–Reagan NIA–Reagan NIA–Reagan
Low Intermediate High
(Braak stage 0–II) (Braak stage III–IV) (Braak stage V–VI)
Lewy body type pathology Brainstem-predominant Low Low Low
Limbic (transitional) High Intermediate Low
Diffuse neocortical High High Intermediate
Abbreviation: NIA, National Institute on Aging.

DLB also differs in the severity of vascular pathology et al., 2013). The TDP-43 pathology in DLB is most associ-
compared with AD. Amyloid angiopathy is prevalent in ated with coexisting AD pathology.
both AD and DLB (Jellinger and Attems, 2008a, 2008b),
but haemorrhages appear more common in DLB due to
trauma associated with falls, while microinfarcts are less
62.3 PREVALENCE OF LB IN
common (Isojima et al., 2006; Jellinger and Attems, 2008b).
White matter vascular changes are more frequent in AD and
AGEING POPULATIONS AND
small mini-bleeds occur in both AD and DLB patients with PATHOLOGICAL PHENOTYPES
AD (De Reuck et al., 2011). The extent of LB pathology is
inversely correlated to the extent of most vascular patholo- Data from large cohorts of older brain donors, including
gies (atherosclerosis, infarcts and small vessel disease) except population-based studies have identified LBs in asymptom-
for cerebral amyloid angiopathy, which correlates with the atic people and also in different distributions (Parkkinen
LB pathology (Ghebremedhin et al., 2010). et al., 2003; Zaccai et al., 2008, 2015). The UK popula-
It has been recognized for some time that tar DNA bind- tion-based study showed some degree of synucleinopathy
ing protein 43 (TDP-43) proteinopathy forms the basis for in 37% of 207 elderly people (Zaccai et al., 2008, 2015).
many cases of frontotemporal dementia (Neumann et al., Similarly, a Japanese population-based study reported
2006), and it has been reported that TDP-43 proteinopathy α-synucleinopathy with a prevalence of 22.5% at autopsy,
can present with clinical features mimicking DLB (Claassen which included 41% who were demented during life
et al., 2008). In AD, TDP-43 deposition is commonly present (Wakisaka et al., 2003). These findings are comparable to
as a minor component of the pathology, especially in hip- work from Finland (Parkkinen et al., 2003) and also from
pocampus and amygdala (Wilson et al., 2011; Youmans and the United States (Beach et al., 2009). The different distri-
Wolozin, 2012). Between 30% and 60% of DLB cases with AD butions of LBs in these populations have led to an alternate
have similar TDP-43 pathology (Nakashima-Yasuda et al., theory of progression through olfactory and limbic brain
2007; Arai et al., 2009) with a subset of TDP-43 inclusions regions in addition to that from the brainstem through to
colocalizing with LB (Higashi et al., 2007). Approximately the cortex (Beach et al., 2009) and have also lead to the con-
5% have associated hippocampal sclerosis (Aoki et al., cept of different phenotypes of DLB (Table 62.3).
2015). Few cases with pure DLB have TDP-43 pathology As detailed in Table 62.3, three main DLB phenotypes
(Nakashima-Yasuda et al., 2007; Yokota et al., 2010; Wilson can be identified based on the amount of LB and AD
Pathology of dementia with Lewy bodies 717

Table 62.3 Phenotypes of cases with different pathological combinations of LB and AD pathologies

Feature Pure DLB DLB + AD AD + amygdala LB Pure AD


Severity of amyloid β deposition High High High High
Severity of tau deposition (Weisman et al., 2007) Low High High High
Severity of LRP (Uchikado et al., 2006) Highest Intermediate Low Absent
Frequency of ApoE-4 and odds ratio ~30% ~40%–50% NA ~35%
(Tsuang et al., 2005, 2013) 6.1 12.6 NA 9.9
Frequency of GBA mutation (Clark et al., 2009; 10%–28% ~5% NA 1%–10%
Tsuang et al., 2012) 7.6 4.6 NA 1.1
Odds for male gender (Nelson et al., 2010) 2.9 2.9 NA 0.66
Odds for depression (Lopez et al., 2006) NA 3.83 8.56 0.96
Memory function (Kraybill et al., 2005; Initially Poor Poor Poor
Nelson et al., 2009) preserved
Executive function/attention (Kraybill et al., Poor Poor NA Initially preserved
2005)
Functional decline (Kraybill et al., 2005) Steady Rapid NA Steady
Median survival and survival after dementia NA 78 and 7.3 years, NA 85 and 8.5 years,
(Williams et al., 2006) adjusted for adjusted for
gender and gender and
ApoE ApoE
Abbreviations: LB, Lewy body; AD, Alzheimer’s disease; LRP, low-density lipoprotein receptor-related protein; GBA, β-glucocerebrosidase;
NA, not available; ApoE, apolipoprotein E.

pathologies. At present, the prevalence of these different on neuroimaging, which highlight underlying patholo-
phenotypes is not clear, as the amygdala-only LB pheno- gies (Mckeith et al., 2005). Positron emission tomography
type appears to be very prevalent – 60% of cases with AD (PET) and single-photon emission computed tomography
(Hamilton, 2000) – but has not been assessed at a popula- (SPECT) imaging using dopamine transporter ligands
tion level. The pure DLB phenotype is considered the least were recommended due to reduced striatal uptake in DLB
prevalent and the DLB + AD phenotype has previously patients compared with AD (Mak et al., 2014). This suggests
been known as the ‘common form’ of DLB or the ‘LB vari- that there is a loss of dopaminergic neurones in the substan-
ant of Alzheimer’s disease’ (Hansen et al., 1990). Pure DLB tia nigra and their projections to the striatum in DLB that
has more preserved memory at onset, the most severe LB is similar to that observed in PD and different from AD. A
pathology and less tau deposition, as well as the lowest fre- multicentre study confirmed the routine clinical utility of
quency of apolipoprotein E (ApoE) ε4 genotype and the using dopamine transporter ligands to assist with the diag-
highest frequency of β-glucocerebrosidase (GBA) mutations nosis of DLB showing over 85% accuracy in diagnosing DLB
(Table 62.3). Compared with AD, DLB + AD has LB pathol- with high inter-rater agreement (Mckeith et al., 2007). A
ogy, a higher frequency of both ApoE ε4 genotype and GBA longitudinal study shows that such scans detect abnormali-
mutations as well as depression, with a more rapid func- ties in the dopaminergic system prior to symptom onset
tional decline (Table 62.3). The amygdala-only phenotype (Siepel et al., 2013). Of importance, in patients followed lon-
has the highest rate of depression (Table 62.3). While these gitudinally to autopsy, sensitivity was 88% and specificity
phenotypes are separable pathologically with evidence of was 100% in diagnosing DLB using dopamine transporter
clinical and genetic differences, more work is needed to dif- imaging, proving that such imaging substantially enhances
ferentiate these phenotypes further, particularly clinically. the diagnosis of DLB (Walker et al., 2007). Although it is
suggested that a proportion of DLB cases do not have dopa-
mine transporter abnormalities (Siepel et al., 2013), these
62.4 NEUROIMAGING CHANGES AND studies have not been confirmed at autopsy.
At autopsy, the loss of neurones in the substantia nigra is
NEURONAL LOSS IN AUTOPSY
generally not as severe while more LB are often seen com-
CONFIRMED DLB pared to PD, although some DLB cases do show substantia
nigra neuronal loss and LB formation in the PD range (Iseki
62.4.1 DOPAMINE TRANSPORTER AND et al., 2005; Tsuboi and Dickson, 2005). This potentially
NEUROMELANIN IMAGING AND suggests that synaptic damage is greater than the absolute
RELATED CELL LOSS loss of neurones in DLB compared with PD. Neuronal loss
and increasing LB severity are related (Oinas et al., 2009),
In addition to revising the pathological definition of DLB also suggesting that more LBs, as seen in DLB, may protect
in 2005, the DLB Consortium also revised guidelines neuronal cell bodies but not terminals. Importantly, the loss
718 Dementia

of the dopaminergic pigmented neurones of the substantia abnormalities, which differentiate DLB from AD (Nedelska
nigra can be visualized using magnetic resonance imaging et al., 2015b). These data again suggest that neuronal cell
(MRI) (Kitao et al., 2013) providing another tool for future bodies are spared in DLB but that significant abnormalities
studies to assess differences between the loss of synapses occur to their processes.
with dopamine transporter labelling and neuronal cell bod- In addition to the more selective hypometabolism of
ies with MRI. the occipital cortex in DLB, a new signature abnormality
on glucose metabolism is called the cingulate island sign,
62.4.2 MEDIAL TEMPORAL LOBE where there is a higher ratio of posterior cingulate to pre-
ATROPHY AND RELATED CELL cuneus and cuneus metabolism (Graff-Radford et al., 2014).
LOSS The regions of lower metabolism have been shown to have
cortical thinning in DLB (Delli Pizzi et al., 2014a). DLB
Medial temporal lobe atrophy has been identified as a cases with this metabolic signature have a high or interme-
highly accurate diagnostic marker for autopsy confirmed diate pathologic likelihood of DLB using the 2005 consensus
AD (sensitivity 91%, specificity 94%), and does not relate to criteria and lower neuritic pathologic stages (Graff-Radford
the severity of plaque or LBs in the temporal lobe (Burton et al., 2014), although changes in these regions of the brain
et al., 2009). Patients with a high or intermediate pathologic have yet to be assessed pathologically. The cingulate island
likelihood of DLB using the 2005 consensus criteria have sign is independent of positive findings on amyloid PiB
normal hippocampal volumes (Kantarci et al., 2012) and imaging (Graff-Radford et al., 2014).
greater atrophy rates in the hippocampus and amygdala cor-
relate with AD-type pathology in DLB patients (Nedelska
et al., 2015a). These studies show that medial temporal lobe 62.4.4 OTHER REGIONS OF NEURONAL
changes are much more likely to relate to AD pathologies in LOSS IDENTIFIED
DLB than to LB pathologies.
In patients with pure DLB, neuronal loss in the amygdala Unfortunately, many studies on other regions of neuronal
is severe (Yamamoto et al., 2006) and there is selective loss loss, particularly those assessing subcortical regions, have not
of neurones in only the lower pyramidal layers of the presu- assessed appropriate comparison groups, and so it is difficult
biculum in DLB compared with PD or AD (Harding et al., to determine whether any changes noted are more related to
2002b). In the hippocampus proper, there is a reduction in AD-type pathologies or LB pathologies, and whether patients
the small interneuronal population that contains parvalbu- with PD and no dementia also have similar neuronal loss.
min immunoreactivity (Bernstein et al., 2011) suggesting Below is a summary of regions where multiple studies have
some changes in calcium regulation. These changes appear assessed the same region using different methods.
to be additional to any changes associated with concomitant Greater structural changes in the thalamus is observed
AD in patients with DLB; however, it should be noted that in DLB compared with AD (Delli Pizzi et al., 2014b) and
the AD changes are overwhelming compared to the DLB thalamic pathology is substantial in DLB, particularly in
changes noted. the midline (innervate medial temporal and prefrontal cor-
tices) and anterior (innervate frontal and parietal cortices)
62.4.3 CORTICAL ATROPHY AND intralaminar nuclei (Brooks and Halliday, 2009). These
RELATED CELL LOSS thalamic regions regulate cortical synchrony, informa-
tion transmission and cognition (Saalmann, 2014). Recent
Overall cortical atrophy is more pronounced in DLB com- studies have identified neuronal loss in the hypothalamic
pared with PD, but AD has more temporal and frontal lobe sleep-­associated centres (Kasanuki et al., 2014; Benarroch
atrophy than DLB (Beyer et al., 2007). Glucose PET imag- et al., 2015), but such loss does not appear to differ between
ing shows hypometabolism in temporoparietal regions and patients with DLB and AD (Kasanuki et al., 2014).
occipital regions in DLB with occipital hypometabolism dif- Severe loss of cholinergic neurones in the basal fore-
ferentiating DLB (Kasanuki et al., 2012; Toledo et al., 2013; brain that innervate the medial temporal lobe is observed
Mak et al., 2014). Neuronal loss in temporoparietal regions in patients with DLB compared with AD (Fujishiro et al.,
is milder in DLB compared with AD, with no difference 2006), and there is greater atrophy of the entire cholinergic
in the degree of cell loss in these regions in DLB patients basal forebrain region in DLB (Kim et al., 2011), although this
with or without AD pathology (Kasanuki et al., 2012). This may occur in a region-specific fashion (Grothe et al., 2014).
suggests that the severity of AD pathology is independent Patients with a high pathologic likelihood of DLB using the
of neuronal loss in DLB, with confirmation that amyloid 2005 consensus criteria have atrophy of the cholinergic dorsal
Pittsburgh compound B (PiB) imaging detects all types of mesopontine grey matter nuclei (Kantarci et al., 2012) and a
amyloid plaques in DLB (Kantarci et al., 2012). Amyloid PiB loss of the main cholinergic projection from the dorsal meso-
imaging in DLB is not related to the severity of hypome- pontine region to the thalamus has been identified in DLB
tabolism (Ishii et al., 2015) confirming this notion. No cell but not AD using acetylcholinesterase (AChE) PET (Kotagal
loss is observed in occipital cortex (Kasanuki et al., 2012), et al., 2012b). Neuronal loss has been observed in this region
but diffusion tensor imaging shows significant white matter in some (Schmeichel et al., 2008), but not all (Hepp et al.,
Pathology of dementia with Lewy bodies 719

2013), cases with DLB. Of interest, amyloid deposition is only contrast to the increase in subcortical D2 receptors, there is
observed in this region in DLB (Hepp et al., 2013). a substantial decrease in cortical D2 receptors (Piggott et al.,
2007b), most likely due to the denervation of the cortex by
62.4.5 SYNAPTIC LOSS dopaminergic neurones. The severity of cortical D2 recep-
tor loss correlates with the severity of cognitive decline and
Synaptic loss has been proposed to be a major process in psychiatric symptoms, as well as with increasing LB pathol-
the pathogenesis of DLB linked to the role of oligomeric ogy, and is likely to explain the intolerance of DLB patients
α-synuclein acting as a synaptotoxin (Kramer and Schulz- to D2 antagonistic neuroleptic medications (Piggott et al.,
Schaeffer, 2007). This finding has been replicated using other 2007b). Importantly, such changes are not observed in pure
synaptic markers in the medial temporal lobe (Mukaetova- AD cases (Piggott et al., 2007b).
Ladinska et al., 2009), occipital (Mukaetova-Ladinska et al.,
2013) and frontal lobes (Whitfield et al., 2014), suggesting 62.5.2 CHOLINERGIC SYSTEMS
that LB pathology does primarily affect synapses more than
neuronal cell bodies. As discussed above, there is a loss of the major cholinergic
neurones innervating the cortex and subcortical structures
in DLB, with subcortical denervation and more marked
hippocampal denervation differentiating DLB from AD.
62.5 NEUROTRANSMITTERS AND
There are two major classes of cholinergic receptors, nico-
NEUROCHEMICAL CHANGES IN
tinic receptors located on postsynaptic neurones, and mus-
DLB carinic receptors located on neurones and blood vessels
(Greig et al., 2013). In addition to the same type of receptor
As discussed above, neuronal loss specific to DLB is more changes as discussed above, enzymatic changes that regu-
similar to PD and involves the cholinergic and dopami- late acetylcholine metabolism can also impact on choliner-
nergic systems, but is also overlaid with changes associated gic function, with certain drugs increasing cholinergic tone
with AD, at least in a proportion of cases. In addition to the targeting this aspect selectively (Greig et al., 2013).
frank loss of neurones, neurotransmitter receptor changes Cortical nicotinic receptors are fast-acting choliner-
in postsynaptic neurones also occur, as detailed below. Such gic receptors that are reduced equally in DLB, AD and
changes have relevance for neurotransmitter replacement PD (Perry et al., 1990b), a dysfunction that correlates
therapies. with a decline in executive function (Colloby et al., 2010).
However, in DLB there is an increase in nicotinic recep-
62.5.1 DOPAMINERGIC SYSTEMS tors in the occipital lobe, a change most pronounced in
patients with recent visual hallucinations, linking cholin-
As discussed above, the loss of dopaminergic neurones in ergic changes in the occipital lobe to visual hallucinations
the substantia nigra and their input to the striatum is highly (O’Brien et al., 2008). Such a receptor change suggests most
accurate at identifying patients with DLB from AD. As the marked cholinergic denervation in the occipital lobe in DLB
membranes of these neurones contain the dopamine trans- compared to the other cortical regions. In contrast to nico-
porter, neuroimaging using this ligand is a very effective tinic receptors, muscarinic receptors affect neurones over
way of seeing this change, as discussed above. There are a longer time frame. In DLB and PD, muscarinic receptors
multiple dopamine receptors (D1–D5) that can be classed are elevated in the cortex (Teaktong et al., 2005), even in the
into two major categories, D1-like receptors (D1 and D5) occipital lobe (Colloby et al., 2006), in contrast to AD (Perry
that are very widespread in the brain, and D2-like recep- et al., 1990b), again reflecting possible upregulation of post-
tors (D2, D3 and D4) that autoregulate dopaminergic synaptic receptors in response to cholinergic denervation.
neurones as well as occurring selectively postsynaptically The increase in muscarinic receptors in the cingulate cortex
(Beaulieu and Gainetdinov, 2011). Receptor loss occurs relates to the severity of psychosis (Teaktong et al., 2005).
when the neuronal structures containing them degener- In contrast, muscarinic receptor density in the striatum is
ate, as discussed, but can also occur with drug treatments. lower in DLB and parallels low D2 receptor density (Piggott
Agonists that activate receptors can decrease receptor den- et al., 2003), suggesting that these receptors may be located
sity, whereas antagonists that block receptor activity or a on the same structures that have degenerated, which is the
reduction in neurotransmitter can increase receptor den- dopamine terminals, as described above.
sity. In this context, it is of interest that D1 receptors do not Neocortical choline acetyltransferase (ChAT) activ-
change substantially in DLB, even in regions of significant ity, the enzyme necessary for acetylcholine production, is
dopamine denervation (Sun et al., 2013). However, D2-like lower in DLB than in AD, and similar to that observed in
receptors are upregulated in the striatum and thalamus in demented PD (Mukaetova-Ladinska et al., 2013). This data
DLB (Piggott et al., 2007a; Sun et al., 2013), particularly in is consistent with the structural loss of cholinergic neu-
those treated with neuroleptic medications, which antago- rones, as discussed above. The other enzyme that has been
nize dopamine receptors, with such changes associated with studied in detail is AChE, the main enzyme that degrades
an increase in extrapyramidalism (Piggott et al., 2007a). In acetylcholine. Cortical activity of AChE is significantly
720 Dementia

reduced in probable DLB compared with probable AD the risk of dementia (Zaccai et al., 2015). Other molecu-
(Shimada et al., 2009; Klein et al., 2010; Shimada et al., lar markers have been associated with cognitive decline,
2015), although this is not as consistent early in these dis- including synaptic markers (Whitfield et al., 2014) and
eases where there is a more consistent deficit in patients dynamin1 (Vallortigara et al., 2014).
with amnestic cognitive impairment (Marcone et al.,
2012). In patients with probable DLB, the greatest differen- 62.6.2 PARKINSONISM
tial reduction is observed in the posterior cingulate cortex
(Shimada et al., 2015). This suggests there is less degrada- The pathological and neurochemical basis of mild parkin-
tion and increased acetylcholine transmitter cortically as sonism in DLB are the most satisfactorily characterized
DLB takes hold, particularly in the posterior cingulate aspect of DLB pathology. The presence of nigral dopamine
cortex, which may be the basis of the cingulate island sign neurone loss with reduced presynaptic dopamine trans-
discussed above. Perhaps surprisingly, reducing AChE porter activity in the striatum now form the basis for a
activity further with the use of inhibitors produces sig- clinical diagnostic strategy to distinguish DLB from other
nificant clinical benefit in DLB (Mori et al., 2012), suggest- dementia syndromes, as discussed above.
ing that decreasing acetylcholine degradation further and
enhancing cholinergic transmission in general is benefi- 62.6.3 VISUAL HALLUCINATIONS
cial. Of interest, glucose metabolism has been reported to
be either increased (Fong et al., 2011) or decreased in DLB The presence of visual hallucinations is common to both
following AChE inhibition (Satoh et al., 2010), which may DLB and PD with dementia. However, it is well recognized
perhaps relate to diagnostic overlap with AD (where cho- clinically that visual hallucinations and other psychotic fea-
linesterase inhibitors increase perfusion [Chaudhary et al., tures such as paranoia, emerge as early as in the first year of
2013]), as none of these studies have had pathologic confir- DLB but not generally seen for several years in the course
mation of diagnosis. of PD (Hely et al., 2008). One of the obvious candidate
regions expected to correlate with visual hallucinations in
DLB would be the components of the visual pathways. There
has been some speculation on the contribution of pathology
62.6 MOLECULAR AND ANATOMICAL in the retina to hallucinations in DLB, but recent studies
PATHOLOGY CORRELATES OF show a lack of either AD or LB pathologies in the retina of
CLINICAL FEATURES IN DLB patients with AD or LB diseases (Ho et al., 2014). However,
DLB patients appear to have greater atrophy of the retinal
Research has attempted to identify key brain structures nerve fibre layer compared with AD or PD (Moreno-Ramos
where pathology underpins the major and minor clinical et al., 2013) and large pale synaptic inclusions between the
features of DLB. Parkinsonism is securely attributable to photoreceptors and retinal horizontal cells (Maurage et al.,
degeneration of the nigrostriatal dopaminergic pathway. 2003; Devos et al., 2005), consistent again with abnormali-
Neurochemical and anatomical substrates for other clini- ties in axonal and synaptic structures in DLB. The pale
cal hallmarks such as visual hallucinations, fluctuating synaptic inclusions in DLB occur only in patients with hal-
consciousness and rapid eye movement (REM) behavioural lucinations, suggesting a specific and less common associ-
sleep disorder (RBD) remain poorly characterized. ated retinopathy (Devos et al., 2005). Visual information
from the retina is relayed to the cortex via the thalamus
with the main rely nucleus being the lateral geniculate.
62.6.1 COGNITIVE DECLINE IN DLB There are no pathological features in the lateral geniculate
relay to the occipital cortex in DLB, in contrast to AD where
While mild cognitive impairment is recognized in around some degeneration is observed (Yamamoto et al., 2006;
50% of PD patients at their time of diagnosis (Foltynie et al., Erskine et al., 2016). However, microstructural changes
2004), the progression to dementia is generally seen over (Delli Pizzi et al., 2014b) and α-synuclein pathologies
5–20 years depending on their age of disease onset (Hely (Yamamoto et al., 2006; Brooks and Halliday, 2009) occur
et al., 2008). In contrast, significant cognitive decline from in both the posterior thalamic regions projecting between
onset is the clinical hallmark of DLB. Attributing specific the occipital and parietal cortices as well as in the medial
molecular and pathological changes as the substrate of thalamic regions projecting to the amygdala, differentiat-
cognitive decline in DLB is complicated by the frequent ing DLB patients from AD. Hallucinations particularly
coexistence of AD-type pathologies. However, some recent related to microstructural changes in the posterior tha-
studies show that the degree of cognitive decline correlates lamic regions (Delli Pizzi et al., 2014b), while unconscious
positively with the severity of cortical α-synuclein pathol- visual information is transferred from the thalamus to the
ogy, but also to the severity of β-amyloid and tau deposition amygdala, and α-synuclein pathology and microstructural
in DLB (Howlett et al., 2015). This differs from the regional changes in the amygdala in conjunction with the loss of
pattern of α-synuclein pathology, which is independent of dopaminergic neurones is also associated with the presence
Pathology of dementia with Lewy bodies 721

of visual hallucinations (Harding et al., 2002a; Yamamoto networks) and visual networks of the brain, allowing for
et al., 2006; Kalaitzakis et al., 2009a; Kantarci et al., 2010; the generation of incorrect perceptions (Shine et al., 2014a).
Zaccai et al., 2015). A requirement for visual hallucinations This construct is consistent with the brain changes identi-
is a relative reduction in the activity/perfusion of the occipi- fied above and allows an appreciation that the generation
tal cortex (Shimada et al., 2009; Firbank et al., 2015) and of hallucinations can arise from pathology across multiple
there is some evidence of a reduction in vascular endothe- regions with contributions from various neurotransmitters
lial growth factor and capillary density in the occipital lobe (Onofrj et al., 2013; Shine et al., 2014b).
in DLB compared with AD (Miners et al., 2014). Of note,
primary visual cortex in the occipital lobe is spared from 62.6.4 FLUCTUATION IN CONSCIOUS
α-synuclein pathologies and abnormalities, and is there- LEVEL
fore not used diagnostically (Table 62.1), although some LB
pathology may be seen in secondary visual cortices in DLB The neuroanatomical structures that are key to regulation
patients (Yamamoto et al., 2006). These secondary visual of consciousness are not well characterized and candidate
regions have been shown to be dysfunctional with thinned brain regions have not been established in DLB. Through
cortices in DLB patients with visual hallucinations (Taylor serendipitous experiments, the claustrum has recently been
et al., 2012; Delli Pizzi et al., 2014a). These data show only identified as the central hub of a network that subserves
subtle changes in primary visual pathways in DLB patients consciousness (Koubeissi et al., 2014) and as described
with hallucinations, although more severe LB pathology above this region has significant LB pathology in DLB
and dysfunction in secondary visual relay pathways as well (Yamamoto et al., 2006; Kalaitzakis et al., 2009a, 2009b).
as in pathways relaying visual information to limbic brain The claustrum connects to many parts of the cortex, and to
regions associates with visual hallucinations. the hippocampus, amygdala and caudate nucleus, allowing
A number of other regional brain changes are docu- it to have widespread coordination of the cerebral cortex.
mented as associated with visual hallucinations in DLB. During disruptions of consciousness by stimulation of the
In patients with visual hallucination there is an increased claustrum, there is increased electroencephalogram (EEG)
burden of α-synucleinopathy in the parahippocampal signal synchrony in medial parietal and posterior frontal
cortices, claustrum and insula (Harding et al., 2002a; networks (Koubeissi et al., 2014). Increased EEG signal syn-
Yamamoto et al., 2006; Kalaitzakis et al., 2009a, 2009b), chrony in these regions distinguishes DLB from AD, PD
with the amount of α-synuclein pathology in these regions and elderly controls with many studies demonstrating its
correlating with that observed in secondary visual cortices utility in clinical practice (Kai et al., 2005; Andersson et al.,
(Yamamoto et al., 2006). The insula has been identified as 2008; Bonanni et al., 2008; Roks et al., 2008; Liedorp et al.,
a core region of degeneration/atrophy using neuroimaging 2009; Micanovic and Pal, 2014; Lee et al., 2015). A recent
in patients with visual hallucinations and associates with study suggests that EEG signal synchrony measures are
hypometabolism in the anterior cingulate and parahippo- 100% accurate in predicting conversion from mild cognitive
campal cortices (Blanc et al., 2014; Heitz et al., 2015). In DLB impairment to DLB within 3 years (Bonanni et al., 2015). It
patients with complex hallucinations, hypometabolism in is of interest that in a small study, AChE inhibitors reduced
parahippocampal/inferior temporal cortices is especially the EEG synchrony in DLB but not AD patients (Kai et al.,
pronounced (Heitz et al., 2015). Decreased cortical cholin- 2005). Pathologically, there are no differences in the sever-
ergic activity appears to be important in DLB patients with ity of cortical cholinergic degeneration or LB pathologies in
hallucinations (Perry et al., 1990a), with AChE inhibitors DLB patients with or without fluctuations in consciousness
that enhance cholinergic transmission and posterior cor- (Ballard et al., 2002; Harding et al., 2002a), but a reduc-
tical perfusion highly effective against this clinical feature tion in temporal lobe nicotinic receptors has been demon-
(Satoh et al., 2010; Ukai et al., 2015). strated (Ballard et al., 2002). Such a reduction is unlikely to
More recent developments in our understanding of explain the efficacy of AChE inhibitors, and the relationship
visual hallucinations have come from an appreciation of between EEG abnormalities and neurochemical changes
the role of higher order neural networks rather than think- and pathology in the claustrum in DLB should be pursued
ing of more simplistic neural circuits or neurotransmit- further.
ters (Onofrj et al., 2013). Much of this thinking has arisen
from functional neuroimaging studies that have identified 62.6.5 RAPID EYE MOVEMENT SLEEP
functional networks that co-activate in response to specific BEHAVIOUR DISORDER
internal and exogenous demands (Smith et al., 2010; Laird
et al., 2011; Fornito and Harrison, 2012) and also in the In the 2005 consensus on DLB diagnosis (McKeith et al.,
absence of an external task (Biswal et al., 1995). Evidence 2005), an additional suggestive clinical feature for DLB was
from functional MRI experiments in non-demented PD introduced – RBD, where patients experience dream enact-
patients with and without visual hallucinations has sug- ment. The prevalence of RBD in DLB patients has not been
gested that, there is a breakdown in the communication determined, but its inclusion in the diagnosis of DLB is help-
between the attentional (Default Mode, Dorsal and Ventral ful in association with other core clinical features (Ferman
722 Dementia

et al., 2011), indicating it is not more highly prevalent than neighbouring cells for degradation in lysosomes (see Lee
these features. In longitudinal studies, RBD appears to be et al., 2014 for review). Under stress conditions, α-synuclein
highly specific for an underlying α-synuclein pathology (not is mainly released from neurones via unconventional exocy-
necessarily DLB) (Iranzo et al., 2013; Schenck et al., 2013), tosis in vesicles in an oligomeric form, but is also observed in
although analysis of large cohorts of patients with dementia exosomes (although inhibition of their formation increases
shows that RBD more frequently occurs in a variety of other α-synuclein secretion) (see Lee et al., 2014 for review).
dementia types (AD, frontotemporal dementia and vascu- Experimental studies have shown that preformed α-synuclein
lar dementia) compared with DLB (Pistacchi et al., 2014). fibrils and oligomers can be internalized by cultured neu-
Further studies are required to determine the prevalence of rones via endocytosis and that direct transfer of α-synuclein
RBD in patients with DLB and other types of dementia. This between cells in culture and in animal models occurs (see
will assist in identifying the underlying brain regions and Lee et al., 2014 for review). In cell culture, the α-synuclein
pathologies associated with RBD. In this context, RBD con- transferred from donor cells to recipient cells can form inclu-
verters to DLB have occipital hypometabolism (Fujishiro sions consistent with LB pathologies, and animal models
et al., 2013) and awake EEG signal synchrony (Inoue et al., show that α-synuclein pathology can spread throughout the
2015) similar to that for DLB described above, and DLB brain (Masuda-Suzukake et al., 2013). These studies suggest
patients with RBD have more severe occipital hypoperfusion that lysosome dysfunction is important for LB formation and
(Chiba et al., 2014). Cross-sectionally, patients with RBD are that once LB form, they can spread throughout the brain.
similar to DLB patients in their dopamine cell loss and stri-
atal denervation (Rupprecht et al., 2013) and their choliner-
gic denervation (Kotagal et al., 2012a) with similar severities
of brainstem LB pathology (Dugger et al., 2012) but more 62.8 SUMMARY
cortical LB pathology (Murray et al., 2013; Postuma et al.,
2015). Cross-sectional neuroimaging shows that patients LB formation is much more highly prevalent in dementia
with RBD have greater neuronal degeneration in the locus patients with AD-type pathology than previously recog-
caeruleus that correlates with increased muscle tone dur- nized, although it is clear that DLB has specific and identifi-
ing REM sleep (Garcia-Lorenzo et al., 2013), although by able pathologies that warrant its recognition as the distinct
end-stage, degeneration in this region or in the cholinergic disorder. Compared with AD, the loss of dopaminergic
pedunculopontine tegmental nucleus is similar in patients neurones is profound while the medial temporal lobe is less
with or without RBD (Dugger et al., 2012). This suggests affected. Pathological comparisons with end-stage PD show
that noradrenergic degeneration is not isolated to RBD but largely similar degenerative changes that differ in their
is more severe at earlier disease stages. The specificity of any tempo and order of onset. This review has identified synap-
of these or other pathologies for the clinical phenomenon of tic and axonal pathologies as particularly prominent in DLB
RBD rather than another core feature associated with such compared with both AD and PD, and genetic associations
pathologies requires significantly more work. suggest that dysfunction of lysosomes plays an important
role. Further research with more consistent comparisons
between patients with DLB, AD and PD are required to
unravel the separate and overlapping factors critical for
62.7 PATHOGENESIS OF DLB AND these different disorders.
OTHER SYNUCLEINOPATHIES
REFERENCES
Most research on LB formation is driven by mechanisms
associated with PD, although genetic association studies Alafuzoff, I., Ince, P.G., Arzberger, T. et al. (2009). Staging/
show similar associations to α-synuclein and genes associ- typing of Lewy body related alpha-synuclein pathol-
ated with lysosomal dysfunction (Bras et al., 2014). In addi- ogy: A study of the BrainNet Europe Consortium. Acta
tion to depositing α-synuclein, impairment in lysosomal Neuropathologica, 117: 635–652.
pathways is a characteristic of most cells that aggregate Andersson, M., Hansson, O., Minthon, L. et al. (2008).
α-synuclein (Tofaris, 2012; Dehay et al., 2013). Lysosomes Electroencephalogram variability in dementia with lewy
degrade proteins and other cellular molecules via three bodies, Alzheimer’s disease and controls. Dementia
pathways, microautophagy (small invagination of the mem- and Geriatric Cognitive Disorders, 26: 284–290.
brane), chaperone-mediated autophagy (CMA, selectively Aoki, N., Murray, M.E., Ogaki, K. et al. (2015).
tagged molecules for lysosomal degradation) and macro- Hippocampal sclerosis in Lewy body disease is a TDP-
autophagy (walling off of large cellular compartments, like 43 proteinopathy similar to FTLD-TDP Type A. Acta
mitochondria, with specialized membranes that then fuse Neuropathologica, 129: 53–64.
with lysosomes). There is strong evidence for selective CMA Arai, T., Mackenzie, I.R., Hasegawa, M. et al. (2009).
dysfunction associated with increased α-synuclein accumu- Phosphorylated TDP-43 in Alzheimer’s disease and
lation (see Bourdenx et al., 2014 for review). α-Synuclein is dementia with Lewy bodies. Acta Neuropathologica,
normally released from neurones and can be taken up by 117: 125–136.
Pathology of dementia with Lewy bodies 723

Ballard, C.G., Court, J.A., Piggott, M. et al. (2002). Bras, J., Guerreiro, R., Darwent, L. et al. (2014). Genetic
Disturbances of consciousness in dementia with Lewy analysis implicates APOE, SNCA and suggests lyso-
bodies associated with alteration in nicotinic receptor somal dysfunction in the etiology of dementia with Lewy
binding in the temporal cortex. Consciousness and bodies. Human Molecular Genetics, 23: 6139–6146.
Cognition, 11: 461–474. Brooks, D. and Halliday, G.M. (2009). Intralaminar nuclei
Ballard, C.G., Jacoby, R., Del Ser, T. et al. (2004). of the thalamus in Lewy body diseases. Brain Research
Neuropathological substrates of psychiatric symp- Bulletin, 78: 97–104.
toms in prospectively studied patients with autopsy- Burton, E.J., Barber, R., Mukaetova-Ladinska, E.B. et al.
confirmed dementia with lewy bodies. The American (2009). Medial temporal lobe atrophy on MRI differ-
Journal of Psychiatry, 161: 843–849. entiates Alzheimer’s disease from dementia with Lewy
Beach, T.G., Adler, C.H., Lue, L. et al. (2009). Unified bodies and vascular cognitive impairment: A prospec-
staging system for Lewy body disorders: Correlation tive study with pathological verification of diagnosis.
with nigrostriatal degeneration, cognitive impairment Brain: A Journal of Neurology, 132: 195–203.
and motor dysfunction. Acta Neuropathologica, 117: Chaudhary, S., Scouten, A., Schwindt, G. et al. (2013).
613–634. Hemodynamic effects of cholinesterase inhibition
Beaulieu, J.M. and Gainetdinov, R.R. (2011). The physiol- in mild Alzheimer’s disease. Journal of Magnetic
ogy, signaling, and pharmacology of dopamine recep- Resonance Imaging, 38: 26–35.
tors. Pharmacological Reviews, 63: 182–217. Chiba, Y., Iseki, E., Fujishiro, H. et al. (2014). Primary visual
Benarroch, E.E., Schmeichel, A.M., Parisi, J.E. and Low, P.A. cortical metabolism and rapid eye movement sleep
(2015). Histaminergic tuberomammillary neuron loss behavior disorder in dementia with Lewy bodies.
in multiple system atrophy and dementia with Lewy Psychiatry and Clinical Neurosciences, 68: 137–144.
bodies. Movement Disorders: Official Journal of the Claassen, D.O., Parisi, J.E., Giannini, C. et al. (2008).
Movement Disorder Society, 30: 1133–1139. Frontotemporal dementia mimicking dementia with
Bernstein, H.G., Johnson, M., Perry, R.H. et al. (2011). Lewy bodies. Cognitive and Behavioral Neurology, 21:
Partial loss of parvalbumin-containing hippocam- 157–163.
pal interneurons in dementia with Lewy bodies. Clark, L.N., Kartsaklis, L.A., Wolf Gilbert, R. et al. (2009).
Neuropathology, 31: 1–10. Association of glucocerebrosidase mutations with
Beyer, M.K., Larsen, J.P. and Aarsland, D. (2007). Gray dementia with lewy bodies. Archives of Neurology, 66:
matter atrophy in Parkinson disease with dementia and 578–583.
dementia with Lewy bodies. Neurology, 69: 747–754. Colloby, S.J., Pakrasi, S., Firbank, M.J. et al. (2006). In vivo
Biswal, B., Yetkin, F.Z., Haughton, V.M. and Hyde, J.S. SPECT imaging of muscarinic acetylcholine receptors
(1995). Functional connectivity in the motor cortex of using (R,R) 123I-QNB in dementia with Lewy bodies and
resting human brain using echo-planar MRI. Magnetic Parkinson’s disease dementia. NeuroImage, 33: 423–429.
Resonance in Medicine, 34: 537–541. Colloby, S.J., Perry, E.K., Pakrasi, S. et al. (2010). Nicotinic
Blanc, F., Noblet, V., Philippi, N. et al. (2014). Right ante- 123I-5IA-85380 single photon emission computed
rior insula: Core region of hallucinations in cognitive tomography as a predictor of cognitive progression in
neurodegenerative diseases. PLoS ONE, 9: e114774. Alzheimer’s disease and dementia with Lewy bodies.
Bonanni, L., Perfetti, B., Bifolchetti, S. et al. (2015). The American Journal of Geriatric Psychiatry: Official
Quantitative electroencephalogram utility in predicting Journal of the American Association for Geriatric
conversion of mild cognitive impairment to dementia Psychiatry, 18: 86–90.
with Lewy bodies. Neurobiology of Aging, 36: 434–445. De Reuck, J., Deramecourt, V., Cordonnier, C. et al.
Bonanni, L., Thomas, A., Tiraboschi, P. et al. (2008). EEG (2011). Prevalence of small cerebral bleeds in
comparisons in early Alzheimer’s disease, dementia patients with a neurodegenerative dementia:
with Lewy bodies and Parkinson’s disease with demen- A ­neuropathological study. Journal of the Neurological
tia patients with a 2-year follow-up. Brain: A Journal of Sciences, 300: 63–66.
Neurology, 131: 690–705. Dehay, B., Martinez-Vicente, M., Caldwell, G.A. et al.
Bourdenx, M., Bezard, E. and Dehay, B. (2014). Lysosomes (2013). Lysosomal impairment in Parkinson’s disease.
and alpha-synuclein form a dangerous duet leading to Movement Disorders: Official Journal of the Movement
neuronal cell death. Frontiers in Neuroanatomy, 8: 83. Disorder Society, 28: 725–732.
Braak, H., Del Tredici, K., Rüb, U. et al. (2003). Staging of Delli Pizzi, S., Franciotti, R., Tartaro, A. et al. (2014a).
brain pathology related to sporadic Parkinson’s dis- Structural alteration of the dorsal visual network in
ease. Neurobiology of Aging, 24: 197–211. DLB patients with visual hallucinations: A cortical thick-
Braak, H., Sastre, M. and Del Tredici, K. (2007). ness MRI study. PLoS ONE, 9: e86624.
Development of alpha-synuclein immunoreactive Delli Pizzi, S., Maruotti, V., Taylor, J.P. et al. (2014b).
astrocytes in the forebrain parallels stages of intraneu- Relevance of subcortical visual pathways disruption
ronal pathology in sporadic Parkinson’s disease. Acta to visual symptoms in dementia with Lewy bodies.
Neuropathologica, 114: 231–241. Cortex, 59: 12–21.
724 Dementia

Devos, D., Tir, M., Maurage, C.A. et al. (2005). ERG and Fujishiro, H., Umegaki, H., Isojima, D. et al. (2006).
anatomical abnormalities suggesting retinopathy in Depletion of cholinergic neurons in the nucleus
dementia with Lewy bodies. Neurology, 65: 1107–1110. of the medial septum and the vertical limb of the
Dubois, B., Feldman, H.H., Jacova, C. et al. (2014). diagonal band in dementia with Lewy bodies. Acta
Advancing research diagnostic criteria for Alzheimer’s Neuropathologica, 111: 109–114.
disease: The IWG-2 criteria. The Lancet Neurology, 13: Garcia-Lorenzo, D., Longo-Dos Santos, C., Ewenczyk, C.
614–629. et al. (2013). The coeruleus/subcoeruleus complex
Duda, J.E. (2004). Pathology and neurotransmitter abnor- in rapid eye movement sleep behaviour disorders in
malities of dementia with Lewy bodies. Dementia and Parkinson’s disease. Brain: A Journal of Neurology, 136:
Geriatric Cognitive Disorders, 17 (Suppl. 1): S3–S14. 2120–2129.
Duda, J.E., Giasson, B.I., Mabon, M.E. et al. (2002). Novel Ghebremedhin, E., Rosenberger, A., Rub, U. et al.
antibodies to synuclein show abundant striatal pathol- (2010). Inverse relationship between cerebrovas-
ogy in Lewy body diseases. Annals of Neurology, 52: cular lesions and severity of lewy body pathology
205–210. in patients with Lewy body diseases. Journal of
Dugger, B.N., Murray, M.E., Boeve, B.F. et al. (2012). Neuropathology and Experimental Neurology, 69:
Neuropathological analysis of brainstem cholinergic and 442–448.
catecholaminergic nuclei in relation to rapid eye move- Graff-Radford, J., Murray, M.E., Lowe, V.J. et al. (2014).
ment (REM) sleep behaviour disorder. Neuropathology Dementia with Lewy bodies: Basis of cingulate island
and Applied Neurobiology, 38: 142–152. sign. Neurology, 83: 801–809.
Erskine, D., Taylor, J.P., Firbank, M.J. et al. (2016). Changes Greig, N.H., Reale, M. and Tata, A.M. (2013). New phar-
to the lateral geniculate nucleus in Alzheimer’s disease macological approaches to the cholinergic system: An
but not dementia with Lewy bodies. Neuropathology overview on muscarinic receptor ligands and cho-
and Applied Neurobiology, 42: 366–376. linesterase inhibitors. Recent Patents on CNS Drug
Ferman, T.J., Boeve, B.F., Smith, G.E. et al. (2011). Inclusion Discovery, 8: 123–141.
of RBD improves the diagnostic classification of demen- Grothe, M.J., Schuster, C., Bauer, F. et al. (2014). Atrophy
tia with Lewy bodies. Neurology, 77: 875–882. of the cholinergic basal forebrain in dementia with
Firbank, M.J., Lloyd, J. and O’Brien, J.T. (2015). The Lewy bodies and Alzheimer’s disease dementia.
relationship between hallucinations and FDG-PET Journal of Neurology, 261: 1939–1948.
in dementia with Lewy bodies. Brain Imaging and Hamilton, R.L. (2000). Lewy bodies in Alzheimer’s disease:
Behavior: 1–4. A neuropathological review of 145 cases using alpha-
Foltynie, T., Brayne, C.E., Robbins, T.W. and Barker, R.A. synuclein immunohistochemistry. Brain Pathology, 10:
(2004). The cognitive ability of an incident cohort of 378–384.
Parkinson’s patients in the UK. The CamPaIGN study. Hansen, L., Salmon, D., Galasko, D. et al. (1990). The Lewy
Brain: A Journal of Neurology, 127: 550–560. body variant of Alzheimer’s disease: A clinical and
Fong, T.G., Inouye, S.K., Dai, W. et al. (2011). Association pathologic entity. Neurology, 40: 1–8.
cortex hypoperfusion in mild dementia with Lewy bod- Harding, A.J., Broe, G.A. and Halliday, G.M. (2002a).
ies: A potential indicator of cholinergic dysfunction? Visual hallucinations in Lewy body disease relate to
Brain Imaging and Behavior, 5: 25–35. Lewy bodies in the temporal lobe. Brain: A Journal of
Fornito, A. and Harrison, B.J. (2012). Brain connectivity Neurology, 125: 391–403.
and mental illness. Frontiers in Psychiatry, 3: 72. Harding, A. and Halliday, G. (1998). Simplified neuro-
Fujimi, K., Sasaki, K., Noda, K. et al. (2008). pathological diagnosis of dementia with Lewy bod-
Clinicopathological outline of dementia with Lewy ies. Neuropathology and Applied Neurobiology, 24:
bodies applying the revised criteria: The Hisayama 195–201.
study. Brain Pathology, 18: 317–325. Harding, A.J., Lakay, B. and Halliday, G.M. (2002b).
Fujishiro, H., Ferman, T.J., Boeve, B.F. et al. (2008). Selective hippocampal neuron loss in dementia with
Validation of the neuropathologic criteria of the third Lewy bodies. Annals of Neurology, 51: 125–128.
consortium for dementia with Lewy bodies for prospec- Heitz, C., Noblet, V., Cretin, B. et al. (2015). Neural cor-
tively diagnosed cases. Journal of Neuropathology and relates of visual hallucinations in dementia with Lewy
Experimental Neurology, 67: 649–656. bodies. Alzheimer’s Research and Therapy, 7: 6.
Fujishiro, H., Iseki, E., Kasanuki, K. et al. (2013). A follow Hely, M.A., Reid, W.G., Adena, M.A. et al. (2008). The
up study of non-demented patients with primary visual Sydney multicenter study of Parkinson’s disease:
cortical hypometabolism: Prodromal dementia with The inevitability of dementia at 20 years. Movement
Lewy bodies. Journal of the Neurological Sciences, Disorders: Official Journal of the Movement Disorder
334: 48–54. Society, 23: 837–844.
Pathology of dementia with Lewy bodies 725

Hepp, D.H., Ruiter, A.M., Galis, Y. et al. (2013). Kalaitzakis, M.E., Pearce, R.K. and Gentleman, S.M.
Pedunculopontine cholinergic cell loss in hallucinating (2009b). Clinical correlates of pathology in the claus-
Parkinson disease patients but not in dementia with trum in Parkinson’s disease and dementia with Lewy
Lewy bodies patients. Journal of Neuropathology and bodies. Neuroscience Letters, 461: 12–15.
Experimental Neurology, 72: 1162–1170. Kantarci, K., Avula, R., Senjem, M.L. et al. (2010).
Higashi, S., Iseki, E., Yamamoto, R. et al. (2007). Dementia with Lewy bodies and Alzheimer disease:
Concurrence of TDP-43, tau and alpha-synuclein Neurodegenerative patterns characterized by DTI.
pathology in brains of Alzheimer’s disease and Neurology, 74: 1814–1821.
dementia with Lewy bodies. Brain Research, 1184: Kantarci, K., Yang, C., Schneider, J.A. et al. (2012).
284–294. Antemortem amyloid imaging and beta-amyloid
Ho, C.Y., Troncoso, J.C., Knox, D. et al. (2014). Beta- pathology in a case with dementia with Lewy bodies.
amyloid, phospho-tau and alpha-synuclein deposits Neurobiology of Aging, 33: 878–885.
similar to those in the brain are not identified in the Kasanuki, K., Iseki, E., Fujishiro, H. et al. (2012).
eyes of Alzheimer’s and Parkinson’s disease patients. Neuropathological investigation of the hypometabolic
Brain Pathology, 24: 25–32. regions on positron emission tomography with [18F]
Howlett, D.R., Whitfield, D., Johnson, M. et al. (2015). fluorodeoxyglucose in patients with dementia with
Regional multiple pathology scores are associated Lewy bodies. Journal of the Neurological Science, 314:
with cognitive decline in Lewy body dementias. Brain 111–119.
Pathology, 25: 401–408. Kasanuki, K., Iseki, E., Kondo, D. et al. (2014).
Inoue, Y., Sasai, T. and Hirata, K. (2015). Neuropathological investigation of hypocretin
Electroencephalographic finding in idiopathic REM sleep expression in brains of dementia with Lewy bodies.
behavior disorder. Neuropsychobiology, 71: 25–33. Neuroscience Letters, 569: 68–73.
Iranzo, A., Tolosa, E., Gelpi, E. et al. (2013). Kim, H.J., Lee, J.E., Shin, S.J. et al. (2011). Analysis of
Neurodegenerative disease status and post-mortem the substantia innominata volume in patients with
pathology in idiopathic rapid-eye-movement sleep Parkinson’s disease with dementia, dementia with lewy
behaviour disorder: An observational cohort study. bodies, and Alzheimer’s disease. Journal of Movement
The Lancet Neurology, 12: 443–453. Disorders, 4: 68–72.
Iseki, E., Marui, W., Yamamoto, R. et al. (2005). The Kitao, S., Matsusue, E., Fujii, S. et al. (2013). Correlation
nigro-striatal and nigro-amygdaloid pathways between pathology and neuromelanin MR imaging in
undergo different degeneration processes in brains Parkinson’s disease and dementia with Lewy bodies.
of dementia with Lewy bodies. Neuroscience Letters, Neuroradiology, 55: 947–953.
380: 161–165. Klein, J.C., Eggers, C., Kalbe, E. et al. (2010).
Ishii, K., Hosokawa, C., Hyodo, T. et al. (2015). Regional Neurotransmitter changes in dementia with Lewy bod-
glucose metabolic reduction in dementia with Lewy ies and Parkinson disease dementia in vivo. Neurology,
bodies is independent of amyloid deposition. Annals 74: 885–892.
of Nuclear Medicine, 29: 78–83. Klucken, J., Mclean, P.J., Gomez-Tortosa, E. et al.
Isojima, D., Togo, T., Kosaka, K. et al. (2006). Vascular com- (2003). Neuritic alterations and neural system
plications in dementia with Lewy bodies: A postmor- dysfunction in Alzheimer’s disease and dementia
tem study. Neuropathology, 26: 293–297. with Lewy bodies. Neurochemical Research, 28:
Jellinger, K.A. and Attems, J. (2008a). Cerebral amyloid 1683–1691.
angiopathy in Lewy body disease. Journal of Neural Kosaka, K., Tsuchiya, K. and Yoshimura, M. (1988).
Transmission, 115: 473–482. Lewy body disease with and without dementia:
Jellinger, K.A. and Attems, J. (2008b). Prevalence and A clinicopathological study of 35 cases. Clinical
impact of vascular and Alzheimer pathologies in Neuropathology, 7: 299–305.
Lewy body disease. Acta Neuropathologica, 115: Kosaka, K., Yoshimura, M., Ikeda, K. and Al, E. (1984).
427–436. Diffuse type of Lewy body disease: Progressive
Kai, T., Asai, Y., Sakuma, K. et al. (2005). Quantitative dementia with abundant cortical Lewy bodies and
electroencephalogram analysis in dementia with senile changes of varying degree – A new disease?
Lewy bodies and Alzheimer’s disease. Journal of the Clinical Neuropathology, 3: 185–192.
Neurological Science, 237: 89–95. Kotagal,V., Albin, R.L., Muller, M.L. et al. (2012a).
Kalaitzakis, M.E., Christian, L.M., Moran, L.B. et al. Symptoms of rapid eye movement sleep behavior
(2009a). Dementia and visual hallucinations associ- disorder are associated with cholinergic denerva-
ated with limbic pathology in Parkinson’s disease. tion in Parkinson disease. Annals of Neurology, 71:
Parkinsonism and Related Disorders, 15: 196–204. 560–568.
726 Dementia

Kotagal, V., Muller, M.L., Kaufer, D.I. et al. (2012b). disease, and dementia with Lewy bodies: A Bayesian
Thalamic cholinergic innervation is spared in Alzheimer method and voxel-based analysis. Journal of
disease compared to parkinsonian disorders. Alzheimer’s Disease, 31: 387–399.
Neuroscience Letters, 514: 169–172. Masuda-Suzukake, M., Nonaka, T., Hosokawa, M. et al.
Koubeissi, M.Z., Bartolomei, F., Beltagy, A. and Picard, (2013). Prion-like spreading of pathological alpha-synu-
F. (2014). Electrical stimulation of a small brain area clein in brain. Brain, 136: 1128–1138.
reversibly disrupts consciousness. Epilepsy and Maurage, C.A., Ruchoux, M.M., De Vos, R. et al. (2003).
Behavior, 37: 32–35. Retinal involvement in dementia with Lewy bodies:
Kovacs, G.G., Breydo, L., Green, R. et al. (2014). A clue to hallucinations? Annals of Neurology, 54:
Intracellular processing of disease-associated alpha- 542–547.
synuclein in the human brain suggests prion-like cell- Mckeith, I.G., Dickson, D.W., Lowe, J. et al. (2005).
to-cell spread. Neurobiology of Disease, 69: 76–92. Diagnosis and management of dementia with
Kramer, M.L. and Schulz-Schaeffer, W.J. (2007). Lewy bodies: Third report of the DLB Consortium.
Presynaptic alpha-synuclein aggregates, not Lewy Neurology, 65: 1863–1872.
bodies, cause neurodegeneration in dementia with Mckeith, I., O’Brien, J., Walker, Z. et al. (2007). Sensitivity
Lewy bodies. The Journal of Neuroscience, 27: and specificity of dopamine transporter imaging with
1405–1410. 123I-FP-CIT SPECT in dementia with Lewy bodies: A
Kraybill, M.L., Larson, E.B., Tsuang, D.W. et al. (2005). phase III, multicentre study. The Lancet Neurology, 6:
Cognitive differences in dementia patients with 305–313.
autopsy-verified AD, Lewy body pathology, or both. Micanovic, C. and Pal, S. (2014). The diagnostic utility of
Neurology, 64: 2069–2073. EEG in early-onset dementia: A systematic review of
Laird, A.R., Fox, P.M., Eickhoff, S.B. et al. (2011). the literature with narrative analysis. Journal of Neural
Behavioral interpretations of intrinsic connectivity Transmission, 121: 59–69.
networks. Journal of Cognitive Neuroscience, 23: Miners, S., Moulding, H., De Silva, R. and Love, S.
4022–4037. (2014). Reduced vascular endothelial growth fac-
Lee, H.J., Bae, E.J. and Lee, S.J. (2014). Extracellular tor and ­capillary density in the occipital cortex in
alpha-synuclein – A novel and crucial factor in Lewy ­dementia with Lewy bodies. Brain Pathology, 24:
body diseases. Nature Reviews Neurology, 10: 92–98. 334–343.
Lee, H., Brekelmans, G.J. and Roks, G. (2015). The EEG as Montine, T.J., Phelps, C.H., Beach, T.G. et al. (2012).
a diagnostic tool in distinguishing between dementia National Institute on Aging-Alzheimer’s Association
with Lewy bodies and Alzheimer’s disease. Clinical guidelines for the neuropathologic assessment of
Neurophysiology: Official Journal of the International Alzheimer’s disease: A practical approach. Acta
Federation of Clinical Neurophysiology , 126: Neuropathologica, 123: 1–11.
1735–1739. Moreno-Ramos, T., Benito-Leon, J., Villarejo, A.
Leverenz, J.B., Hamilton, R., Tsuang, D.W. et al. (2008). and Bermejo-Pareja, F. (2013). Retinal nerve
Empiric refinement of the pathologic assessment of fiber layer thinning in dementia associated with
Lewy-related pathology in the dementia patient. Brain Parkinson’s disease, dementia with Lewy bodies,
Pathology, 18: 220–224. and Alzheimer’s disease. Journal of Alzheimer’s
Lewy, F.H. (1913). Zur pathologischen anatomie Disease, 34: 659–664.
der Paralysis Agitans. Deutsche Zeitschrift für Mori, E., Ikeda, M., Kosaka, K. and Donepezil DLB Study
Nervenheilkunde, 50: 50–55. Investigators. (2012). Donepezil for dementia with
Liedorp, M., Van Der Flier, W.M., Hoogervorst, E.L. Lewy bodies: A randomized, placebo-controlled trial.
et al. (2009). Associations between patterns of EEG Annals of Neurology, 72: 41–52.
abnormalities and diagnosis in a large memory clinic Mukaetova-Ladinska, E.B., Andras, A., Milne, J. et al.
cohort. Dementia and Geriatric Cognitive Disorders, (2013). Synaptic proteins and choline acetyltrans-
27: 18–23. ferase loss in visual cortex in dementia with Lewy
Lopez, O.L., Becker, J.T., Sweet, R.A. et al. (2006). Lewy bodies. Journal of Neuropathology and Experimental
bodies in the amygdala increase risk for major depres- Neurology, 72: 53–60.
sion in subjects with Alzheimer disease. Neurology, 67: Mukaetova-Ladinska, E.B., Xuereb, J.H., Garcia-Sierra, F.
660–665. et al. (2009). Lewy body variant of Alzheimer’s disease:
Mak, E., Su, L., Williams, G.B. and O’Brien, J.T. Selective neocortical loss of t-SNARE proteins and loss
(2014). Neuroimaging characteristics of demen- of MAP2 and alpha-synuclein in medial temporal lobe.
tia with Lewy bodies. Alzheimer’s Research and TheScientificWorldJournal, 9: 1463–1475.
Therapy, 6: 18. Murray, M.E., Ferman, T.J., Boeve, B.F. et al. (2013).
Marcone, A., Garibotto, V., Moresco, R.M. et al. (2012). MRI and pathology of REM sleep behavior disor-
[11C]-MP4A PET cholinergic measurements in amnes- der in dementia with Lewy bodies. Neurology, 81:
tic mild cognitive impairment, probable Alzheimer’s 1681–1689.
Pathology of dementia with Lewy bodies 727

Nakashima-Yasuda, H., Uryu, K., Robinson, J. et al. (2007). Piggott, M.A., Ballard, C.G., Dickinson, H.O. et al.
Co-morbidity of TDP-43 proteinopathy in Lewy body (2007a). Thalamic D2 receptors in dementia with
related diseases. Acta Neuropathologica, 114: 221–229. Lewy bodies, Parkinson’s disease, and Parkinson’s
National Institute on Aging and Reagan Institute disease dementia. The International Journal of
Working Group on Diagnostic Criteria for the Neuropsychopharmacology, 10: 231–244.
Neuropathological Assessment of Alzheimer’s Piggott, M.A., Ballard, C.G., Rowan, E. et al. (2007b).
Disease. (1997). Consensus recommendations Selective loss of dopamine D2 receptors in temporal
for the postmortem diagnosis of Alzheimer’s dis- cortex in dementia with Lewy bodies, association with
ease. The National Institute on Aging and Reagan cognitive decline. Synapse, 61: 903–911.
Institute Working Group on Diagnostic Criteria for Piggott, M.A., Owens, J., O’Brien, J… et al. (2003).
the Neuropathological Assessment of Alzheimer’s Muscarinic receptors in basal ganglia in demen-
Disease. Neurobiology of Aging, 18: S1–S2. tia with Lewy bodies, Parkinson’s disease
Nedelska, Z., Ferman, T.J., Boeve, B.F. et al. (2015a). and Alzheimer’s disease. Journal of Chemical
Pattern of brain atrophy rates in autopsy-confirmed Neuroanatomy, 25: 161–173.
dementia with Lewy bodies. Neurobiology of Aging, Pistacchi, M., Gioulis, M., Contin, F. et al. (2014). Sleep dis-
36: 452–461. turbance and cognitive disorder: Epidemiological analy-
Nedelska, Z., Schwarz, C.G., Boeve, B.F. et al. (2015b). sis in a cohort of 263 patients. Neurological Sciences:
White matter integrity in dementia with Lewy bodies: Official Journal of the Italian Neurological Society and
A voxel-based analysis of diffusion tensor imaging. of the Italian Society of Clinical Neurophysiology, 35:
Neurobiology of Aging, 36: 2010–2017. 1955–1962.
Nelson, P.T., Kryscio, R.J., Jicha, G.A. et al. (2009). Polymeropoulos, M.H., Lavedan, C., Leroy, E. et al.
Relative preservation of MMSE scores in autopsy- (1997). Mutation in the alpha-synuclein gene identi-
proven dementia with Lewy bodies. Neurology, 73: fied in familes with Parkinson’s disease. Science, 276:
1127–1133. 2045–2047.
Nelson, P.T., Schmitt, F.A., Jicha, G.A. et al. (2010). Postuma, R.B., Adler, C.H., Dugger, B.N. et al. (2015).
Association between male gender and cortical Lewy REM sleep behavior disorder and neuropathology
body pathology in large autopsy series. Journal of in Parkinson’s disease. Movement Disorders: Official
Neurology, 257: 1875–1881. Journal of the Movement Disorder Society, 30 (10):
Neumann, M., Sampathu, D.M., Kwong, L.K. et al. (2006). 1413–1417.
Ubiquitinated TDP-43 in frontotemporal lobar degen- Roberts, R.F., Wade-Martins, R. and Alegre-Abarrategui,
eration and amyotrophic lateral sclerosis. Science, 314: J. (2015). Direct visualization of alpha-synuclein
130–133. oligomers reveals previously undetected pathol-
O’Brien, J.T., Colloby, S.J., Pakrasi, S. et al. (2008). ogy in Parkinson’s disease brain. Brain: A Journal of
Nicotinic alpha4beta2 receptor binding in dementia Neurology, 138: 1642–1657.
with Lewy bodies using 123I-5IA-85380 SPECT demon- Roks, G., Korf, E.S., Van Der Flier, W.M. et al. (2008). The
strates a link between occipital changes and visual use of EEG in the diagnosis of dementia with Lewy
hallucinations. NeuroImage, 40: 1056–1063. bodies. Journal of Neurology, Neurosurgery and
Oinas, M., Polvikoski, T., Sulkava, R. et al. (2009). Psychiatry, 79: 377–380.
Neuropathologic findings of dementia with lewy bod- Rupprecht, S., Walther, B., Gudziol, H. et al. (2013). Clinical
ies (DLB) in a population-based Vantaa 85+ study. markers of early nigrostriatal neurodegeneration in
Journal of Alzheimer’s Disease, 18: 677–689. idiopathic rapid eye movement sleep behavior disor-
Onofrj, M., Taylor, J.P., Monaco, D. et al. (2013). Visual hal- der. Sleep Medicine, 14: 1064–1070.
lucinations in PD and Lewy body dementias: Old and Saalmann, Y.B. (2014). Intralaminar and medial thalamic
new hypotheses. Behavioural Neurology, 27: 479–493. influence on cortical synchrony, information transmis-
Parkkinen, L., Soininen, H. and Alafuzoff, I. (2003). Regional sion and cognition. Frontiers in Systems Neuroscience,
distribution of a-synuclein pathology in unimpaired aging 8: 83.
and Alzheimer disease. Journal of Neuropathology and Satoh, M., Ishikawa, H., Meguro, K. et al. (2010).
Experimental Neurology, 62: 363–367. Improved visual hallucination by donepezil and
Perry, E.K., Marshall, E., Kerwin, J. et al. (1990a). Evidence occipital glucose metabolism in dementia with Lewy
of monoaminergic-cholinergic imbalance related to bodies: The Osaki-Tajiri project. European Neurology,
visual hallucinations in Lewy body dementia. Journal of 64: 337–344.
Neurochemistry, 55: 1456–1456. Schenck, C.H., Boeve, B.F. and Mahowald, M.W. (2013).
Perry, E.K., Smith, C.J., Court, J.A. and Perry, R.H. Delayed emergence of a parkinsonian disorder or
(1990b). Cholinergic nicotinic and muscarinic receptors dementia in 81% of older men initially diagnosed with
in dementia of Alzheimer, Parkinson and Lewy body idiopathic rapid eye movement sleep behavior disor-
types. Journal of Neural Transmission. Parkinson’s der: A 16-year update on a previously reported series.
Disease and Dementia Section, 2: 149–158. Sleep Medicine, 14: 744–748.
728 Dementia

Schmeichel, A.M., Buchhalter, L.C., Low, P.A. et al. (2008). Terada, S., Ishizu, H., Haraguchi, T. et al. (2000).
Mesopontine cholinergic neuron involvement in Tau-negative astrocytic star-like inclusions and
Lewy body dementia and multiple system atrophy. coiled bodies in dementia with Lewy bodies. Acta
Neurology, 70: 368–373. Neuropathologica, 100: 464–468.
Seidel, K., Mahlke, J., Siswanto, S. et al. (2015). The brain- Tiraboschi, P., Attems, J., Thomas, A. et al. (2015).
stem pathologies of Parkinson’s disease and dementia Clinicians’ ability to diagnose dementia with Lewy
with lewy bodies. Brain Pathology, 25: 121–135. bodies is not affected by beta-amyloid load.
Shimada, H., Hirano, S., Shinotoh, H. et al. Neurology, 84: 496–499.
(2009). Mapping of brain acetylcholinester- Tofaris, G.K. (2012). Lysosome-dependent pathways as
ase alterations in Lewy body disease by PET. a unifying theme in Parkinson’s disease. Movement
Neurology, 73: 273–278. Disorders: Official Journal of the Movement Disorder
Shimada, H., Hirano, S., Sinotoh, H. et al. (2015). Dementia Society, 27: 1364–1369.
with Lewy bodies can be well-differentiated from Toledo, J.B., Cairns, N.J., Da, X. et al. (2013). Clinical
Alzheimer’s disease by measurement of brain ace- and multimodal biomarker correlates of ADNI neu-
tylcholinesterase activity-a [ C]MP4A PET study. ropathological findings. Acta Neuropathologica
International Journal of Geriatric Psychiatry, 30 (11): Communications, 1: 65.
1105–1113. Tsuang, D., Leverenz, J.B., Lopez, O.L. et al. (2013). APOE
Shine, J.M., Halliday, G.M., Gilat, M. et al. (2014a). The epsilon4 increases risk for dementia in pure synucle-
role of dysfunctional attentional control networks in inopathies. JAMA Neurology, 70: 223–228.
visual misperceptions in Parkinson’s disease. Human Tsuang, D., Leverenz, J.B., Lopez, O.L. et al. (2012). GBA
Brain Mapping, 35: 2206–2219. mutations increase risk for Lewy body disease with
Shine, J.M., O’Callaghan, C., Halliday, G.M. and Lewis, and without Alzheimer disease pathology. Neurology,
S.J. (2014b). Tricks of the mind: Visual hallucinations as 79: 1944–1950.
disorders of attention. Progress in Neurobiology, 116: Tsuang, D.W., Wilson, R.K., Lopez, O.L. et al. (2005).
58–65. Genetic association between the APOE*4 allele and
Siepel, F.J., Rongve, A., Buter, T.C. et al. (2013). (123I) Lewy bodies in Alzheimer disease. Neurology, 64:
FP-CIT SPECT in suspected dementia with Lewy bodies: 509–513.
A longitudinal case study. BMJ Open, 3 (4): e002642. Tsuboi, Y. and Dickson, D.W. (2005). Dementia with Lewy
Smith, J.F., Pillai, A., Chen, K. and Horwitz, B. (2010). bodies and Parkinson’s disease with dementia: Are
Identification and validation of effective connectivity they different? Parkinsonism and Related Disorders, 11
networks in functional magnetic resonance imaging (Suppl. 1) S47–S51.
using switching linear dynamic systems. NeuroImage, Uchikado, H., Lin, W.L., Delucia, M.W. and Dickson, D.W.
52: 1027–1040. (2006). Alzheimer disease with amygdala Lewy bod-
Sofroniew, M.V. and Vinters, H.V. (2010). Astrocytes: ies: A distinct form of alpha-synucleinopathy. Journal
Biology and pathology. Acta Neuropathologica, 119: of Neuropathology and Experimental Neurology, 65:
7–35. 685–697.
Song, Y.J., Halliday, G.M., Holton, J.L. et al. (2009). Ukai, K., Fujishiro, H., Iritani, S. and Ozaki, N. (2015). Long-
Degeneration in different parkinsonian syndromes term efficacy of donepezil for relapse of visual hallu-
relates to astrocyte type and astrocyte protein expres- cinations in patients with dementia with Lewy bodies.
sion. Journal of Neuropathology and Experimental Psychogeriatrics,15: 133–137.
Neurology, 68: 1073–1083. Vallortigara, J., Rangarajan, S., Whitfield, D. et al. (2014).
Spillantini, M.G., Schmidt, M.L., Lee, V.M.-Y. et al. (1997). Dynamin1 concentration in the prefrontal cortex is
a-Synuclein in Lewy bodies. Nature, 388: 839–840. associated with cognitive impairment in Lewy body
Sun, J., Cairns, N.J., Perlmutter, J.S. et al. (2013). dementia. F1000Research, 3: 108.
Regulation of dopamine D(3) receptor in the striatal Wakabayashi, K., Hayashi, S., Yoshimoto, M. et al.
regions and substantia nigra in diffuse Lewy body (2000). NACP/alpha-synuclein-positive filamentous
disease. Neuroscience, 248: 112–126. inclusions in astrocytes and oligodendrocytes of
Taylor, J.P., Firbank, M.J., He, J. et al. (2012). Visual cortex Parkinson’s disease brains. Acta Neuropathologica,
in dementia with Lewy bodies: Magnetic resonance 99: 14–20.
imaging study. The British Journal of Psychiatry: The Wakisaka, Y., Furuta, A., Tanizaki, Y. et al. (2003). Age-
Journal of Mental Science, 200: 491–498. associated prevalence and risk factors of Lewy body
Teaktong, T., Piggott, M.A., Mckeith, I.G. et al. (2005). pathology in a general population: the Hisayama
Muscarinic M2 and M4 receptors in anterior cingu- study. Acta Neuropathologica, 106: 374–382.
late cortex: Relation to neuropsychiatric symptoms Walker, Z., Jaros, E., Walker, R.W. et al. (2007). Dementia
in dementia with Lewy bodies. Behavioural Brain with Lewy bodies: A comparison of clinical diag-
Research, 161: 299–305. nosis, FP-CIT single photon emission computed
Pathology of dementia with Lewy bodies 729

t­ omography imaging and autopsy. Journal of Yamamoto, R., Iseki, E., Murayama, N. et al. (2006).
Neurology, Neurosurgery and Psychiatry, 78: Investigation of Lewy pathology in the visual pathway
1176–1181. of brains of dementia with Lewy bodies. Journal of the
Weisman, D., Cho, M., Taylor, C. et al. (2007). In demen- Neurological Sciences, 246: 95–101.
tia with Lewy bodies, Braak stage determines phe- Yokota, O., Davidson, Y., Arai, T. et al. (2010). Effect of
notype, not Lewy body distribution. Neurology, 69: topographical distribution of alpha-synuclein pathol-
356–359. ogy on TDP-43 accumulation in Lewy body disease.
Whitfield, D.R., Vallortigara, J., Alghamdi, A. et al. (2014). Acta Neuropathologica, 120: 789–801.
Assessment of ZnT3 and PSD95 protein levels in Lewy Youmans, K.L. and Wolozin, B. (2012). TDP-43: A new
body dementias and Alzheimer’s disease: Association player on the AD field? Experimental Neurology, 237:
with cognitive impairment. Neurobiology of Aging, 35: 90–95.
2836–2844. Zaccai, J., Brayne, C., Matthews, F.E. and Ince, P.G.
Williams, M.M., Xiong, C., Morris, J.C. and Galvin, J.E. (2015). Alpha-synucleinopathy and neuropsychological
(2006). Survival and mortality differences between symptoms in a population-based cohort of the elderly.
dementia with Lewy bodies vs Alzheimer disease. Alzheimer’s Research and Therapy, 7(1): 19.
Neurology, 67: 1935–1941. Zaccai, J., Brayne, C., Mckeith, I. et al. (2008). Patterns
Wilson, A.C., Dugger, B.N., Dickson, D.W. and Wang, and stages of alpha-synucleinopathy: Relevance in a
D.S. (2011). TDP-43 in aging and Alzheimer’s dis- population-based cohort. Neurology, 70: 1042–1048.
ease – A review. International Journal of Clinical and
Experimental Pathology, 4: 147–155.
Wilson, R.S., Yu, L., Trojanowski, J.Q. et al. (2013). TDP-43
pathology, cognitive decline, and dementia in old age.
JAMA Neurology, 70: 1418–1424.
63
The treatment of dementia with Lewy bodies

JOHN-PAUL TAYLOR

symptoms and prevent multiple concurrent clinic/hospital


63.1 INTRODUCTION attendances, although local resources will dictate what is
pragmatically possible.
The management of dementia with Lewy bodies (DLB) is not In this chapter, we focus on DLB management. However,
easy. Clinicians face a daunting multiplicity of symptoms clinical uncertainty often exists in terms of whether the
where the treatment of one symptom can potentially exac- diagnosis of the dementia is DLB or Parkinson’s disease
erbate another. Classic examples of these include the use of with dementia (PDD; see Chapter 64). While such delin-
dopaminergic medications to help ameliorate motor symp- eation is important for research, from a clinical perspec-
toms but which conversely exacerbate or cause psychiatric tive there are significant overlaps in the treatments, which
symptoms; in a similar vein, the use of cholinesterase inhibi- are applied to both conditions and thus, management
tors (ChEIs) may improve cognition, yet lead to unacceptable suggestions described here also have broad applicability
urinary urgency and incontinence. However, this should not to PDD.
engender treatment nihilism, as often, with well-informed
and balanced management plans, patients with DLB can do
very well. Indeed, as discussed in Section 63.2.2, treatment
effects for ChEI can be noticeably greater in DLB patients 63.2 PHARMACOLOGICAL
than in patients with Alzheimer’s disease (AD). INTERVENTIONS
Practical management of DLB, reflecting the widespread
impact of the core pathology of alpha-synuclein across mul-
63.2.1 GENERAL PRINCIPLES
tiple neural systems (both peripheral and central), often
means that clinicians need to consider a wide range of con- Prior to adding in any new medications, it is worthwhile
current treatments targeting different symptom clusters reviewing the patient’s current medicines and rationalize
(Figure 63.1). these in conjunction with their prescribers, e.g. physical
The key to this process is making sure that the patient and health specialists or primary care physicians. This is par-
patient’s family have a clear understanding of the condition ticularly important in DLB as polypharmacy (particularly
and its myriad of symptoms. In the clinical consultation, with drugs that have psychotropic effects such as opiates,
asking direct questions in each of the symptom domains antihistamines, anticonvulsants, benzodiazepines etc.)
can unearth what seem like apparently unrelated yet trou- can exacerbate or mimic DLB related symptoms such as
bling problems and allow patients, families and caregivers to worsening confusion, sedation and behavioural distur-
rank these in order of priority and severity so that a rational bances, as well as contribute to risk of falls in the patient.
management plan can be chalked out for the patient. Frank In particular, be alert to those drugs with potent central
discussions on the benefits versus side effects of particular anticholinergic effects – generally, these should be avoided
interventions can also help determine what symptoms are in patients with DLB. However, one should also be aware
perhaps most important to patients and their families. that there is increasing evidence that even drugs with mild
Experienced multidisciplinary teams (e.g. geriatricians, anticholinergic effects (e.g. opiates, digoxin, beta-blockers
psychiatrists, neurologists, physiotherapists, nurse special- etc.), which are often prescribed in old age, additively, can
ists etc.) working in close collaboration probably represent have a deleterious effect on cognition, worsen long-term
the idealized service model to deal with the wide array of functional outcomes in dementia and increase the risk

730
The treatment of dementia with Lewy bodies 731

Cognitive
impairment and
cognitive
fluctuations

Autonomic
dysfunction, e.g.
Depression,
OH, urinary
anxiety, apathy
symptoms,
constipation etc.

DLB
symptoms

Psychosis, especially
Parkinsonism visual
hallucinations

Sleep
disturbances, e.g.
RBD, insomnia,
daytime
somnolence etc.

Figure 63.1 Examples of symptom clusters in dementia with Lewy bodies (DLB). OH, orthostatic hypotension; RBD, rapid
eye movement sleep behaviour disorder.

of mortality (Jewart et al., 2005; Fox et al., 2011; Kalisch effect may be an effective method by which to treat cognitive
Ellett et al., 2014; Gray et al., 2015). Furthermore, from and neuropsychiatric symptoms of DLB.
a DLB perspective, drugs with an anticholinergic effect Supporting this assertion is the observation that, in gen-
may also obviate any therapeutic benefit of any prescribed eral treatment, effect size for cognitive impairment appears to
ChEI. be larger in Lewy body dementias (LBDs) than AD (Samuel
Another challenge in DLB is the inherent variabil- et al., 2000; Emre et al., 2007). All three currently prescribed
ity in symptom severity, which can make it difficult to ChEIs, donepezil, rivastigmine and galantamine have
determine any positive or negative response to treatment. been tested in DLB with improvements noted in cognition
Pragmatically, for the initiation of new medications in (Rolinski et al., 2012; Stinton et al., 2015; Wang et al., 2015).
patients with DLB, the established mantra of starting low A recent meta-analysis (Stinton et al., 2015) across a combina-
and going slow is a sensible approach. Careful upward titra- tion of eight major trials of either rivastigmine or donepezil
tion with regular monitoring of the mental and physical in DLB and PDD suggested that, on average, the Mini-Mental
state can help as well as only introducing one treatment at a State Examination (MMSE) improved by 1.26 points (95%
time to determine response while being mindful to the fact confidence interval [CI] 0.66–1.86) with cognitive benefits
that any short-term worsening may be related to the disease favouring donepezil in DLB and rivastigmine in PDD.
itself rather than the treatment. Post-mortem studies have suggested that visual hal-
lucination occurrence is also associated with more pro-
63.2.2 CHOLINESTERASE INHIBITORS found cholinergic deficits (Perry et al., 1990) and this
aligns with clinical trial data demonstrating improvement
There is extensive evidence to suggest that unlike AD, there in this symptom in DLB with rivastigmine and donepezil
is relatively less neuronal loss but relatively more profound (McKeith et al., 2000; Emre et al., 2004; Mori et al., 2012).
cholinergic loss in DLB (Tiraboschi et al., 2002) and this Notably, the presence of visual hallucinations appears to
provides the scientific basis that restoration of cholinergic predict a better cognitive response to ChEI, perhaps, as
function by blocking acetylcholine breakdown by cholines- a result of greater cholinergic deficits in those individu-
terases in the synaptic cleft, and prolonging its postsynaptic als who experience those (McKeith et al., 2004). Other
732 Dementia

psychiatric domains that may also improve with ChEI With regard to side effects, meta-analyses have suggested
in DLB include apathy, delusions, depression and cogni- that there is perhaps a tendency for more adverse events with
tive fluctuations (McKeith et al., 2000; Mori et al., 2012). rivastigmine than donepezil (Stinton et al., 2015). However,
Behavioural symptoms such as agitation and aggression rivastigmine now comes as a transdermal patch also, which
can sometimes improve with ChEI, particularly, if there is appears to have a lower proclivity for gastrointestinal side
a clear cognitive component driving them; however, ChEI, effects and may have benefits in terms of providing a more
as alerting agents, can, on rare occasions, paradoxically sustained release (Emre et al., 2010a).
aggravate agitation in DLB patients. When and if, one should withdraw ChEI in DLB is not
Functionally, ChEI use in both DLB and PDD has been clear; certainly, if there are side effects there may be a clear
associated with improvements in activities of daily living need to do this. Sudden withdrawal of ChEI in DLB (and
(ADLs) and clinical global impression and reductions in PDD), however, has been associated with worsening neu-
caregiver burden scales (Aarsland et al., 2002; Emre et al., ropsychiatric symptoms and marked declines in cognition
2004; Dubois et al., 2012; Mori et al., 2012). Even if improve- (Minett et al., 2003) and even switching from one ChEI to
ments are not evidenced, absence of deterioration data have another may have negative sequelae (Bhanji and Gauthier,
tended to favour the use of ChEI in DLB with one study 2005), so careful cross-tapering may be required in these
(Mori et al., 2012) observing a lack of clinical decline in a instances.
treated donepezil group (96% with no decline) versus pla- There are limited data for the long-term effect of ChEI
cebo group (50% with no decline) over the 3-month trial treatment in DLB although benefits appear to be sustained
period. for up to 2 years (Grace et al., 2001; Werner et al., 2006; Mori
Discerning whether one ChEI is better than another in et al., 2015) and use of ChEI may delay nursing home admis-
DLB is less clear, as there are no direct comparison trials sion (Rongve et al., 2014). Intriguingly, meta-analyses have
to favour one agent over another. Double-blind randomized also suggested that across the LBD spectrum, ChEI use may
controlled trial (RCT) evidence exists for rivastigmine and be associated with reduced mortality with odds ratio of 0.28
donepezil, whereas there has been only one uncontrolled (CI 0.09–0.84) (Rolinski et al., 2012).
trial in DLB with galantamine (Edwards et al., 2007). Thus, There is also some tentative evidence to suggest that there
while from an efficacy perspective, there is no evidence to may be added benefits to patients by increasing ChEI doses
suggest that any one ChEI is better than another in DLB above the recommended maximum, albeit, at an increased
(Bhasin et al., 2007; Stinton et al., 2015), the relative lack of likelihood of side effects (Pakrasi et al., 2006). There have
high-quality data for galantamine has perhaps biased cur- been no systematic trials to suggest that higher than recom-
rent clinical practice towards the use of rivastigmine and mended doses are truly beneficial.
donepezil in DLB.
Overall, ChEIs appear to be tolerated reasonably in 63.2.3 MEMANTINE
patients with DLB. Notable side effects, however, include
gastrointestinal symptoms, sleep disturbance, dizziness, Memantine, N-methyl-d-aspartate (NMDA) antagonist has
urinary frequency, insomnia, leg cramps, hypersalivation been trialled in DLB but results have not been conclusive.
and increased lacrimation. Sleep disturbances may respond One study by Aarsland et al. (2009) included 72 patients
to prescribing ChEI in the morning (or giving second dose with either PDD or DLB of mild-to-moderate severity who
in the afternoon for twice daily preparations) and gastroin- were randomized to 20 mg of memantine or to placebo for
testinal symptoms arising after ChEI initiation may settle 24 weeks. Key findings included a significant benefit for
over a few days. Sometimes, anti-emetics may be needed memantine over placebo in the primary outcome measure
and 5-HT3 antagonists such as ondansetron or granis- of the Clinical Global Impressions of Change (CGIC), which
etron might be suggested as helpful by some clinicians appeared stronger in PDD than DLB. There was no effect
(Boot, 2015). on secondary outcome measures such as neuropsychiatric
Symptomatic bradycardia and conduction delays as a symptoms or ADLs, although attentional speed did appear
result of Lewy body disease related to cardiac dysautono- better on memantine while parkinsonian symptoms did not
mia and are perhaps more serious, albeit, rare side effects of worsen. In a second larger study by Emre et al. (2010b), 199
ChEI in DLB (Ballard et al., 2006). Some authorities advo- patients with DLB or PDD were randomized to memantine
cate the use of baseline electrocardiograms to check for any 20 mg or placebo. However, while the primary outcome
pre-existing issues and for some patients permanent cardiac measure of CGIC was lower in the memantine group, this
pacemakers may be required although whether this should did not reach significance except in the DLB group, who also
be done specifically to allow the patient to undergo ChEI demonstrated improvements in their total Neuropsychiatric
treatment is controversial (Boot, 2015). Inventory (NPI) scores. Cognitive scores and other sec-
Finally, despite the reciprocal interplay between dopa- ondary outcome measures were not statistically different
mine and acetylcholine in the striatum, only a minority of between treatment and placebo groups.
patients appear to have exacerbations of their motor symp- It is likely that patient heterogeneity played a large role
toms with ChEI treatment (Thomas et al., 2005). in the above studies and introduced a degree of response
The treatment of dementia with Lewy bodies 733

variability. In addition, notably, in the Aarsland et al. (2009) can provoke profound neuroleptic sensitivity reactions in up
study, concurrent use of ChEI was allowed, whereas they to 50% of DLB patients and these reactions have been asso-
were not in the Emre et al. (2010b) trial, which may have ciated with significant morbidity and mortality (McKeith
influenced the findings. et al., 1995). Given the frequent occurrence of cardiac
Overall, these mixed data provide some tentative evi- denervation in DLB (King et al., 2011), QT prolongation
dence that memantine might be beneficial, but it is equivocal may also be a risk. Across the dementia landscape, there
whether it helps patients with DLB and/or PDD and it remains have been significant concerns about the use of antipsy-
unclear what symptom domains would be best targeted by chotics in dementia, not in the least as a result of evidence
the memantine. Clearly, further studies are needed in this suggesting significant increases in mortality, cerebrovascu-
area, making use of different designs with responder analy- lar disease and cognitive decline associated with the use of
ses to clarify the utility of memantine in DLB. Longitudinal these medications (Wang et al., 2005; Schneider et al., 2006;
data from a subsample of the patients in the Aarsland et al. Ballard et al., 2009).
(2009) trial have suggested that early treatment and positive Specific evidence for the utility of antipsychotics in DLB
clinical response to memantine might predict longer survival is also lacking. A secondary analysis of an RCT of olanzap-
in patients with DLB and PDD (Stubendorff et al., 2014) and ine in AD where patients were reclassified retrospectively
a secondary analysis of reaction time data has favoured the as DLB (Cummings et al., 2002) found that this antipsy-
use of memantine in terms of improvements in attention in chotic leads to reductions in hallucinations and delusions;
DLB and PDD patients (Wesnes et al., 2015). however, this finding was balanced with marked intolerance
Practically, despite some case reports suggesting of DLB patients to olanzapine even when treated with low
increased agitation in DLB (Ridha et al., 2005), the major doses (Marsh et al., 2001).
trials described above indicate that memantine is generally Risperidone has also been trialled in an RCT of 31 DLB
a well-tolerated agent and thus, it may be worth trialling in patients (Culo et al., 2010). Similar to olanzapine, this anti-
selected patients, particularly, those in whom ChEI are con- psychotic led to a worsening in psychiatric and cognitive
traindicated or cannot be tolerated. symptoms with marked withdrawals in the study.
Clozapine has an evidence base for efficacy in PD with
63.2.4 DOPAMINE REPLACEMENT psychosis (Eng and Welty, 2010) and has also been tried in
PDD (Lee et al., 2007), but there is no evidence for its use
Use of levodopa/carbidopa in Parkinson’s disease (PD) is well in DLB; aside from the obvious risk of agranulocytosis it is
established; however, their use in LBD and, in particular, DLB likely that side effects such as drooling, sedation, tremors,
is less clear due to a lack of adequate trial data. Uncontrolled constipation and delirium, which have been reported in PD/
studies have demonstrated acute and chronic improvements PDD patients taking clozapine would also limit the utility of
in motor functioning and reductions in tremor for individuals clozapine in DLB.
with DLB and PDD with levodopa (Bonelli et al., 2004; Molloy A favourite antipsychotic choice of clinicians, at present,
et al., 2005; Goldman et al., 2008; Onofrj et al., 2013). Motor for DLB, appears to be quetiapine, given its lower tendency
improvements in response to levodopa treatment appear more for side effects. However, evidence for efficacy in treatment
common in PDD (65%–70%) than DLB (32%–50%) (Bonelli of agitation or psychosis in DLB is limited with regard to
et al., 2004; Onofrj et al., 2013; Stinton et al., 2015). this drug (Takahashi et al., 2003; Kurlan et al., 2007).
A major challenge with levodopa use in DLB is the In the context of the above, given the relative burden of
increased risk of psychotic symptoms such as hallucina- side effects to possible efficacy, a high level of caution needs
tions; certainly about one-third of those who derive motor to be applied to the use of antipsychotics in DLB. They
benefits also experience worsening of their psychotic symp- should only be considered as a second or third option when
toms (Goldman et al., 2008). other therapeutic avenues have been exhausted and in gen-
There are little or no data for the use of other anti-­ eral, they should not be used to treat non-specific agitation
parkinsonian agents such as amantadine, catechol-O-­ or behavioural disturbances but should only be reserved for
methyltransferase (COMT) inhibitors, rotigotine selegiline those patients with clear psychosis as these symptoms are
etc. in DLB although their tolerability is likely to be poor the ones most likely to respond to an antipsychotic (Ballard
and their propensity towards causing psychosis and compul- et al., 2004).
sive symptoms significantly limits their use in DLB (Stinton
et al., 2015). Thus, treatment of any motor symptoms in DLB 63.2.6 ANTIDEPRESSANTS, ANXIOLYTICS
is typically recommended to be limited to a levodopa mono- AND STIMULANTS
therapy regime.
Depression and anxiety are common symptoms in DLB
63.2.5 ANTIPSYCHOTICS affecting up to approximately two thirds (59%) and one-
third (27%) of patients, respectively (Fritze et al., 2011;
Alternatives to ChEI for the management of behavioural Boot et al., 2013). However, there is a paucity of evidence
symptoms include the use of antipsychotics, but these regarding what treatments might be helpful for these symp-
agents, particularly typical antipsychotics (e.g. haloperidol) toms in DLB. One small trial in DLB of citalopram versus
734 Dementia

risperidone in terms of neuropsychiatric/behavioural ben- circadian modulator is anecdotal in DLB but other data in
efits found more than 70% of participants withdrew from PD and related disorders suggest it might be effective (Boeve
the study due to worsening of psychiatric symptoms or side et al., 2003; Dowling et al., 2005). Overall, given its low side
effects (Culo et al., 2010). effect profile, melatonin may be reasonable first choice in
There are some unpublished data in PDD to support the DLB for treatment of RBD.
use of duloxetine, escitalopram and trazodone in treating Restless legs and periodic limb movements can also be
depression but little of such other kind of data are available an issue in DLB and treatment can be a challenge; in PD
(Stinton et al., 2015). Pragmatically, avoidance of tricyclics, these symptoms can be treated with dopamine agonists,
given their anticholinergic properties, is sensible; low dose gabapentin, pregabalin and benzodiazepines, but the pro-
trials of selective Serotonin re-uptake inhibitors (SSRIs), clivity of these medications for significant side effects in
Serotonin-norepinephrine re-uptake inhibitors (SNRIs) DLB limits their use.
(duloxetine, venlafaxine) or noradrenergic and specific
serotonergic antidepressants (NaSSAs) (mirtazapine) may 63.2.8 MEDICATIONS FOR AUTONOMIC
represent reasonable alternatives for treatment of depres- SYMPTOMS IN DLB
sion and/or anxiety. One should also be cautious regard-
ing the use of benzodiazepines and pregabalin for anxiety There are a wide range of autonomic symptoms in DLB
in DLB, given the propensity of these medications to cause ranging from orthostatic hypotension, urinary and sexual
sedation, falls and increased confusion. dysfunction, dysphagia and gastroparesis to constipation,
Notably, depression was one of four symptoms in an aggre- altered sweating and sialorrhoea. These symptoms are often
gate NPI symptom score, which demonstrated improvement neglected in clinical settings which focus on cognitive, neu-
in the major ChEI trials in DLB (McKeith et al., 2000; Mori ropsychiatric or motor symptoms; yet autonomic-related
et al., 2012). Thus, it is possible that ChEI may also have a problems can be severe and functionally limiting for people
role to play in treating depression in DLB patients. with DLB. It is important to enquire after these symptoms
Apathy is another major, yet often unrecognized symp- as treatment of them can have significant positive impacts
tom in DLB. It can have significant impacts on patients and on the quality of life (QoL) of patients and their carers.
their families (Bjoerke-Bertheussen et al., 2012). If apathy First-line treatment of autonomic symptoms is typically
is part of a depressive constellation, it may respond to anti- non-pharmacological (Table 63.1) and rationalizing; if pos-
depressant treatment although, again evidence is lacking sible, other medications that can exacerbate these symptoms
for any specifically efficacious agent. Alternatively, apathy is also an important consideration. For example, orthostatic
as part of impaired arousal clustered with daytime somno- hypotension may be worsened by anti-parkinsonian drugs,
lence/drowsiness may respond to ChEI. antidepressants, antihypertensives etc. However, if these
Stimulants such as methylphenidate, modafinil/ measures fail, then pharmacological interventions may be
armodafinil have also been advocated by some special- required. For orthostatic hypotension, fludrocortisone or
ists with case series data, suggesting the latter agents may midodrine may be used, although, these may need to be ini-
lead to global improvements as well as increased wakeful- tiated and monitored by a specialist. As noted above, car-
ness and attention in DLB (Varanese et al., 2013). However, diac sympathetic innervation can be compromised in DLB
data are limited to make recommendations about the use of leading to syncope in some patients which, in some cases,
stimulants in DLB and the marked side effect propensity of may require a pacemaker.
these agents is a significant factor limiting their use. For troublesome sialorrhoea, anticholinergics should
be avoided, given their negative effects on cognition; botu-
63.2.7 SLEEP MEDICATIONS linum toxin injections offer a good alternative to salivary
glands. There is also evidence in PD for glycopyrrolate in
Sleep disturbances are common and troublesome symp- the treatment of sialorrhoea (Arbouw et al., 2010), although
toms in DLB and often have significant impacts on carers there is no trial data in DLB.
(Galvin et al., 2010a). The archetypal sleep symptom in For constipation symptom in PD, there are exten-
DLB is REM sleep behaviour disorder (RBD), which affects sive management guidelines (Seppi et al., 2011) and these
approximately 70% of DLB patients (Ferman et al., 2011). are also, probably applicable to DLB. For example, in PD
Pharmacological management of RBD has focused on the patients, macrogol had demonstrable efficacy in an RCT
use of low dose clonazepam (0.5 mg–1.0 mg nocte) and while (Zangaglia et al., 2007) and similarly, lubiprostone also
there is case series data to support its efficacy (Massironi appears to have a good evidence base for improving consti-
et al., 2003), there is a lack of trial data. Furthermore, the pation (Ondo et al., 2012).
potential for significant side effects by use of benzodiaze- Urinary symptoms including urge and nocturia can be
pines in a fall-prone dementia group such as DLB is a con- particularly challenging as ChEI can exacerbate urinary
cern and benzodiazepines can also worsen any concurrent symptoms making the situation difficult to manage and urge
sleep apnoea (Postuma et al., 2012). incontinence medications have centrally had anticholiner-
Some clinicians use melatonin for RBD and other sleep gic effects; newer drugs such as solifenacin, darifenacin and
disturbances in DLB. The evidence for efficacy of this trospium that have less proclivity to cross the blood–brain
The treatment of dementia with Lewy bodies 735

Table 63.1 Examples of symptoms in DLB and their possible non-pharmacological management

Symptom Non-pharmacological advice


Cognitive fluctuations and May respond to enhanced environmental novelty (e.g. new places) and improving social
reduced arousal interactions (e.g. attendance at day care centres may help). Occupational therapy
input may be useful in helping to develop cognitive and social remediation strategies.
Visual hallucinations Can be exacerbated by eye disease and environmental issues. Correcting for refractive
errors, cataract removal, providing improved illumination and removal of patterned
furnishings that can provoke illusionary experiences may all be helpful. For some,
focusing attention on the visual hallucination experience (especially in cognitively less
impaired individuals) may reduce them. Others find looking away from visual
hallucination and then looking back at where the hallucination was can cause them to
disappear.
Parkinsonism and falls A physiotherapy assessment may clarify if there are any modifiable contributors to poor
mobility, which could be treated, e.g. frailty, arthritis. Occupational therapists can
advise on home modifications, e.g. fitting of grab bars, railings etc.
REM sleep behaviour These symptoms are often more distressing for spouses than the patient themselves and
symptoms it may be necessary for bed partners to sleep separately, although, this often needs to
be sensitively discussed.
If the patient is at risk of injury or falling out of bed, lowering the bed/putting the
mattress on floor is an option. Using a sleeping bag may be beneficial and removal of
dangerous/sharp objects in the bedroom is advisable.
Constipation High fibre diets, encouraging fluid intake, regular exercise and use of stool softeners
may help.
Urinary incontinence Avoid excessive fluid intake including fizzy, acidic or caffeinated drinks. Regular voiding
pattern and incontinence pads may help some. If symptoms are particularly
troublesome, seeking advice from an incontinence nurse and/or urologist may be
appropriate.
Orthostatic hypotension Advise the patient to be careful about the increase in fluid and salt intake. Compression
hosiery may be suitable for some. Advise the patient to take time to rise from a supine
to standing position. For orthostatic hypotension occurring early in the morning,
raising the head of the bed can help.
Abbreviations: DLB, dementia with Lewy bodies; REM, rapid eye movement.

barrier may be better (Kessler et al., 2011; Pagoria et al.,


2011) in such cases. Mirabegron, a beta-3 adrenergic ago- 63.4 FUTURE THERAPIES
nist, may be alternative for urinary urgency although there
is no evidence for its use in DLB. As is evident from the previous sections, there is a dearth of
high-level evidence in the treatment of DLB. The strongest
evidence is the use of ChEI, specifically rivastigmine and
donepezil for cognitive impairment, cognitive fluctuations,
63.3 NON-PHARMACOLOGICAL
neuropsychiatric symptoms and global improvements in
TREATMENTS function. ChEIs, therefore, probably represent good first-
line and broad-spectrum treatment options in DLB.
Systematic evaluation of non-pharmacological interven- However, even with ChEI, there is still a relative lack
tions in DLB is lacking and most recommendations are on of data in terms of the number of trials and numbers of
the basis of anecdote, clinical experience, case study and participants within these studies compared to those trials
analogy of treatment benefits in other conditions that over- which have been carried out in AD. Another major chal-
lap to some degree with DLB (e.g. PD or AD). Examples of lenge in DLB trials is a high degree of heterogeneity in
possible non-pharmacological strategies, based on clinical patient response to interventions and this is typified by the
experience and anecdotal evidence which may help DLB mixed results seen in the major memantine trials (Aarsland
symptoms are described in Table 63.1. Broadly, multifaceted et al., 2009; Emre et al., 2010b). Specific aetio-pathological
and tailored behavioural strategies and caregiver training processes, perhaps in the distribution of alpha-synuclein
programmes for agitation and behavioural disturbances and rate of pathological change, which leads to divergences
and which are applicable to a range of dementias should also in the clinical phenotype, for example, in some leading to
be considered (Gitlin et al., 2012; Kales et al., 2015). the manifestation of PD/PDD and in others, DLB, are likely
736 Dementia

to be important factors. Better patient stratification on the Ballard, C., Hanney, M.L., Theodoulou, M. et al. (2009).
basis of biomarkers is likely to be highly relevant for future The dementia antipsychotic withdrawal trial (DART-AD):
DLB trials. Long-term follow-up of a randomised placebo-con-
Targeting neurotransmitter systems other than the cho- trolled trial. The Lancet Neurology, 8: 151–157.
linergic system may hold promise. As noted earlier, there are Ballard, C., Lane, R., Barone, P. et al. (2006). Cardiac safety
limited trial data to support the notion that improvements of rivastigmine in Lewy body and Parkinson’s disease
in the noradrenergic system using agents such as amodi- dementias. International Journal of Clinical Practice, 60
final may have benefits related to improved alertness and (6): 639–645.
decreased drowsiness. In PD, a recent double-blind RCT Ballard, C.G., Thomas, A., Fossey, J. et al. (2004). A
Phase III study of pimavanserin, a serotonin 2A receptor 3-month, randomized, placebo-controlled, neuroleptic
inverse agonist, suggested that this agent may help amelio- discontinuation study in 100 people with dementia:
rate psychotic symptoms (Cummings et al., 2014); whether The neuropsychiatric inventory median cutoff is a
mechanistically this medication may have benefits in DLB predictor of clinical outcome. The Journal of Clinical
remains an intriguing unanswered question. Psychiatry, 65: 114–119.
Deep brain stimulation (DBS) has been used with success Bhanji, N.H. and Gauthier, S. (2005). Emergent complica-
to treat motor symptoms in PD and there are ongoing trials tions following donepezil switchover to galantamine in
focusing on the stimulation of the nucleus basalis of Meynert three cases of dementia with Lewy bodies. The Journal
in DLB patients with cognitive impairment as a novel of Neuropsychiatry and Clinical Neurosciences, 17:
approach to enhance cognition (ClinicalTrials.gov, 2015). 552–555.
Furthermore, non-invasive stimulation techniques such as Bhasin, M., Rowan, E., Edwards, K. and Mckeith, I. (2007).
transcranial magnetic stimulation (TMS) have demonstrated Cholinesterase inhibitors in dementia with Lewy bod-
some antidepressant effects in DLB (Takahashi et al., 2009). ies – A comparative analysis. International Journal of
As a final point, the prevalence of DLB is estimated to Geriatric Psychiatry, 22: 890–895.
be about 4%–7% of dementia diagnoses (Vann Jones and Bjoerke-Bertheussen, J., Ehrt, U., Rongve, A. et al. (2012).
O’Brien, 2014) depending upon the clinical sample, which Neuropsychiatric symptoms in mild dementia with
contrasts with the higher prevalence of Lewy body disease lewy bodies and Alzheimer’s disease. Dementia and
of up to 20% in dementia cases evidenced on post-mortem Geriatric Cognitive Disorders, 34: 1–6.
(McKeith et al., 2005). Thus, one of the main obstacles pro- Boeve, B.F., Silber, M.H. and Ferman, T.J. (2003).
viding best care to DLB patients is obtaining the initial Melatonin for treatment of REM sleep behavior dis-
diagnosis and this is reflected in the fact that often, patients order in neurologic disorders: Results in 14 patients.
with DLB attend numerous different specialist services Sleep Medicine, 4: 281–284.
prior to obtaining a diagnosis and it is estimated that, cur- Bonelli, S.B., Ransmayr, G., Steffelbauer, M. et al. (2004).
rently, only one in three DLB cases is correctly diagnosed, L-dopa responsiveness in dementia with Lewy bod-
even after referral to specialist dementia services (Galvin ies, Parkinson disease with and without dementia.
et al., 2010b). The remaining cases are typically to be misdi- Neurology, 63: 376–378.
agnosed as being AD or vascular dementia (VaD) cases, or Boot, B.P. (2015). Comprehensive treatment of demen-
given another psychiatric diagnosis instead. Specialist train- tia with Lewy bodies. Alzheimer’s Research and
ing in the diagnosis and use of biomarkers may improve this Therapy, 7: 45.
situation. Boot, B.P., Orr, C.F., Ahlskog, J.E. et al. (2013). Risk factors
for dementia with Lewy bodies: A case-control study.
Neurology, 81 (9): 833–840.
REFERENCES
ClinicalTrials.gov. (2015). Deep Brain Stimulation for
Aarsland, D., Ballard, C., Walker, Z. et al. (2009). Patients with Dementia with Lewy Bodies. Available
Memantine in patients with Parkinson’s disease at https://clinicaltrials.gov/ct2/show/NCT02263937.
dementia or dementia with Lewy bodies: A double- Accessed 22 December 2015.
blind, placebo-controlled, multicentre trial. The Lancet Culo, S., Mulsant, B.H., Rosen, J. et al. (2010). Treating
Neurology, 8: 613–618. neuropsychiatric symptoms in dementia with Lewy
Aarsland, D., Laake, K., Larsen, J.P. and Janvin, C. (2002). bodies: A randomized controlled-trial. Alzheimer
Donepezil for cognitive impairment in Parkinson’s Disease and Associated Disorders, 24: 360–364.
disease: A randomised controlled study. Journal Cummings, J., Isaacson, S., Mills, R. et al. (2014).
of Neurology, Neurosurgery, and Psychiatry, 72: Pimavanserin for patients with Parkinson’s disease
708–712. Erratum appears in Journal of Neurology, psychosis: A randomised, placebo-controlled phase 3
Neurosurgery, and Psychiatry (2002 Sep), 73 (3): 354. trial. Lancet, 383: 533–540.
Arbouw, M.E., Movig, K.L., Koopmann, M. et al. (2010). Cummings, J.L., Street, J., Masterman, D. and Clark,
Glycopyrrolate for sialorrhea in Parkinson disease: A W.S. (2002). Efficacy of olanzapine in the treatment of
randomized, double-blind, crossover trial. Neurology, psychosis in dementia with lewy bodies. Dementia and
74: 1203–1207. Geriatric Cognitive Disorders, 13: 67–73.
The treatment of dementia with Lewy bodies 737

Dowling, G.A., Mastick, J., Colling, E. et al. (2005). Goldman, J.G., Goetz, C.G., Brandabur, M. et al. (2008).
Melatonin for sleep disturbances in Parkinson’s dis- Effects of dopaminergic medications on psychosis
ease. Sleep Medicine, 6: 459–466. and motor function in dementia with Lewy bodies.
Dubois, B., Tolosa, E., Katzenschlager, R. et al. (2012). Movement Disorders, 23: 2248–2250.
Donepezil in Parkinson’s disease dementia: A ran- Grace, J., Daniel, S., Stevens, T. et al. (2001). Long-
domized, double-blind efficacy and safety study. term use of rivastigmine in patients with dementia
Movement Disorders, 27: 1230–1238. with Lewy bodies: An open-label trial. International
Edwards, K., Royall, D., Hershey, L. et al. (2007). Efficacy Psychogeriatrics, 13: 199–205.
and safety of galantamine in patients with dementia with Gray, S.L., Anderson, M.L., Dublin, S. et al. (2015).
Lewy bodies: A 24-week open-label study. Dementia and Cumulative use of strong anticholinergics and incident
Geriatric Cognitive Disorders, 23: 401–405. dementia: A prospective cohort study. JAMA Internal
Emre, M., Aarsland, D., Albanese, A. et al. (2004). Medicine, 175: 401–407.
Rivastigmine for dementia associated with Parkinson’s Jewart, R.D., Green, J., Lu, C.J. et al. (2005). Cognitive,
disease. The New England Journal of Medicine, 351: behavioral, and physiological changes in Alzheimer
2509–2518. disease patients as a function of incontinence medica-
Emre, M., Bernabei, R., Blesa, R. et al. (2010a). Drug tions. The American Journal of Geriatric Psychiatry, 13:
profile: Transdermal rivastigmine patch in the treat- 324–328.
ment of Alzheimer disease. CNS Neuroscience and Kales, H.C., Gitlin, L.N. and Lyketsos, C.G. (2015).
Therapeutics, 16 (4): 246–253. Assessment and management of behavioral and
Emre, M., Cummings, J.L. and Lane, R.M. (2007). psychological symptoms of dementia. British Medical
Rivastigmine in dementia associated with Parkinson’s Journal, 350: h369.
disease and Alzheimer’s disease: Similarities and dif- Kalisch Ellett, L.M., Pratt, N.L., Ramsay, E.N. et al.
ferences. Journal of Alzheimer’s Disease, 11: 509–519. (2014). Multiple anticholinergic medication use and
Emre, M., Tsolaki, M., Bonuccelli, U. et al. (2010b). risk of hospital admission for confusion or demen-
Memantine for patients with Parkinson’s disease tia. Journal of the American Geriatrics Society, 62:
dementia or dementia with Lewy bodies: A ran- 1916–1922.
domised, double-blind, placebo-controlled trial. The Kessler, T.M., Bachmann, L.M., Minder, C. et al. (2011).
Lancet Neurology, 9: 969–977. Adverse event assessment of antimuscarinics for
Eng, M.L. and Welty, T.E. (2010). Management of hal- treating overactive bladder: A network meta-analytic
lucinations and psychosis in Parkinson’s disease. The approach. PLoS ONE, 6: e16718.
American Journal of Geriatric Pharmacotherapy, 8: King, A.E., Mintz, J. and Royall, D.R. (2011). Meta-analysis
316–330. of 123I-MIBG cardiac scintigraphy for the diagnosis of
Ferman, T.J., Boeve, B.F., Smith, G.E. et al. (2011). Lewy body-related disorders. Movement Disorders, 26
Inclusion of RBD improves the diagnostic classifica- (7): 1218–1224.
tion of dementia with Lewy bodies. Neurology, 77: Kurlan, R., Cummings, J., Raman, R. and Thal, L. (2007).
875–882. Quetiapine for agitation or psychosis in patients
Fox, C., Richardson, K., Maidment, I.D. et al. (2011). with dementia and parkinsonism. Neurology, 68:
Anticholinergic medication use and cognitive impair- 1356–1363.
ment in the older population: The Medical Research Lee, H.B., Hanner, J.A., Yokley, J.L. et al. (2007). Clozapine
Council Cognitive Function and Ageing Study. Journal for treatment-resistant agitation in dementia. Journal
of the American Geriatrics Society, 59: 1477–1483. of Geriatric Psychiatry and Neurology, 20: 178–182.
Fritze, F., Ehrt, U., Sonnesyn, H. et al. (2011). Depression Marsh, L., Lyketsos, C. and Reich, S.G. (2001). Olanzapine
in mild dementia: Associations with diagnosis, APOE for the treatment of psychosis in patients with
genotype and clinical features. International Journal of Parkinson’s disease and dementia. Psychosomatics, 42:
Geriatric Psychiatry, 26: 1054–1061. 477–481.
Galvin, J.E., Duda, J.E., Kaufer, D.I. et al. (2010a). Lewy Massironi, G., Galluzzi, S. and Frisoni, G.B. (2003). Drug
body dementia: Caregiver burden and unmet needs. treatment of REM sleep behavior disorders in demen-
Alzheimer Disease and Associated Disorders, 24: tia with Lewy bodies. International Psychogeriatrics, 15:
177–181. 377–383.
Galvin, J.E., Duda, J.E., Kaufer, D.I. et al. (2010b). Lewy Mckeith, I.G., Ballard, C.G. and Harrison, R.W. (1995).
body dementia: The caregiver experience of clini- Neuroleptic sensitivity to risperidone in Lewy body
cal care. Parkinsonism and Related Disorders, 16 (6): dementia. Lancet, 346: 699.
388–392. Mckeith, I., Del Ser, T., Spano, P. et al. (2000). Efficacy of
Gitlin, L.N., Kales, H.C. and Lyketsos, C.G. (2012). rivastigmine in dementia with Lewy bodies: A ran-
Nonpharmacologic management of behavioral symp- domised, double-blind, placebo-controlled interna-
toms in dementia. JAMA, 308: 2020–2029. tional study. Lancet, 356: 2031–2036.
738 Dementia

Mckeith, I.G., Dickson, D.W., Lowe, J. et al. (2005). Rongve, A., Vossius, C., Nore, S. et al. (2014). Time until
Diagnosis and management of dementia with nursing home admission in people with mild demen-
Lewy bodies. Third report of the DLB consortium. tia: Comparison of dementia with Lewy bodies and
Neurology, 65: 1863–1872. Alzheimer’s dementia. International Journal of Geriatric
Mckeith, I.G., Wesnes, K.A., Perry, E. and Ferrara, R. (2004). Psychiatry, 29: 392–398.
Hallucinations predict attentional improvements with Samuel, W., Caligiuri, M., Galasko, D. et al. (2000). Better
rivastigmine in dementia with Lewy bodies. Dementia cognitive and psychopathologic response to donepezil
and Geriatric Cognitive Disorders, 18: 94–100. in patients prospectively diagnosed as dementia with
Minett, T.S., Thomas, A., Wilkinson, L.M. et al. (2003). Lewy bodies: A preliminary study. International Journal
What happens when donepezil is suddenly withdrawn? of Geriatric Psychiatry, 15: 794–802.
An open label trial in dementia with Lewy bodies Schneider, L.S., Tariot, P.N., Dagerman, K.S. et al. (2006).
and Parkinson’s disease with dementia. International Effectiveness of atypical antipsychotic drugs in
Journal of Geriatric Psychiatry, 18: 988–993. patients with Alzheimer’s disease. The New England
Molloy, S., Mckeith, I.G., O’Brien, J.T. and Burn, D.J. Journal of Medicine, 355 (15): 1525–1538.
(2005). The role of levodopa in the management of Seppi, K., Weintraub, D., Coelho, M. et al. (2011). The
dementia with Lewy bodies. Journal of Neurology, Movement Disorder Society Evidence-Based Medicine
Neurosurgery and Psychiatry, 76: 1200–1203. Review Update: Treatments for the non-motor symp-
Mori, E., Ikeda, M. and Kosaka, K. (2012). Donepezil for toms of Parkinson’s disease. Movement Disorders, 26
dementia with Lewy bodies: A randomized, placebo- (Suppl. 3): S42–S80.
controlled trial. Annals of Neurology, 72: 41–52. Stinton, C., Mckeith, I., Taylor, J.P. et al. (2015).
Mori, E., Ikeda, M., Nagai, R. et al. (2015). Long-term Pharmacological management of Lewy body demen-
donepezil use for dementia with Lewy bodies: Results tia: A systematic review and meta-analysis. American
from an open-label extension of Phase III trial. Journal of Psychiatry, 172 (8): 731–742.
Alzheimer’s Research and Therapy, 7 (1): 5. Stubendorff, K., Larsson, V., Ballard, C. et al. (2014).
Ondo, W.G., Kenney, C., Sullivan, K. et al. (2012). Placebo- Treatment effect of memantine on survival in dementia
controlled trial of lubiprostone for constipation with Lewy bodies and Parkinson’s disease with demen-
associated with Parkinson disease. Neurology, 78: tia: A prospective study. BMJ Open, 4 (7): e005158.
1650–1654. Takahashi, S., Mizukami, K., Yasuno, F. and Asada, T.
Onofrj, M., Varanese, S., Bonanni, L. et al. (2013). Cohort (2009). Depression associated with dementia with
study of prevalence and phenomenology of tremor in Lewy bodies (DLB) and the effect of somatotherapy.
dementia with Lewy bodies. Journal of Neurology, 260 Psychogeriatrics, 9: 56–61.
(7): 1731–1742. Takahashi, H., Yoshida, K., Sugita, T. et al. (2003).
Pagoria, D., O’Connor, R.C. and Guralnick, M.L. (2011). Quetiapine treatment of psychotic symptoms and
Antimuscarinic drugs: Review of the cognitive impact aggressive behavior in patients with dementia with
when used to treat overactive bladder in elderly Lewy bodies: A case series. Progress in Neuro-
patients. Current Urology Reports, 12: 351–357. Psychopharmacology and Biological Psychiatry, 27:
Pakrasi, S., Thomas, A., Mosimann, U.P. et al. (2006). 549–553.
Cholinesterase inhibitors in advanced Dementia with Thomas, A.J., Burn, D.J., Rowan, E.N. et al. (2005). A
Lewy bodies: Increase or stop? International Journal of comparison of the efficacy of donepezil in Parkinson’s
Geriatric Psychiatry, 21: 719–721. disease with dementia and dementia with Lewy bod-
Perry, E.K., Kerwin, J., Perry, R.H. et al. (1990). Visual ies. International Journal of Geriatric Psychiatry 20:
­hallucinations and the cholinergic system in 938–944.
­dementia. Journal of Neurology, Neurosurgery, and Tiraboschi, P., Hansen, L.A., Alford, M. et al.
Psychiatry, 53: 88. (2002). Early and widespread cholinergic losses
Postuma, R.B., Gagnon, J.F. and Montplaisir, J.Y. (2012). ­differentiate dementia with Lewy bodies from
REM sleep behavior disorder: From dreams to neuro- Alzheimer ­disease. Archives of General Psychiatry, 59:
degeneration. Neurobiology of Disease, 46: 553–558. 946–951.
Ridha, B.H., Josephs, K.A. and Rossor, M.N. (2005). Vann Jones, S.A. and O’Brien, J.T. (2014). The preva-
Delusions and hallucinations in dementia with Lewy lence and incidence of dementia with Lewy bodies: A
bodies: Worsening with memantine. Neurology, 65: systematic review of population and clinical studies.
481–482. Psychological Medicine, 44: 673–683.
Rolinski, M., Fox, C., Maidment, I. and Mcshane, R. (2012). Varanese, S., Perfetti, B., Gilbert-Wolf, R. et al. (2013).
Cholinesterase inhibitors for dementia with Lewy bod- Modafinil and armodafinil improve attention and
ies, Parkinson’s disease dementia and cognitive impair- global mental status in Lewy bodies disorders:
ment in Parkinson’s disease. The Cochrane Database Preliminary evidence. International Journal of Geriatric
of Systematic Reviews, (3): CD006504. Psychiatry, 28: 1095–1097.
The treatment of dementia with Lewy bodies 739

Wang, P.S., Schneeweiss, S., Avorn, J. et al. (2005). Werner, P., Erik, W., Murat, E. et al. (2006). Long-term
Risk of death in elderly users of conven- benefits of rivastigmine in dementia associated with
tional vs. atypical antipsychotic medications. Parkinson’s disease: An active treatment extension
The New England Journal of Medicine, 353: study. Movement Disorders, 21: 456–461.
2335–2341. Wesnes, K.A., Aarsland, D., Ballard, C. and Londos, E.
Wang, H.F., Yu, J.T., Tang, S.W. et al. (2015). Efficacy (2015). Memantine improves attention and episodic
and safety of cholinesterase inhibitors and meman- memory in Parkinson’s disease dementia and dementia
tine in cognitive impairment in Parkinson’s dis- with Lewy bodies. International Journal of Geriatric
ease, Parkinson’s disease dementia, and d ­ ementia Psychiatry, 30: 46–54.
with Lewy bodies: Systematic review with Zangaglia, R., Martignoni, E., Glorioso, M. et al. (2007).
­meta-­analysis and trial sequential analysis. Journal Macrogol for the treatment of constipation in
of Neurology, Neurosurgery, and Psychiatry, 86: Parkinson’s disease. A randomized placebo-controlled
135–143. study. Movement Disorders, 22: 1239–1244.
64
Cognitive impairment and dementia in
Parkinson’s disease

DAG AARSLAND, KOLBJØRN BRØNNICK, MILICA G. KRAMBERGER AND


JOANA B. PEREIRA

other neuropsychiatric symptoms affect the quality of life,


64.1 INTRODUCTION contribute to caregiver distress and increased risk of nurs-
ing home placement, and is associated with more psychiat-
Parkinson’s disease (PD) is a common neurodegenerative ric symptoms such as depression and hallucinations, higher
disorder affecting about 1.5% of people aged 65 or older (de mortality and functional disability and higher risk for drug
Rijk et al., 1997). PD is defined pathologically as cell loss toxicity. This chapter discusses the epidemiology, pathophys-
in the pigmented dopaminergic cells of the substantia nigra iologic mechanism, clinical presentation and management of
pars compacta and synuclein pathology (Lewy neurites and cognitive impairment and dementia in patients with PD.
Lewy bodies) in the surviving cells. In addition, cholinergic
forebrain nuclei and other brain stem nuclei including the
serotonergic raphe nuclei and the noradrenergic locus coe-
64.2 COGNITIVE IMPAIRMENT IN PD
ruleus are usually affected. The topographical progression
subsequently involves the anteromedial temporal meso-
cortex, including the transentorhinal region, and reaches Cognitive impairment is common in PD and has a major clin-
into adjoining high-order sensory association areas and ical impact on the patient, carers and society (Svenningsson
important limbic structures such as amygdalae and hippo- et al., 2012). Although the risk for cognitive decline increases
campus (Braak et al., 2003). The cardinal clinical features of with disease duration, mild cognitive impairment (MCI)
PD are resting tremor, bradykinesia, rigidity and postural can be detected in a considerable proportion: around 20%,
abnormalities. However, owing to the wide distribution of already at the diagnosis of PD (Foltynie et al., 2004; Aarsland
neurodegeneration, it is not surprising that a wide range of et al., 2009; Litvan et al., 2011; Weintraub et al., 2015). It is
non-motor symptoms occur as well, including neuropsychi- usually but not always (see below) a progressive decline, and
atric symptoms and autonomic dysfunction. thus the early and mild changes tend to progress to dementia
The treatment of PD involves dopamine replacement even to severe dementia in many patients.
therapy with l-3,4-dihydroxyphenylalanine (l-DOPA) and
several dopamine agonists, including the selective dopa- 64.2.1 DIAGNOSIS OF COGNITIVE
mine D3 receptor agonists ropinirole and pramipexole. In IMPAIRMENT IN PD
addition, the monoamine oxidase inhibitors selegiline and
rasagiline with a potential neuroprotective effect may slow It is crucial that clinicians seeing PD patients screen for cog-
disease progression. Deep cerebral stimulation with the nitive impairment throughout the disease course. The cog-
implantation of electrodes can be very helpful for selected nitive profile in PD varies but is usually distinct from the
patients. A wide range of behavioural side effects can occur profiles seen in Alzheimer’s disease (AD) and other demen-
during drug and surgical treatments. tias. Executive control impairment is paramount, including
It is now increasingly recognized that PD is a neuro- deficits in working memory (Possin et al., 2008), set-shifting,
psychiatric disorder and not merely a movement disorder. planning, error monitoring (Willemssen et al., 2009), response
Independent of the motor symptoms of PD, dementia and inhibition (Wylie et al., 2005) and dual tasking (Brown et al.,
740
Cognitive impairment and dementia in Parkinson’s disease 741

1991). Memory impairment is also common (Weintraub, Morphological changes involved in cognitive impair-
2004; Brønnick et al., 2011), although may partly be second- ment and dementia include cortical and limbic Lewy bodies
ary to executive/attentional deficits (Higginson et al., 2003). (Aarsland et al., 2005), but also Alzheimer-like changes, and
Visuospatial dysfunction is another hallmark of the cognitive the alpha-synuclein, amyloid and tau-related changes seem to
profile in PD (Aarsland et al., 2009; Litvan et al., 2011). act in a synergistic way (Compta et al., 2011). Consistent with
The motor symptoms and apathy can make the diagnosis the heterogeneous clinical pattern, the underlying pathology
of cognitive impairment and dementia difficult in PD, and is heterogeneous, with some patients exhibiting early and
both overdiagnosis and underdiagnosis may occur (Litvan widespread cortical neurodegeneration and an aggressive
et al., 1998). Cognitive rating scales should be used, taking clinical course, whereas others have less cortical involvement
into account the disabilities from motor symptoms. Screening and a slower decline (Ballard et al., 2006; Halliday et al., 2008).
tests such as the Mini-Mental State Examination (MMSE)
may be employed, although instruments including execu- 64.2.3 CLINICAL FACTORS ASSOCIATED
tive dysfunction, such as the Dementia Rating Scale and the WITH COGNITIVE DECLINE IN PD
Montreal Cognitive Assessment (MoCA) are more sensitive to
the early cognitive disorders in PD. Neuropsychological tests The median time to dementia in PD is 10 years, but there
are required for a reliable diagnosis of MCI (see Section 64.3.1). are wide variations in the time from onset of PD to demen-
Particular care should be taken to distinguish between cogni- tia. Age, advanced parkinsonism and MCI are predictors of
tive impairment due to confusional states from drug toxicity or dementia in PD (Svenningsson et al., 2012). Rigidity and
other diseases. Criteria to diagnose dementia in PD have been symptoms ­mediated mainly by non-dopaminergic systems,
proposed (Emre et al., 2007), and a practical guide to assist in such as speech, gait and postural disorders, are particularly
the diagnosis of Parkinson’s disease with dementia (PDD) has related to subsequent development of dementia, whereas
been published (Dubois et al., 2007). patients with a tremor-dominant pattern have a lower risk
There is considerable clinical and pathological overlap (Alves et al., 2005). The relationship with age and severity of
between dementia with Lewy bodies (DLB) and PDD. A parkinsonism seems to be due to their combined effect rather
diagnosis of PDD should be made in patients who are diag- than separate effects (Levy et al., 2002). In addition, halluci-
nosed as PD and develop dementia after at least 1 year with nations, rapid eye ­movement (REM) sleep behavior disorder
motor symptoms. If dementia develops before or within (Boot et al., 2012), early severe olfactory dysfunction (Anang
1 year after the diagnosis of PD, a diagnosis of DLB should et al., 2014), orthostatic blood pressure drop, abnormal colour
be made (McKeith et al., 2005), although this time window vision and baseline gait dysfunction are clinical predictors for
is rather arbitrary. There is a debate whether they represent global cognitive decline (Anang et al., 2014).
distinct diseases or rather two syndromes on a spectrum of
Lewy body diseases, and there are no major clinical conse- 64.2.4 GENETICS OF COGNITIVE
quences related to this differentiation. IMPAIRMENT IN PD
64.2.2 MECHANISMS UNDERLYING Some, but not all, studies have found an association between
COGNITIVE IMPAIRMENT IN PD dementia and apolipoprotein (ApoE) ε4 in PD (Williams-Gray
et al., 2009), whereas the ε2 allele may be protective (Berge
The heterogeneous cognitive deficits in PD probably reflect et al., 2014). A positive association of dementia with the H1
differing forms and location of neuropathological involve- haplotype of the microtubule-associated protein tau (MAPT)
ment. Relatively mild executive impairment, usually pres- gene has been noted (Goris et al., 2007). Others have not
ent very early in the course of the disease, is related to reported this association and also failed to find an association
the dopaminergic deficits, caused by either disruption of with the synuclein gene (Mata et al., 2014).
nigrostriatal circuitry with altered outflow of the caudate
nuclei to frontal cortex via thalamus (Rinne et al., 2000) 64.2.5 IMAGING AND COGNITIVE
or diminished dopamine activity in the frontal projections DECLINE IN PD
consequent to degeneration of mesocortical projections
(Mattay et al., 2002). Loss of cells in the cholinergic nucleus Structural brain imaging with magnetic resonance imag-
basalis of Meynert and cortical cholinergic losses of similar ing (MRI) has become a powerful technique to assess neu-
degrees as in AD have been reported, and these changes are rodegeneration in vivo in PD. Using a range of imaging
more pronounced in PD patients with dementia compared analyses, a pattern of widespread cortical atrophy in fron-
with non-demented patients (Perry et al., 1993; Tiraboschi tal, temporal, occipital and parietal regions has been found
et al., 2000). There is also evidence linking the cortical nor- in patients with dementia (Burton et al., 2004; Melzer et al.,
adrenergic system to extradimensional shift performance, 2012; Zarei et al., 2013). Limbic structures such as the hippo-
whereas, in general, visuospatial deficits may be dependent campus (Apostolova et al., 2010) and the amygdala (Junqué
on acetylcholine rather than dopamine, and these deficits et al., 2005; Bouchard et al., 2008) in addition to the caudate
are also associated with the emergence of visual hallucina- (Almeida et al., 2002), putamen and thalamus (Summerfield
tions (Kehagia et al., 2010). et al., 2005) are also atrophied in PDD. In PD patients with
742 Dementia

MCI, less extensive grey matter loss or cortical thinning considered diagnostic and possibly predictive markers for
have been found, mainly confined to prefrontal, temporal PDD. Indeed, CSF biomarkers are among the most promis-
and parietal regions (Melzer et al., 2012; Pereira et al., 2014). ing biomarker candidates for cognitive impairment in PD.
Brain structure has been consistently associated with impair- Patients with PDD have lower CSF levels of Aβ1–42 com-
ment in specific cognitive functions, with hippocampal atro- pared with patients with PD without cognitive impairment
phy correlating with verbal memory deficits in both PDD or age-matched control subjects (Siderowf et al., 2010; Alves
(Laakso et al., 1996) and PD-MCI (Beyer et al., 2012). et al., 2014) and have also shown to predict future cognitive
Diffusion tensor imaging (DTI) has also been used to decline (Siderowf et al., 2010). Inconsistent findings have
detect abnormalities in white matter microstructure in PD. been reported for levels of total tau (t-tau) and phosphor-
Reductions of fractional anisotropy, indicating white mat- ylated tau (p-tau), reporting slightly lower or unchanged
ter integrity loss, have been reported in the longitudinal levels compared with normal controls. Many studies have
fasciculi, internal capsule (Hattori et al., 2012), posterior attempted to measure alpha-synuclein levels in CSF in PD,
cingulate bundles (Matsui et al., 2007) as well as other tracts and decreased levels have been found (Kang et al., 2013). The
(Melzer et al., 2013) in PDD. In PD patients with MCI, a variation is large, and there is overlap with normal controls,
pattern of white matter abnormalities involving the corona and it has yet proven difficult to correlate alphasynuclein-
radiata, uncinate and corpus callosum was identified in the levels with cognitive decline in PD.
absence of significant grey matter changes (Agosta et al., Several studies have also explored blood-based mark-
2014), suggesting that white matter abnormalities might ers, and promising results have been shown for epidermal
precede grey matter atrophy in PD. growth factor but need to be replicated (Mollenhauer et al.,
A number of studies have also applied functional MRI 2014). There is also evidence that electroencephalogram
(fMRI) to assess the functional network characteristics of (EEG) may be a marker of cognitive impairment in PD
PD at the resting state. In patients with dementia, reduced (Bonanni et al., 2008).
functional connectivity between regions of the default
mode and visual networks has been found (Rektorova et al.,
2012), while in patients with MCI decreased connectivity in
the fronto-parietal, dorsal attention and default-mode net- 64.3 MCI IN PD
works (Amboni et al., 2014; Baggio et al., 2015) are amongst
some of the most consistent findings. 64.3.1 CLASSIFICATION AND DIAGNOSIS
Several studies have explored amyloid pathology in PD
in vivo using 11C-Pittsburgh compound B positron emis- A task force commissioned by the Movement Disorders
sion tomography (PIB PET). In a subgroup of patients with recently proposed graded criteria for the clinical diagnosis
dementia in PD, increased amyloid load has been found and of MCI in PD (Litvan et al., 2012). These include subjec-
seems to be associated with Alzheimer-like characteristics tive and objective evidence of cognitive impairment, and
both for clinical phenotype and cerebrospinal fluid (CSF) no significant functional impairment due to the cogni-
(Maetzler et al., 2009). Most studies show low amyloid bur- tive decline. The diagnosis of PD-MCI can be made using
den in PD-MCI patients that is comparable to that observed screening instruments, which are validated for PD, for
in healthy controls, although there is some evidence sug- example MOCA, or a small battery of cognitive tests at level
gesting that amyloid might have a greater value in predicting 1, whereas two standardized neuropsychological tests for
cognitive decline over time in PD (Gomperts et al., 2013). each of the five cognitive domains (memory, executive, lan-
Although both structural and functional imaging tech- guage, visuospatial and attention) are required for a diag-
niques may assist in the diagnostic workup of patients with nosis at level 2. The criterion ‘subjective cognitive decline’
parkinsonism and cognitive impairment, the final diagno- can be difficult to ascertain in PD; due to the combined
sis must be based on the history and clinical examination motor features both over- and under-reporting has been
including routine supplementary tests. shown. Another challenge is to draw the line between MCI
and dementia based on functional impairment, since both
64.2.6 CSF AND OTHER BIOMARKERS OF motor and cognitive symptoms can contribute to cognitive
COGNITION IN PD impairment. The Parkinson’s Disease-Cognitive Functional
Rating Scale (Kulisevsky et al., 2013) has been reported to be
There is much focus on identifying biomarkers, which can helpful in this regard.
assist in the diagnosis and, even more importantly, predic-
tion of cognitive decline in PD (Mollenhauer et al., 2014). 64.3.2 THE COURSE OF MCI IN PD
In addition to imaging techniques reviewed above, many
studies have explored CSF biomarkers. CSF can be useful The annual decline on MMSE in PD is one point, but with wide
in predicting development of AD. Since Alzheimer’s-type interindividual variations (Aarsland et al., 2004). There is now
brain lesions that contain amyloid beta (Aβ) proteins and convincing evidence that PD patients with MCI have a shorter
neurofibrillary tangles are associated with dementia in time to dementia than those with normal cognition, shown
PD, CSF biomarkers for assessing Aβ and tau levels may be in several large longitudinal cohort studies (Williams-Gray
Cognitive impairment and dementia in Parkinson’s disease 743

et al., 2007; Pedersen et al., 2013). There is some evidence et al., 2004). The response is particularly marked in those
that tests with a more posterior cortical basis (e.g. visuospatial with visual hallucinations (Burn et al., 2006), and an open-
tests) predicted more rapid cognitive decline compared with label extension study suggests sustained benefits for up to
fronto-striatal tests (Williams-Gray et al., 2007). This cogni- 48 weeks (Poewe et al., 2006). Similar, but somewhat less
tive dissociation was paralleled by a genetic dissociation: poly- robust findings were reported for donepezil (Dubois et al.,
morphisms of the microtubule-associated protein tau (MAPT) 2012). These findings have received further support from
gene, but not catechol O-methyltransferase (COMT) gene, meta-analyses (Rolinski et al., 2012; Wang et al., 2014) In
was associated with dementia (Goris et al., 2007), whereas a addition to the typical side effects including gastrointestinal
­polymorphism of the COMT gene was associated with atten- problems, worsening of tremor may occur in some patients,
tional dysfunction and underlying frontal activation, but not but is usually mild and transient. Whereas all PD patients
dementia risk (Williams-Gray et al., 2008). with dementia should be treated with a cholinesterase
inhibitor, there is yet no convincing evidence for PD-MCI.
Changes in the glutamatergic system have been reported
in PD. Memantine, a partial N-methyl-d-aspartate (NMDA)
64.4 DEMENTIA antagonist, suggested benefit in PDD was found to have
a significant effect in a small placebo-controlled study
64.4.1 EPIDEMIOLOGY OF DEMENTIA (Aarsland et al., 2009), and some effect was also reported
IN PD in a larger multicentre study (Emre et al., 2010); but overall,
the evidence is inconsistent (Wang et al., 2014). Memantine
Dementia develops in a considerable proportion of patients is usually well tolerated.
with PD. In a systematic review including 13 studies with Neuropsychiatric symptoms such as psychosis, apathy
1767 patients, a point prevalence of 31.3% (95% confidence and depression are common in PD and are particularly
interval 29.2–33.6), was found, and between 3% and 4% of common in PDD (Aarsland et al., 2014). There is good evi-
patients with dementia in the general population were due to dence for using clozapine for hallucinations, and a recent
PD, with an estimated prevalence of PDD in the general pop- study found effect after pimavanserin, a selective serotonin
ulation aged 65 and over of 0.3%–0.5% (Zaccai et al., 2005). 5-hydroxytryptamine receptor 2A (5-HT2A) inverse agonist
In two community-based studies, the annual incidence (Cummings et al., 2014). As is the case in DLB, PD patients
was 95.3 (Aarsland et al., 2001) and 112.5 per thousand are particularly sensitive to the motor but also cognitive side
(Marder et al., 1995), indicating that about 10% of PD effects of antipsychotic agents. There is also good evidence
patients develop dementia per year, although during the for drug treatment against depression, including paroxetine,
first years the incidence may be lower (Williams-Gray et al., venlafaxine and nortriptyline (Rocha et al., 2013). Non-
2007). The risk for developing dementia is nearly six times specific measures such as reviewing and removing drugs
higher than in non-PD subjects (Aarsland et al., 2001). In that may cause worsening of cognition, that is, anticholin-
the Sydney study, de novo PD patients were followed for ergic agents, and treating comorbid conditions, are thus of
up to 20 years, with 75% being diagnosed with dementia importance. Informing the patient and family about the risk
in the follow-up period (Hely et al., 2008). Since differen- of progressive worsening of cognitive functioning and emer-
tial mortality among demented and non-demented was not gence of psychiatric symptoms is also important.
adjusted for, the cumulative prevalence is likely to be even
higher. In the Stavanger study, based on a prevalence sam-
ple, the 8-year cumulative prevalence of dementia was cal-
culated to be 78% after controlling for attrition due to death 64.6 CONCLUSIONS
(Aarsland et al., 2003), and a high likelihood of dementia
and mortality for the higher age groups has been shown Cognitive impairment and dementia are very common in PD
(Buter et al., 2008). patients, with important clinical consequences for them and
their carers. Clinicians need to focus on cognitive impair-
ment and other neuropsychiatric symptoms in addition to the
motor symptoms in order to provide optimal care for patients.
64.5 TREATMENT OF COGNITIVE The underlying aetiology and the clinical presentation of cog-
IMPAIRMENT IN PD nitive impairment in PD are highly variable, and the noso-
logical classification of dementia in PD and its relationship to
The cholinergic loss in dementia in PD, an increase of mus- other dementias, in particular DLB, is not yet clarified. The
carinic receptor binding (Perry et al., 1993) and less neu- complex clinical presentation in these frail and elderly indi-
rodegenerative changes in neocortex compared with AD, viduals poses considerable challenges for the clinical manage-
indicate that cholinergic drugs may be useful. A large-scale ment. Rivastigmine is approved for treatment of dementia in
placebo-controlled trial over 24 weeks showed that rivastig- PD, and emerging evidence indicates that memantine may be
mine can improve cognition, activities of daily living and useful as well. The identification of multimodal biomarkers,
neuropsychiatric symptoms in patients with PDD (Emre including clinical, biochemical, neuroimaging and genetic
744 Dementia

markers will provide crucial insight into the pathophysiology Apostolova, L.G., Beyer, M., Green, A.E. et al. (2010).
of cognitive impairment in PD and future disease-modifying Hippocampal, caudate, and ventricular changes
therapies (Lin and Wu, 2015). in Parkinson’s disease with and without dementia.
Movement Disorders, 25 (6): 687–695.
Baggio, H.‐C., Segura, B., Sala‐Llonch, R. et al. (2015).
REFERENCES Cognitive impairment and resting‐state network
connectivity in Parkinson’s disease. Human Brain
Aarsland, D., Andersen, K., Larsen, J.P. et al. (2001). Risk Mapping, 36 (1): 199–212.
of dementia in Parkinson’s disease: A community- Ballard, C., Ziabreva, I., Perry, R. et al. (2006).
based, prospective study. Neurology, 56: 730–736. Differences in neuropathologic characteristics across
Aarsland, D., Andersen, K., Larsen, J.P. et al. (2003). the Lewy body dementia spectrum. Neurology, 67 (11):
Prevalence and characteristics of dementia in 1931–1934.
Parkinson disease: An 8-year prospective study. Berge, G., Sando, S.B., Rongve, A. et al. (2014).
Archives of Neurology, 60: 387–392. Apolipoprotein E ε2 genotype delays onset of demen-
Aarsland, D., Andersen, K., Larsen, J.P. et al. (2004). tia with Lewy bodies in a Norwegian cohort. Journal
The rate of cognitive decline in Parkinson’s disease. of Neurology, Neurosurgery and Psychiatry, 85 (11):
Archives of Neurology, 61: 1906–1911. 1227–1231.
Aarsland, D., Ballard, C., Walker, Z. et al. (2009). Beyer, M.K., Bronnick, K.S., Hwang, K.S. et al. (2012).
Memantine in patients with Parkinson’s disease Verbal memory is associated with structural hippocam-
dementia or dementia with Lewy bodies: A double- pal changes in newly diagnosed Parkinson’s disease.
blind, placebo-controlled, multicentre trial. Lancet Journal of Neurology, Neurosurgery and Psychiatry, 84
Neurology, 8 (7): 613–618. (1): 23–28.
Aarsland, D., Brønnick, K., Larsen, J.P. et al. (2009). Bonanni, L., Thomas, A., Tiraboschi, P. et al. (2008). EEG
Cognitive impairment in incident, untreated Parkinson comparisons in early Alzheimer’s disease, demen-
disease: The Norwegian ParkWest study. Neurology, tia with Lewy bodies and Parkinson’s disease with
72 (13): 1121–1126. dementia patients with a 2-year follow-up. Brain, 131
Aarsland, D., Perry, R., Brown, A. et al. (2005). (3): 690–705.
Neuropathology of dementia in Parkinson’s disease: Boot, B.P., Boeve, B.F., Roberts, R.O. et al. (2012).
A prospective, community-based study. Annals of Probable rapid eye movement sleep behavior disor-
Neurology, 58 (5): 773–776. der increases risk for mild cognitive impairment and
Aarsland, D., Taylor, J.P. and Weintraub, D. (2014). Parkinson disease: A population‐based study. Annals
Psychiatric issues in cognitive impairment. Movement of Neurology, 71 (1): 49–56.
Disorders, 29 (5): 651–662. Bouchard, T.P., Malykhin, N., Martin, W.R.W. et al.
Agosta, F., Canu, E., Stefanova, E. et al. (2014). Mild cogni- (2008). Age and dementia-associated atrophy
tive impairment in Parkinson’s disease is associated ­predominates in the hippocampal head and amygdala
with a distributed pattern of brain white matter dam- in Parkinson’s disease. Neurobiology of Aging, 29 (7):
age. Human Brain Mapping, 35 (5): 1921–1929. 1027–1039.
Almeida, O.P., Burton, E.J., McKeith, I. et al. (2002). MRI Braak, H., Del Tredici, K., Rub, U. et al. (2003). Staging of
study of caudate nucleus volume in Parkinson’s disease brain pathology related to sporadic Parkinson’s dis-
with and without dementia with Lewy bodies and ease. Neurobiological Aging, 24: 197–211.
Alzheimer’s disease. Dementia and Geriatric Cognitive Brønnick, K., Alves, G., Aarsland, D. et al. (2011). Verbal
Disorders, 16 (2): 57–63. memory in drug-naive, newly diagnosed Parkinson’s
Alves, G., Lange, J., Blennow, K. et al. (2014). CSF Aβ42 disease. The retrieval deficit hypothesis revisited.
predicts early-onset dementia in Parkinson disease. Neuropsychology, 25 (1): 114.
Neurology, 82 (20): 1784–1790. Brown, R.G. and Marsden, C.D. (1991). Dual task perfor-
Alves, G., Wentzel-Larsen, T., Aarsland, D. and Larsen, J.P. mance and processing resources in normal subjects and
(2005). Progression of motor impairment and disabil- patients with Parkinson’s disease. Brain, 114 (Pt 1A):
ity in Parkinson disease: A population-based study. 215–231.
Neurology, 65: 1436–1441. Burn, D., Emre, M., McKeith, I. et al. (2006). Effects of riv-
Amboni, M., Tessitore, A., Esposito, F. et al. (2014). astigmine in patients with and without visual hallucina-
Resting-state functional connectivity associated with tions in dementia associated with Parkinson’s disease.
mild cognitive impairment in Parkinson’s disease. Movement Disorders, 21 (11): 1899–1907.
Journal of Neurology, 262 (2): 425–434. Burton, E.J., McKeith, I.G., Burn, D.J. et al. (2004). Cerebral
Anang, J.B.M., Gagnon, J.F., Bertrand, J.A. et al. atrophy in Parkinson’s disease with and without
(2014). Predictors of dementia in Parkinson disease ­dementia: A comparison with Alzheimer’s disease,
A prospective cohort study. Neurology, 83 (14): dementia with Lewy bodies and controls. Brain, 127
1253–1260. (Pt 4): 791–800.
Cognitive impairment and dementia in Parkinson’s disease 745

Buter, T.C., van den Hout, A., Matthews, F.E. et al. Hattori, T., Orimo, S., Aoki, S. et al. (2012). Cognitive
(2008). Dementia and survival in Parkinson disease: status correlates with white matter alteration in
A 12-year population study. Neurology, 70 (13): Parkinson’s disease. Human Brain Mapping, 33 (3):
1017–1022. 727–739.
Compta, Y., Parkkinen, L., O’Sullivan, S.S. et al. (2011). Hely, M.A., Reid, W.G., Adena, M.A. et al. (2008). The
Lewy-and Alzheimer-type pathologies in Parkinson’s Sydney multicenter study of Parkinson’s disease:
disease dementia: Which is more important? Brain, The inevitability of dementia at 20 years. Movement
134 (5): 1493–1505. Disorders, 23 (6): 837–844.
Cummings, J., Isaacson, S., Mills, R. et al. (2014). Higginson, C.I., King, D.S., Levine, D. et al. (2003). The
Pimavanserin for patients with Parkinson’s disease relationship between executive function and verbal
psychosis: A randomised, placebo-controlled phase 3 memory in Parkinson’s disease. Brain and Cognition,
trial. The Lancet, 383 (9916): 533–540. 52 (3): 343–352.
de Rijk, M.C., Tzourio, C., Breteler, M.M. et al. (1997). Junqué, C., Ramírez‐Ruiz, B., Tolosa, E. et al. (2005).
Prevalence of parkinsonism and Parkinson’s disease Amygdalar and hippocampal MRI volumetric reduc-
in Europe: The EUROPARKINSON Collaborative tions in Parkinson’s disease with dementia. Movement
Study. European Community Concerted Action on Disorders, 20 (5): 540–544.
the Epidemiology of Parkinson’s disease. Journal of Kang, J.-H., Irwin, D.J., Chen-Plotkin, A.S. et al. (2013).
Neurology, Neurosurgery, and Psychiatry, 62: 10–15. Association of cerebrospinal fluid β-amyloid 1-42,
Dubois, B., Burn, D., Goetz, C. et al. (2007). Diagnostic T-tau, P-tau181, and α-synuclein levels with clinical
procedures for Parkinson’s disease dementia: features of drug-naive patients with early Parkinson
Recommendations from the movement disorder disease. JAMA Neurology, 70 (10): 1277–1287.
society task force. Movement Disorders, 22 (16): Kehagia, A.A., Barker, R.A. and Robbins, T.W. (2010).
2314–2324. Neuropsychological and clinical heterogeneity of
Dubois, B., Tolosa, E., Katzenschlager, R. et al. (2012). cognitive impairment and dementia in patients with
Donepezil in Parkinson’s disease dementia: A ran- Parkinson’s disease. The Lancet Neurology, 9 (12):
domized, double‐blind efficacy and safety study. 1200–1213.
Movement Disorders, 27 (10): 1230–1238. Kulisevsky, J., de Bobadilla, R.F., Pagonabarraga, J. et al.
Emre, M., Aarsland, D., Albanese, A. et al. (2004). (2013). Measuring functional impact of cognitive
Rivastigmine for dementia associated with Parkinson’s impairment: Validation of the Parkinson’s disease cog-
disease. New England Journal of Medicine, 351: nitive functional rating scale. Parkinsonism and Related
2509–2518. Disorders, 19 (9): 812–817.
Emre, M., Aarsland, D., Brown, R. et al. (2007). Clinical Laakso, M.P., Partanen, K., Riekkinen, P. et al. (1996).
diagnostic criteria for dementia associated with Hippocampal volumes in Alzheimer’s disease,
Parkinson’s disease. Movement Disorders, 22 (12): Parkinson’s disease with and without dementia, and
1689–1707. in vascular dementia: An MRI study. Neurology, 46:
Emre, M., Tsolaki, M., Bonuccelli, U. et al. (2010). 678–681.
Memantine for patients with Parkinson’s disease Levy, G., Schupf, N., Tang, M.X. et al. (2002). Combined
dementia or dementia with Lewy bodies: A ran- effect of age and severity on the risk of dementia in
domised, double-blind, placebo-controlled trial. The Parkinson’s disease. Annals of Neurology, 51: 722–729.
Lancet Neurology, 9 (10): 969–977. Lin, C.H. and Wu, R.M. (2015). Biomarkers of cognitive
Foltynie, T., Brayne, C.E., Robbins, T.W. and Barker, R.A. decline in Parkinson’s disease. Parkinsonism and
(2004). The cognitive ability of an incident cohort of Related Disorders, 21 (5): 431–443.
Parkinson’s patients in the UK. The CamPaIGN study. Litvan, I., Aarsland, D., Adler, C.H. (2011). MDS task force
Brain, 127 (Pt 3): 550–560. on mild cognitive impairment in Parkinson’s disease:
Gomperts, S.N., Locascio, J.L., Rentz, D. et al. (2013). Critical review of PD‐MCI. Movement Disorders, 26
Amyloid is linked to cognitive decline in patients with (10): 1814–1824.
Parkinson disease without dementia. Neurology, 80 (1): Litvan, I., Goldman, J.G., Tröster, A.I. et al. (2012).
85–91. Diagnostic criteria for mild cognitive impairment in
Goris, A., Williams-Gray, C.H., Clark, G.R. et al. (2007). Tau Parkinson’s disease: Movement Disorder Society
and alpha-synuclein in susceptibility to, and dementia Task Force guidelines. Movement Disorders, 27 (3):
in, Parkinson’s disease. Annals of Neurology, 62 (2): 349–356.
145–153. Litvan, I., MacIntyre, A., Goetz, C.G. et al. (1998). Accuracy
Halliday, G., Hely, M., Reid, W. and Morris, J. (2008). of the clinical diagnoses of Lewy body disease,
The progression of pathology in longitudinally fol- Parkinson disease, and dementia with Lewy bodies:
lowed patients with Parkinson’s disease. Acta A clinicopathologic study. Archives of Neurology, 55:
Neuropathologica, 115: 409–415. 969–978.
746 Dementia

Maetzler, W., Liepelt, I., Reimold, M. et al. (2009). Cortical Rektorova, I., Krajcovicova, L., Marecek, R. and Mikl, M.
PIB binding in Lewy body disease is associated (2012). Default mode network and extrastriate visual
with Alzheimer-like characteristics. Neurobiology of resting state network in patients with Parkinson’s disease
Disease, 34 (1): 107–112. dementia. Neurodegenerative Diseases, 10 (1): 232.
Marder, K., Tang, M.X., Cote, L. et al. (1995). The fre- Rinne, J.O., Portin, R., Ruottinen, H. et al. (2000). Cognitive
quency and associated risk factors for dementia impairment and the brain dopaminergic system in
in patients with Parkinson’s disease. Archives of Parkinson disease: [18F]fluorodopa positron emission
Neurology, 52: 695–701. tomographic study. Archives of Neurology, 57: 470–475.
Mata, I.F., Leverenz, J.B., Weintraub, D. et al. (2014). APOE, Rocha, F.L., Murad, M.G., Stumpf, B.P. et al. (2013).
MAPT, and SNCA genes and cognitive performance in Antidepressants for depression in Parkinson’s dis-
Parkinson disease. JAMA Neurology, 71 (11): 1405–1412. ease: Systematic review and meta-analysis. Journal of
Mattay, V.S., Tessitore, A., Callicott, J.H. et al. (2002). Psychopharmacology, 27 (5): 417–423.
Dopaminergic modulation of cortical function in Rolinski, M., Fox, C., Maidment, I. and McShane, R. (2012).
patients with Parkinson’s disease. Annals of Neurology, Cholinesterase inhibitors for dementia with Lewy bod-
51: 156–164. ies, Parkinson’s disease dementia and cognitive impair-
Matsui, H., Nishinaka, K., Oda, M. et al. (2007). Dementia ment in Parkinson’s disease. The Cochrane Database
in Parkinson’s disease: Diffusion tensor imaging. Acta of Systematic Reviews, (3): CD006504.
Neurologica Scandinavica, 116 (3): 177–181. Siderowf, A., Xie, S.X., Hurtig, H. et al. (2010). CSF amy-
McKeith, I.G., Dickson, D.W., Lowe, J. et al. (2005). loid β 1-42 predicts cognitive decline in Parkinson
Diagnosis and management of dementia with disease. Neurology, 75 (12): 1055–1061.
Lewy bodies: Third report of the DLB Consortium. Summerfield, C., Junqué, C., Tolosa, E. et al. (2005).
Consortium on DLB. Neurology, 65 (12): 1863–1872. Structural brain changes in Parkinson disease with
Melzer, T.R., Watts, R., MacAskill, M.R. et al. (2012). Grey dementia: A voxel-based morphometry study. Archives
matter atrophy in cognitively impaired Parkinson’s of Neurology, 62 (2): 281–285.
disease. Journal of Neurology, Neurosurgery and Svenningsson, P., Westman, E., Ballard, C. and Aarsland,
Psychiatry, 83 (2): 188–194. D. (2012). Cognitive impairment in patients with
Melzer, T.R., Watts, R., MacAskill, M.R. et al. (2013). White Parkinson’s disease: Diagnosis, biomarkers, and treat-
matter microstructure deteriorates across cogni- ment. The Lancet Neurology, 11 (8): 697–707.
tive stages in Parkinson disease. Neurology, 80 (20): Tiraboschi, R., Hansen, L.A., Alford, M. et al. (2000).
1841–1849. Cholinergic dysfunction in diseases with Lewy bodies.
Mollenhauer, B., Rochester, L., Chen‐Plotkin, A. and Neurology, 54: 407–411.
Brooks, D. (2014). What can biomarkers tell us about Wang, H.-F., Yu, J.T., Tang, S.-W. et al. (2014). Efficacy
cognition in Parkinson’s disease? Movement Disorders, and safety of cholinesterase inhibitors and meman-
29 (5): 622–633. tine in cognitive impairment in Parkinson’s disease,
Pedersen, K.F., Larsen, J.P., Tysnes, O.B. and Alves, G. Parkinson’s disease dementia, and dementia with
(2013). Prognosis of mild cognitive impairment in early Lewy bodies: Systematic review with meta-analysis
Parkinson disease: The Norwegian ParkWest study. and trial sequential analysis. Journal of Neurology,
JAMA Neurology, 70 (5): 580–586. Neurosurgery and Psychiatry, 86 (2): 135–143.
Pereira, J.B., Svenningsson, P., Weintraub, D. et al. (2014). Weintraub, D., Moberg, P.J., Culbertson, W.C. et al.
Initial cognitive decline is associated with cortical thin- (2004). Evidence for impaired encoding and retrieval
ning in early Parkinson disease. Neurology, 82 (22): memory profiles in Parkinson disease. Cognitive and
2017–2025. Behavioral Neurology, 17 (4): 195–200.
Perry, E.K., Irving, D., Kerwin, J.M. et al. (1993). Weintraub, D., Simuni, T., Caspell‐Garcia, C. et al.
Cholinergic transmitter and neurotrophic activities (2015). Cognitive performance and neuropsychiatric
in Lewy body dementia: Similarity to Parkinson’s and symptoms in early, untreated Parkinson’s disease.
distinction from Alzheimer disease. Alzheimer Disease Movement Disorders, 30 (7): 919–927.
and Associated Disorders, 7: 69–79. Willemssen, R., Muller, T., Schwarz, M. et al. (2009).
Poewe, W., Wolters, E., Emre, M. et al. (2006). Long-term Response monitoring in de novo patients with
benefits of rivastigmine in dementia associated with Parkinson’s disease. PLoS ONE, 4 (3): e4898.
Parkinson’s disease: An active treatment extension Williams-Gray, C.H., Foltynie, T., Brayne, C.E. et al. (2007).
study. Movement Disorders, 21 (4): 456–461. Evolution of cognitive dysfunction in an incident
Possin, K.L., Filoteo, J.V., Song, D.D. and Salmon, D.P. Parkinson’s disease cohort. Brain, 130 (Pt 7): 1787–1798.
(2008). Spatial and object working memory deficits in Williams-Gray, C.H., Goris, A., Saiki, M. et al. (2009).
Parkinson’s disease are due to impairment in differ- Apolipoprotein E genotype as a risk factor for suscep-
ent underlying processes. Neuropsychology, 22 (5): tibility to and dementia in Parkinson’s disease. Journal
585–595. of Neurology, 256 (3): 493–498.
Cognitive impairment and dementia in Parkinson’s disease 747

Williams-Gray, C.H., Hampshire, A., Barker, R.A. and Zaccai, J., McCracken, C. and Brayne, C. (2005). A sys-
Owen, A.M. (2008). Attentional control in Parkinson’s tematic review of prevalence and incidence studies
disease is dependent on COMT val 158 met genotype. of dementia with Lewy bodies. Age Ageing, 34 (6):
Brain, 131 (Pt 2): 397–408. 561–566.
Wylie, S.A., Stout, J.C. and Bashore, T.R. (2005). Activation Zarei, M., Ibarretxe-Bilbao, N., Compta, Y. et al. (2013).
of conflicting responses in Parkinson’s disease: Evidence Cortical thinning is associated with disease stages and
for degrading and facilitating effects on response time. dementia in Parkinson’s disease. Journal of Neurology,
Neuropsychologia, 43 (7): 1033–1043. Neurosurgery and Psychiatry, 84 (8): 875–882.
Part     7
Focal Dementias and Other
Neuropsychiatric Syndromes Involving
Cognitive Decline

65 Frontotemporal dementia 749


Peter J. Nestor
66 Pick’s disease: Its relationship to progressive aphasia, semantic dementia and frontotemporal dementia 759
John Hodges
67 The genetics and molecular pathology of frontotemporal lobar degeneration 771
David M.A. Mann
68 Semantic dementia 783
Julie S. Snowden and Matthew Jones
69 Primary progressive aphasia and posterior cortical atrophy 795
Jonathan D. Rohrer, Sebastian J. Crutch and Jason D. Warren
70 The cerebellum and cognitive impairment 812
Elsdon Storey and Evelyn A. Lindsay
71 Delirium: A clinical overview 823
Andrew Teodorczuk and Daniel Davis
72 Depression with cognitive impairment 832
Sarah Shizuko Morimoto, Genevieve S. Yuen, Stephen Beres and George S. Alexopoulos
73 Schizophrenia, cognitive impairment and dementia 846
Flavie Waters, Andrew Ford and Osvaldo P. Almeida
74 Dementia in intellectual disabilities 852
Jennifer Torr
75 Alcohol-related dementia and Wernicke–Korsakoff syndrome 857
Stephen C. Bowden and Simon J. Scalzo
76 Huntington’s disease 868
Phyllis Chua, Samantha Loi and Edmond Chiu
77 Uncommon dementias (including the prion diseases) 885
Matthew Jones and David Neary
65
Frontotemporal dementia

PETER J. NESTOR

in which case the behavioural form is typically referred to


65.1 INTRODUCTION as ‘behavioural variant FTD’ (bvFTD). This last term was
used in the more recent diagnostic guidelines proposed by
Clinical understanding of frontotemporal dementia Rascovsky et al. (2011) that unlike the earlier Neary criteria
(FTD) has improved immensely over recent decades. focused exclusively on the behavioural presentation (Table
For instance, the entry in the 1983 edition of the Oxford 65.1) rather than FTLD as a whole. Other terms for the clini-
Textbook of Psychiatry for Pick’s Disease, one of the major cal syndrome that have been used in the past include ‘fron-
pathologies underpinning FTD, stated, ‘There are no tal variant FTD’ or ‘dementia of frontal type’. For simplicity,
specific clinical features to separate Pick’s disease from this chapter will use the term ‘FTD’.
Alzheimer’s, and the distinction is generally made at FTLD is unique among the neurodegenerative dementias
autopsy not in life’. In contrast to what is now known, in that, as already shown, it is not a single clinical entity. It is
this statement could barely be further from the truth but also, however, not a single pathological entity. The unifying
highlights that if one applies the simplistic algorithm of feature of this group of clinical syndromes and pathologies
noting progressive decline; falling below a cut-off on some is that they are degenerative and that they have a predilec-
global measure of cognition and excluding non-degener- tion for the frontal and rostral temporal lobes. It is also now
ative pathologies with a brain scan, then all degenerative well recognized that FTD can coexist with amyotrophic lat-
dementias can seem similar. eral sclerosis (ALS) – people initially diagnosed with ALS
A seminal step in putting FTD on the map as a clini- can later develop FTD and vice versa or the two syndromes
cal diagnosis – rather than just a spectrum of pathological can occur contemporaneously. Furthermore, in some of the
diagnoses – was the publication of the Lund–Manchester genetic forms, one can find cases with ALS and others with
criteria (1994) that employed the term ‘frontotemporal FTD in the same kindred.
dementia’. Neary et al. (1998) subsequently revised these cri-
teria proposing frontotemporal lobar degeneration (FTLD)
as the umbrella term for patients presenting with any of
the recognized clinical variants. According to this system,
65.2 DEMOGRAPHICS
those with a predominantly neuropsychiatric syndrome
characterized by behavioural change were labelled ‘fron- The age of onset of FTD typically ranges from late forties to
totemporal dementia’; the other two variants in this clas- early sixties with a mean in the late fifties (e.g. Rascovsky
sification that affect language and conceptual knowledge et al., 2011). Prevalence estimates suggest rates in the order
were labelled ‘progressive non-fluent aphasia’ and ‘semantic of 10–20/100,000 (Onyike and Diehl-Schmid, 2013). It is
dementia’, respectively – these two categories are covered in likely to be the second commonest cause of degenerative
Chapters 68 and 69. dementia under the age of 65 years after Alzheimer’s dis-
It should be noted, however, that ‘FTD’ as a label is ease (AD). Survival estimates suggest a median of around
used inconsistently; some follow the Neary (1998) scheme 10 years from symptom onset (Roberson et al., 2005;
in which FTLD is the umbrella term for the group of dis- Nunnemann et al., 2011) though this may be significantly
eases and ‘FTD’ is the syndromic subgroup with behav- shortened with comorbid ALS. Males and females appear to
ioural changes while other use ‘FTD’ as the umbrella term be affected equally.

749
750 Dementia

Table 65.1 International consensus criteria for the behavioural form of FTD.
I. Neurodegenerative (must be present for a diagnosis of FTD):
A. Shows progressive decline of behaviour and/or cognition by observation or history (provided by a knowledgeable
informant)
II. Possible FTD (Three of the following must be present; symptoms must be persistent or recurring not single or rare events):
A. Early behavioural disinhibition (one of following must be present):
A.1. Socially inappropriate behaviour
A.2. Loss of manners or decorum
A.3. Impulsive, rash or careless actions
B. Early apathy or inertia (one of the following must be present):
B.1. Apathy
B.2. Inertia
C. Early loss of sympathy or empathy (one of the following must be present):
C.1. Diminished response to other people’s needs and feelings
C.2. Diminished social interest, interrelatedness or personal warmth
D. Early perseverative, stereotyped or compulsive behaviour (one of the following must be present):
D.1. Simple repetitive movements
D.2. Complex, compulsive or ritualistic behaviours
D.3. Stereotypy of speech
E. Hyperorality and dietary changes (one of the following must be present):
   E.1. Altered food preferences
E.2. Binge eating, increased consumption of alcohol or cigarettes
E.3. Oral exploration or consumption of inedible objects
F. Neuropsychological profile: executive/generation deficits with relative sparing of memory and visuospatial functions
(all must be present):
F.1. Deficits on executive tasks
F.2. Relative sparing of episodic memory
F.3. Relative sparing of visuospatial skills
III. Probable FTD (all of the following must be present):
A. Meets criteria for possible FTD
B. Exhibits significant functional decline (by caregiver report or as evidenced by Clinical Dementia Rating Scale or
Functional Activities Questionnaire scores)
C. Imaging results consistent with FTD
C.1. Frontal and/or temporal lobe atrophy on MRI or CT
C.2. Frontal and/or temporal lobe hypometabolism (PET) or hypoperfusion (SPECT)
IV. FTD with definite FTLD pathology
A. Meets criteria for possible or probable FTD
B. Histopathological evidence of FTLD on biopsy or at necropsy
C. Presence of a known pathogenic mutation
V. Exclusion criteria for FTD (A and B must be answered negatively for an FTD diagnosis. C can be positive for possible
FTD but must be negative for probable FTD):
A. Pattern of deficits better explained by other non-degenerative nervous system or medical disorder
B. Behavioural disturbance is better accounted for by a psychiatric diagnosis
C. Biomarkers strongly indicative of Alzheimer’s disease or other neurodegenerative process
Source: Rascovsky, K. et al., Brain, 134, 2456–2477, 2011.
Notes:  Note the emphasis on ‘early’ emergence of clinical features in Section II. This separates the behavioural features of FTD from those
that can occur late in the course of other degenerative dementias; the authors proposed within the first 3 years to denote ‘early’.
Abbreviations: FTD, frontotemporal dementia; MRI, magnetic resonance imaging; CT, computed tomography; PET, positron emission
tomography; SPECT, single-photon emission computed tomography; FTLD, frontotemporal lobar degeneration.

prominent early features. This contrasts to AD, for instance,


65.3 CLINICAL FEATURES in which behavioural symptoms occur typically later in the
course of the illness while the early clinical course is domi-
The hallmark of FTD is change in behaviour with neglect of nated by cognitive difficulties.
personal care. While such changes can accompany any form Behavioural changes manifest as being self-centred
of degenerative dementia, the key is that in FTD they are and mentally rigid. Disinhibition is a prominent feature
Frontotemporal dementia 751

manifesting with such things as being overfamiliar with


strangers including sharing very personal information. 65.4 EXAMINATION
It can also include making embarrassing remarks such as
about the weight or appearance of others or comments of Careful observation of the patient’s behaviour during the
a sexual nature. While sexually disinhibited comments are consultation will typically identify abnormalities. For
not uncommon, it is notable that patients typically show instance, fatuous giggling, use of stereotypic catch phrases
a lack of libido and typically do not engage in sexual acts. or echolalia; signs of disinhibition such as kissing staff
Disinhibition in women with FTD can, however, make members or not respecting the personal space of others;
them vulnerable to sexual advances from strange men. environment-bound behaviour such as attention shifting
Loss of empathy and failure to understand the emotional to events outside a window in the street or looking over
states of others is typical; spouses often report, for instance, the examiner’s shoulder at the computer monitor; impul-
that the patient appeared emotionally unaffected by a fam- siveness such as losing patience with the consultation and
ily bereavement. In the case of serious illness in a spouse, leaving the room to pace the corridor or insisting that it is
the patient may appear more concerned about the impact on time to go home. Conversely, repeatedly returning to the
themselves, for instance, getting agitated about not having room when the informant interview is taking place is also
their meals prepared. common.
Stereotypic behaviours are characteristic with the patient The general neurological examination is often unre-
developing very fixed routines such as walking precisely markable, particularly in the early stages of the illness.
the same route at the same time of day; clock-watching, Primitive reflexes such as grasp and palmo-mental reflexes
for instance, expecting to eat meals at precisely the same are often present. To this end, it is worth noting that the lat-
time each day; hoarding useless items; checking repeatedly ter is frequently criticized for the lack of specificity because
that doors and windows are locked and so on. Stereotipes it is often an incidental finding in the elderly. This is not the
in speech are also common, such as the use of repetitive case in middle age, however, so should be taken seriously as
catchphrases in conversation. Becoming fixated on eating a sign of frontal pathology with patients in their fifties or
the same types of foods is also very common. Often a prefer- younger.
ence for sweet foods emerges that can be extreme and lead Because of the association of FTD with ALS, a careful
to considerable weight gain. Excessive consumption of alco- motor examination looking for features of the latter such
hol may also occur and can make care considerably more as wasting, fasciculation and brisk tendon reflexes should
difficult. The manner of eating is also often disturbed with always be undertaken. Extra-pyramidal signs of rigidity
patients eating very quickly and overfilling their mouths; and akinesia may be present, though they are usually a fairly
having very poor table manners; taking food from the plates late feature.
of others and so on. It is not uncommon to hear relatives
complain of ‘pouching’ – i.e. overfilling the mouth such that
the cheeks bulge out like those of a hamster eating.
65.5 NEUROPSYCHOLOGY/COGNITIVE
Apathy is possibly the most frequently reported symp-
tom, although from a diagnostic perspective it is prob-
PROFILE
ably the least helpful in that it is a prominent feature of all
forms of degenerative dementia as well as in depression. The A dysexecutive syndrome is often, albeit erroneously, con-
behavioural features that are most specific for FTD tend to sidered the hallmark of FTD. Although it forms an axis in
be stereotypic behaviours and change in food preference the diagnostic criteria proposed by Rascovsky et al. (2011),
(Bozeat et al., 2000). it is notable that in applying these criteria, one can reach a
Patients characteristically lack insight into their illness diagnosis of possible, probable or even definite FTD without
meaning that in order to obtain a full picture of the behav- this neuropsychological profile; it is not a mandatory fea-
ioural disturbance, an informant’s account, from one who ture (Table 65.1). There has possibly been some backwards
lives with the patient if at all possible, is mandatory. It is also reasoning in the belief of the primacy of a dysexecutive syn-
desirable that the informant interview takes place without drome in FTD – i.e. there is frontal degeneration, therefore,
the patient – for instance while the patient has their cogni- they must be dysexecutive. Empirical evidence for a focal
tive examination – as many find giving a full picture of the dysexecutive syndrome in FTD is, however, rather hard to
manifestations of the behavioural disturbance embarrass- find. Typically, studies looking at comparisons between AD
ing in the patient’s presence. and FTD struggle to find significant differences, even at a
Psychotic features (delusions and hallucinations) are suf- group level, let alone changes that are robust enough to sup-
ficiently unusual in FTD to be a prompt to consider other port an individual diagnosis.
diagnoses. They do, however, occur in a minority of cases Nevertheless, it has been proposed (Hornberger, Piguet
and if present, are typically associated with ubiquitin- et al., 2010) that the best neuropsychological tests of executive
staining pathological inclusions such as FTD-ALS, C9orf72 function for discriminating FTD from AD are reverse digit
mutations or fused-in sarcoma protein (FUS) pathology span, letter fluency (generating as many words as possible
(Sections 65.8 and 65.9). in a minute beginning with a specific letter) and the Hayling
752 Dementia

test in which subjects are presented with a sentence that has features themselves – this scenario has often been referred
the last word missing; this missing word is strongly implied to as the FTD phenocopy syndrome.
from the rest of the sentence, e.g. ‘Last night I watched an As already discussed, one can only appreciate the full
interesting film on ____’ and one has to suppress the salient range of behavioural changes from an informant’s testi-
urge to say ‘television’ and instead supply a word that does not mony. If one relies exclusively on the informant’s account,
make sense. however, problems can occur. There is potential for an infor-
Memory is often assumed to be relatively preserved in mant to be led by probing to zealously for specific behav-
FTD when compared with AD, though again, empirical iours; particular danger arises where the diagnosis has been
evidence indicates that this is not necessarily the case in suggested by a previous medical assessment and the infor-
that FTD patients can display comparable levels of mem- mant, seeking information from the internet, unwittingly
ory impairment to AD (Hornberger, Savage et al., 2010). fills in the blanks after reading the clinical features – for
Memory impairment can even be the prominent early instance, over-interpreting a request for a second slice of
­clinical feature, although, in such circumstances, the char- birthday cake as sustained emergence of a sweet food prefer-
acteristic behavioural profile may emerge later (Graham ence and so on. Although these issues can exaggerate symp-
et al., 2005). toms to give a false impression of FTD, they cannot explain
Similar cognitive test scores between FTD and other forms what brings such patients to medical attention in the first
of dementia do not necessarily imply a similar neural sub- place, and therefore, what their actual diagnosis might be.
strate for the impairment – for instance, one can score badly Patients with an apparent phenocopy of FTD are typically
on a cognitive test simply by not engaging in the task. One male, have good cognitive performance, normal imaging
often sees a rather patchy pattern of deficits across a range of and remain stable over time (Kipps et al., 2009). It appears
cognitive domains in FTD and one is left with the impression that the answer is likely to be multifactorial with person-
that a lack of interest on the part of the patient is playing a ality traits, relationship problems and life events being the
role. As with the clinical examination, it is the behaviour of most important factors (Gossink et al., 2016).
the patient during testing, rather than the scores themselves, To ensure that a false-positive diagnosis is not made, one
that often points to FTD: answering impulsively without should consider the informant-based symptom profile as
appearing to be particularly engaged in the testing; becom- mandatory but not rely on it exclusively. It must be supple-
ing fixated on some tests or arbitrarily deciding to give up mented by empirical evidence – does the patient behave like
for no apparent reason. In fact, distractibility means that it someone with FTD in the consulting room (Section 65.4)?
is not uncommon for FTD patients to be untestable because Do they have imaging features of FTD (Section 65.7.1)? Is
the simply fail to engage in any formal testing. there evidence of impairment on cognitive testing, accept-
The reason for stressing the rather messy and haphaz- ing the caveat that this will typically not take the form of
ard approach that many FTD patients exhibit in neuropsy- an isolated dysexecutive syndrome (Section 65.5)? One
chological testing is that in this author’s experience, one further caveat is that patients with the C9orf72 mutation
of the commonest reasons for an incorrect diagnosis of (Section 65.9.1) can have a very indolent disease with unre-
FTD is the belief that FTD is diagnosed on the basis of a markable imaging (Devenney et al., 2014). Also, as with all
focal dysexecutive syndrome on neuropsychological test- forms of degenerative dementia, sometimes it can be diffi-
ing. The problem is that so-called executive tasks, although cult to decide in the early stages and, in such circumstances,
weighted to the frontal lobes compared with other tests, ­evidence of progression with longitudinal follow-up is the
really require the whole brain to be functioning. As such, key – to this end, evidence of progressive decline on cogni-
non-specific neurological insults typically show up with tive testing is particularly useful.
impairments on executive tasks. In this regard, a miscel-
lany of neurological disorders can cause an apparent dys-
executive test pattern. Examples of referral to our clinic
with a previous incorrect FTD diagnosis include epilepsy, 65.7 INVESTIGATIONS
multiple sclerosis, depression and head injury – all having
been misdiagnosed as FTD on neuropsychological grounds 65.7.1 IMAGING
in spite of a complete absence of any of the typical behav-
ioural features. Structural and nuclear imaging can be particularly ­helpful
in FTD (Varma et al., 2002). A pattern of d ­ isproportionate
frontal and rostal temporal lobe atrophy has diagnostic pre-
dictive value for FTLD in general, t­ ypically with the empha-
65.6 FALSE-POSITIVE DIAGNOSES sis being on the frontal lobes in FTD. Atrophy is often gross
and unmistakable, but even in more subtle degrees, a clear
In addition to the problem of misdiagnosing FTD on the pattern of disproportionate frontal atrophy – compared
basis of neuropsychological performance without consider- with occipito-parietal regions – is evident (Figure 65.1).
ing the patient’s behavioural features, there is also the risk Magnetic resonance imaging is the preferred modality but
of misdiagnosing FTD on the basis of apparent behavioural computerized tomography is often adequate.
Frontotemporal dementia 753

Figure 65.1 Magnetic resonance imaging (MRI) scan in frontotemporal dementia (FTD) showing frontal atrophy with
relative preservation of posterior regions. On the sagittal image, this gradient is clearly seen with the widening of sulci
rostrally (i.e. more cerebrospinal fluid [CSF]) that is not evident in occipital lobe and precuneus. Note also the patient had
hippocampal atrophy (arrows) – this is quite typical in FTD and highlights that a cursory inspection of the medial temporal
lobes in isolation could mislead the viewer to think of Alzheimer’s disease.

In cases where the structural imaging findings are equiv- cortical layers (see Chapter 67 for an in-depth discussion).
ocal, nuclear imaging with fluorodeoxyglucose positron Histopathologically, FTD is associated with either tau-pos-
emission tomography (FDG-PET) will often show meta- itive inclusions or inclusions staining with ubiquitin. These
bolic changes in the frontal lobes (Figure 65.2). Cerebral two histopathologies occur with approximately equal fre-
perfusion imaging with hexamethylpropyleneamineoxime quency in FTD (e.g. Rohrer et al., 2011); in sporadic disease,
(HMPAO)-single-photon emission computed tomography it is usually impossible to predict whether a patient will have
(SPECT) can also be useful in this setting though it is less tau or ubiquinated inclusions in life. The key exception to
sensitive than to FDG-PET. this is FTD with clinically manifesting ALS where pathol-
Completely normal imaging, particularly a normal FDG- ogy is always tau-negative/ubiquitin positive.
PET, should make one very cautious about diagnosing FTD. Tau-positive (FTLD-tau) pathology can be subdivided
by the number of microtubule-binding domains in the pro-
65.7.2 MISCELLANEOUS INVESTIGATIONS tein into three-repeat (3R) or four-repeat (4R) pathologies.
The major forms of FTLD-tau include Pick’s disease (3R)
It has been known for many years that visual reading of (Chapter 66), those with tau mutations (3R and 4R) and,
electroencephalography (EEG) is essentially unremarkable rarely, argyrophilic grain disease (4R). The tauopathies, cor-
in FTD (e.g. Neary et al., 1988) – in particular, there is per- ticobasal degeneration and progressive supranuclear palsy
severation of the occipital alpha rhythm. EEG still occasion- pathology (both 4R) can also be found occasionally in cases
ally has a role, in that, suspected FTD with an abnormal that were diagnosed as FTD in life, though these patholo-
EEG should prompt a search for other causes (Figure 65.3). gies are more commonly noted with non-fluent aphasic pre-
Electromyography is often indicated to exclude/confirm sentations of FTLD (Josephs et al., 2011).
denervation related to ALS. Cerebrospinal fluid, although FTD with ubiquitin-staining inclusions was typically
now a routine investigation in many places as a marker of referred to as FTLD-U until the substrate to which the
Alzheimer pathology, has not yet become a standard clini- stain bound was identified. It was subsequently discov-
cal investigation to support a positive diagnosis of FTD. ered that, in the vast majority, this was a transactive
Genetic testing is often highly relevant in FTD; the known response DNA-binding protein 43 (TDP-43) pathol-
mutations are discussed further in Section 65.9. ogy (Neumann et al., 2006). TDP-43 pathology (FTLD-
TDP) is, in turn, divided into subtypes (A–D) based on
the histological pattern (Mackenzie et al., 2011), although
65.8 PATHOLOGY interrater agreement for subtype classification can be
somewhat variable (Alafuzoff et al., 2015). Clinical FTD
Atrophy is most evident in frontal and rostral temporal is mostly associated with types A and B, the latter being
lobes with neuronal loss predominantly in the superficial typical of FTD-ALS. Type D is specifically designated for
754 Dementia

Figure 65.2 (See colour insert.) Fluorodeoxyglucose positron emission tomography (FDG-PET) scan from a patient with
FTD whose structural imaging did not show convincing evidence of disproportionate frontal atrophy. Note the clear evi-
dence of asymmetric frontal hypometabolism (arrows).

(a)

(b)

(c)

Figure 65.3 (See colour insert.) Use of electroencephalography (EEG) in the investigation of suspected FTD. This 63-year-
old male presented with marked changes in personality and comportment, typical of those seen in FTD, that had evolved
over the preceding 8 months. FDG-PET, shown as a parametric map of hypometabolism (a), revealed a marked prefrontal
hypometabolism. His EEG was grossly abnormal with a considerable delta-wave activity (b). This is not compatible with
FTD. His diagnosis was presumed to be an autoimmune encephalitis, and after treatment with high-dose methylpredniso-
lone both his FTD-like behavior and EEG (c) improved dramatically.
Frontotemporal dementia 755

the pathology of Valosin-containing protein (VCP) gene for dementia that is not necessarily relevant to FTD. On the
mutations (Section 65.9.6). other hand, if FTD was present in previous generations, it is
Following the discovery of TDP-43, it was noted that unlikely to have been diagnosed as such. Enquiring specifi-
not all FTLD-U cases had this pathology. A minority that cally about the age of onset and symptoms, if known, can be
were negative for TDP-43 were designated atypical FTLD-U particularly useful pointers to suggest a forebear had FTD.
(aFTLD-U) and were found subsequently to have FUS Of the current known genes, the chromosome 9 open
pathology (Neumann et al., 2009). In a pathological series reading frame 72 (C9orf72) mutation is probably the most
of FTD, it has been estimated that about 5% have FUS and prevalent cause of familial FTD. Tau and progranulin (PGRN)
in a clinical series of FTD possibly around 3% (Snowden mutations are also important causes though there appears,
et al., 2011). FUS pathology, in turn, also has two further from reported series, to be considerably more geographical
pathological variants – neuronal intermediate filament variability in their occurrence. Several further gene muta-
inclusion disease (NIFID) and basophilic inclusion body tions, namely TAR DNA-binding protein (TARDBP), VCP
disease (BIBD) both of which can be associated with FTD. and CHMP2B, have been associated with FTD but appear
Collectively, the FUS-related pathologies are designated to be very rare. The pathology of tau mutations is obviously
FTLD-FUS (Mackenzie et al., 2010). Aside from FTLD-TDP FTLD-tau, all of the remainder have FTLD-TDP with the
and FTLD-FUS, there remain rare cases with ubiquitin- exception of CHMP2B that is FTLD-UPS (see Section 65.8).
binding pathology that has neither TDP-43 nor FUS as its Several studies have looked for differences between the
substrate; these are designated FTLD-ubiquitin proteasome genetic forms of FTD in clinical profile, imaging and so on.
system (FTLD-UPS) (Mackenzie et al., 2010). The pathology The following list highlights some of the key clinical features
associated with the charged multivesicular body protein 2B to consider as ‘red flags’ that relate to specific mutations.
(CHMP2B) mutation (Section 65.9.7) is an example of this.
Finally, cases may exhibit frontotemporal neurode- 65.9.1 C9orf72 (CHROMOSOME 9)
generation with no specific histological inclusions. This
is referred to as dementia lacking distinctive histology This hexanucleotide repeat expansion (DeJesus-Hernandez
(DLDH) and was more commonly reported in the era before et al., 2011; Renton et al., 2011) is also associated with ALS.
routine staining for ubiquitin inclusions – in other words, ALS can coexist with FTD in the same patient or can exist
the majority of such cases in the historical literature likely in the kindred simply as ALS. The presence of ALS in the
had a form of FTLD-U but without the appropriate stains to kindred of a patient with FTD makes this mutation highly
identify them as such. Nevertheless, there probably remains likely. Another feature of this mutation is that although cases
a small number that have DLDH (Cairns et al., 2007). typically display the poor prognosis of FTD and ALS, reports
are emerging of very slowly progressive cases with disease
duration exceeding two decades (e.g. Suhonen et al., 2015).
65.9 GENETICS
65.9.2 TAU (CHROMOSOME 17)
Compared with other degenerative dementias, there is an When this mutation was first discovered, emphasis was
increased likelihood of monogenetic mutations causing placed on the coexistence of FTD with parkinsonism
FTD. That said, it is difficult to put a precise figure on genetic (FTDP-17) (Hutton et al., 1998). Parkinsonism, particularly
prevalence because most estimates have come from regional occurring later in the disease course, is not uncommon in
samples, often from a single centre; there is quite possibly also clinical FTD in general and so is probably not particularly
a reporting bias in that regions with a few genetic cases might helpful in making a specific pathologic or genetic prediction.
be less likely to publish prevalence data. Estimates of preva- More relevant to clinical dementia differential diagnosis,
lence typically are of the order of 20%–30%. Notably, the however, is that tau mutations can present with prominent
FTD phenotype, as opposed to the language presentations of memory disturbance and profound hippocampal atrophy
FTLD, is most likely to occur with an autosomal-dominant making it easy to mistake this mutation for AD.
mutation (Rohrer et al., 2009). Given that FTD is relatively
less common compared with AD and that only a minority 65.9.3 PGRN (CHROMOSOME 17)
of cases appear to have autosomal-dominant disease, even
a single large kindred in a clinical catchment area can skew These mutations (Baker et al., 2006; Cruts et al., 2006) are
estimates of genetic prevalence. In contrast, it is often the case typically not associated with a sufficiently unique pheno-
that an individual clinic may have very few or even no genetic type to separate it from FTD in general. Visual hallucina-
cases based on the genetic epidemiology of the region. tions have been reported in up to 25% of cases (Le Ber et al.,
Genetically determined disease should always be consid- 2008), so if present, these offer a possible clue being so rare
ered when assessing a patient with FTD, and, obviously, the in FTD otherwise. Marked hemispheric asymmetry of atro-
greatest clue is family history. To this end, it is important to phy pattern has also been emphasized in some studies (Le
consider that with late-onset dementia – of any type – being so Ber et al., 2008; Rohrer et al., 2010), though this is, arguably,
common, many will have an apparently positive family history not uncommon in FTD in general.
756 Dementia

65.9.4 FUS (CHROMOSOME 16) were entirely negative (Boxer et al., 2013). Although
­placebo-controlled trial evidence is largely lacking, selec-
These mutations have been identified in ALS without tive serotonin reuptake inhibitor (SSRI) medications are
dementia (Vance et al., 2009), convincing evidence that often prescribed to deal with disturbing behaviours. In a
such mutations can also cause FTD remains to be estab- small randomized, placebo-controlled trial, trazodone was
lished. Nevertheless, it is included here because FUS pathol- associated with some improvements in behaviour (Lebert
ogy (aFTLD-U) – without a FUS gene mutation – is seen in et al., 2004), while another using paroxetine showed no
FTD and has some distinctive clinical features (Neumann benefit (Deakin et al., 2004). Anecdotally, low-dose topira-
et al., 2009). This pathology is associated with a very young mate may be ­useful to ameliorate abnormal eating behav-
onset FTD, typically in the thirties and forties, markedly iour (Nestor, 2012).
ritualistic/stereotypic behaviour and a relatively high preva-
lence of psychotic delusions and hallucinations (MacKenzie
et al., 2008; Snowden et al., 2011).
65.11 CONCLUSIONS
65.9.5 TARDBP (CHROMOSOME 1)
FTD is an important form of dementia in, predominantly,
These mutations, first identified in ALS (Sreedharan et al., middle age. The massive impact it has on personality and
2008), can be associated with ALS/FTD (e.g. Chiò et al., behaviour make it, arguably, the most distressing of the
2010), though from the number of reported cases this degenerative dementias in terms of caregiver burden. The
appears to be a rare cause of FTD. In addition to FTD, lan- relative youth of patients combined with the non-cognitive
guage disorders have been reported though it appears that disturbances of FTD are a major challenge to support ser-
behavioural disturbance is the norm (Floris et al., 2015). vices that are typically geared to older patients and cogni-
tive manifestations.
65.9.6 VCP (CHROMOSOME 9)
Mutations in this gene give rise to a phenotype of FTD with REFERENCES
inclusion body myopathy and Paget’s bone disease (Watts
et al., 2004). Alafuzoff, I., Pikkarainen, M., Neumann, M. et al. (2015).
Neuropathological assessments of the pathology
65.9.7 CHMP2B in frontotemporal lobar degeneration with TDP43-
positive inclusions: An inter-laboratory study by
This mutation (Skibinski et al., 2005) located on chromo- the BrainNet Europe consortium. Journal of Neural
some 3, and thus known as FTD-3 prior to the gene dis- Transmission, 122: 957–972.
covery, is essentially restricted to a large kindred from the Baker, M., Mackenzie, I.R.A., Pickering-Brown, S.M. et al.
Jutland peninsula of Denmark. (2006). Mutations in progranulin cause tau-negative
frontotemporal dementia linked to chromosome 17.
Nature, 442: 916–919.
Boxer, A.L., Knopman, D.S., Kaufer, D.I. et al. (2013).
65.10 TREATMENT Memantine in patients with frontotemporal lobar
degeneration: A multicentre, randomised, double-
Treatment of FTD is supportive in that there are no evi- blind, placebo-controlled trial. Lancet Neurology, 12:
dence-based treatments that change the course of the ill- 149–156.
ness. A clinical psychologist with experience of FTD can be Bozeat, S., Gregory, C.A., Ralph, M.A. and Hodges, J.R.
an invaluable asset both to support the caregiver but also (2000). Which neuropsychiatric and behavioural
to offer behavioural strategies that minimize the burden of features distinguish frontal and temporal variants of
care. In particular, environmental manipulations aimed at frontotemporal dementia from Alzheimer’s disease?
minimizing burden of care by reducing the impact of stim- Journal of Neurology, Neurosurgery and Psychiatry,
ulus-bound behaviour can be useful. This can even include 69: 178–186.
exploiting stimulus-bound behaviour for a positive out- Cairns, N.J., Bigio, E.H., Mackenzie, I.R. et al. (2007).
come – for instance, by setting out potential activities, such Neuropathologic diagnostic and nosologic criteria for
as placing broom in the middle of a room, a patient may frontotemporal lobar degeneration: Consensus of the
start sweeping and so on. Consortium for frontotemporal lobar degeneration.
There is no evidence that the drugs licensed for the Acta Neuropathologica, 114: 5–22.
­treatment of AD, cholinesterase inhibitors and meman- Chiò, A., Calvo, A., Moglia, C. et al. (2010). Amyotrophic
tine, offer benefit in FTD. Memantine, in particular, lateral sclerosis–frontotemporal lobar dementia in
was the s­ubject of the largest placebo-controlled trial of 3 families with p.Ala382Thr TARDBP Mutations.
a symptomatic therapy in FTD to date, and the results Archives of Neurology, 67: 1002–1009.
Frontotemporal dementia 757

Cruts, M., Gijselinck, I., van der Zee, J. et al. (2006). Null Lund and Manchester groups (1994). Clinical and neuro-
mutations in progranulin cause ubiquitin-positive pathological criteria for frontotemporal dementia.
frontotemporal dementia linked to chromosome Journal of Neurology, Neurosurgery and Psychiatry,
17q21. Nature, 442: 920–924. 57: 416–418.
Deakin, J.B., Rahman, S., Nestor, P.J. et al. (2004). Mackenzie, I.R., Foti, D., Woulfe, J. and Hurwitz, T.A.
Paroxetine does not improve symptoms and impairs (2008). Atypical frontotemporal lobar degeneration
cognition in frontotemporal dementia: a double-blind with ubiquitin-positive, TDP-43-negative neuronal
randomized controlled trial. Psychopharmacology, inclusions. Brain, 131: 1282–1293.
172: 400–408. Mackenzie, I.R., Neumann, M., Baborie, A. et al. (2011).
DeJesus-Hernandez, M., Mackenzie, I.R., Boeve, B.F. et al. A harmonized classification system for FTLD-TDP
(2011) Expanded GGGGCC hexanucleotide repeat in pathology. Acta Neuropathologica, 122: 111–113.
noncoding region of C9ORF72 causes chromosome Mackenzie, I.R., Neumann, M., Bigio, E.H. et al. (2010).
9p-linked FTD and ALS. Neuron, 72: 245–256. Nomenclature and nosology for neuropathologic
Devenney, E., Hornberger, M., Irish, M. et al. (2014). subtypes of frontotemporal lobar degeneration: An
Frontotemporal dementia associated with the update. Acta Neuropathologica, 119: 1–4.
C9ORF72 mutation: A unique clinical profile. JAMA Neary, D., Snowden, J.S., Gustafson, L. et al. (1998).
Neurology, 71: 331–339. Frontotemporal lobar degeneration: A consensus on
Floris, G., Borghero, G., Cannas, A. et al. (2015). Clinical clinical diagnostic criteria. Neurology, 51: 1546–1554.
phenotypes and radiological findings in frontotempo- Neary, D., Snowden, J.S., Northen, B. and Goulding, P.
ral dementia related to TARDBP mutations. Journal of (1988). Dementia of frontal lobe type. Journal of
Neurology, 262: 375–384. Neurology, Neurosurgery and Psychiatry, 51: 353–61.
Gossink, F.T., Dols, A., Kerssens, C.J. et al. (2016). Nestor, P.J. (2012). Reversal of abnormal eating and
Psychiatric diagnoses underlying the phenocopy drinking behaviour in a frontotemporal lobar
syndrome of behavioural variant frontotemporal degeneration patient using low-dose topiramate.
dementia. Journal of Neurology, Neurosurgery and Journal of Neurology, Neurosurgery and Psychiatry,
Psychiatry, 87: 64–68. 83: 349–350.
Graham, A., Davies, R., Xuereb, J. et al. (2005). Neumann, M., Rademakers, R., Roeber, S. et al. (2009).
Pathologically proven frontotemporal dementia pre- A new subtype of frontotemporal lobar degeneration
senting with severe amnesia. Brain, 128: 597–605. with FUS pathology. Brain, 132: 2922–2931.
Hornberger, M., Piguet, O., Graham, A.J. et al. (2010). How Neumann, M., Sampathu, D.M., Kwong, L.K. et al. (2006).
preserved is episodic memory in behavioral variant Ubiquitinated TDP-43 in frontotemporal lobar degen-
frontotemporal dementia? Neurology, 74: 472–479. eration and amyotrophic lateral sclerosis. Science,
Hornberger, M., Savage, S., Hsieh, S. et al. (2010). 314: 130–133.
Orbitofrontal dysfunction discriminates behavioral Nunnemann, S., Last, D., Schuster, T. et al. (2011). Survival
variant frontotemporal dementia from Alzheimer’s in a German population with frontotemporal lobar
disease. Dementia and Geriatriatric Cognitive degeneration. Neuroepidemiology, 37: 160–165.
Disorders, 30: 547–52. Onyike, C.U. and Diehl-Schmid, J. (2013). The epidemiol-
Hutton, M., Lendon, C.L., Rizzu, P. et al. (1998). Association ogy of frontotemporal dementia. International Review
of missense and 5′-splice site mutations in tau with the of Psychiatry, 25: 130–137.
inherited dementia FTDP-17. Nature, 393: 702–705. Rascovsky, K., Hodges, J.R., Knopman, D. et al. (2011).
Josephs, K.A., Hodges, J.R., Snowden, J.S. et al. Sensitivity of revised diagnostic criteria for the behav-
(2011). Neuropathological background of pheno- ioural variant of frontotemporal dementia. Brain, 134:
typical variability in frontotemporal dementia. Acta 2456–2477.
Neuropathologica, 122: 137–153. Renton, A.E., Majounie, E., Waite, A. et al. (2011). A hexa-
Kipps, C.M., Hodges, J.R., Fryer, T.D. and Nestor, P.J. (2009). nucleotide repeat expansion in C9ORF72 is the cause
Combined magnetic resonance imaging and positron of chromosome 9p21-linked ALS-FTD. Neuron, 72:
emission tomography brain imaging in behavioural vari- 257–268.
ant frontotemporal degeneration: Refining the clinical Roberson, E.D., Hesse, J.H, Rose, K.D. et al. (2005).
phenotype. Brain, 132: 2566–2578. Frontotemporal dementia progresses to death faster
Le Ber, I., Camuzat, A., Hannequin, D. et al. (2008). than Alzheimer disease. Neurology, 65: 719–725.
Phenotype variability in progranulin mutation carriers: Rohrer, J D., Guerreiro, R., Vandrovcova, J. et al. (2009).
A clinical, neuropsychological, imaging and genetic The heritability and genetics of frontotemporal lobar
study. Brain, 131: 732–746. degeneration. Neurology, 73: 1451–1456.
Lebert, F., Stekke, W., Hasenbroekx, C. and Pasquier, F. Rohrer, J.D., Ridgway, G.R., Modat, M. et al. (2010).
(2004). Frontotemporal dementia: A randomised, Distinct profiles of brain atrophy in frontotemporal
controlled trial with trazodone. Dementia and lobar degeneration caused by progranulin and tau
Geriatric Cognitive Disorders, 17: 355–359. mutations. Neuroimage, 53: 1070–1076.
758 Dementia

Rohrer, J.D., Lashley, T., Schott, J.M. et al. (2011). Clinical degeneration caused by the C9ORF72 repeat
and neuroanatomical signatures of tissue pathology expansion: A 20-year follow-up study. Neurocase,
in frontotemporal lobar degeneration. Brain, 134: 21: 85–89.
2565–2581. Vance, C., Rogelj, B., Hortobágyi, T. et al. (2009). Mutations
Skibinski, G., Parkinson, N.J., Brown, J.M. et al. (2005). in FUS, an RNA processing protein, cause familial
Mutations in the endosomal ESCRTIII-complex sub- amyotrophic lateral sclerosis type 6. Science, 323:
unit CHMP2B in frontotemporal dementia. Nature 1208–1211.
Genetics, 37: 806–808. Varma, A.R., Adams, W., Lloyd, J.J. et al. (2002). Diagnostic
Snowden, J.S., Hu, Q., Rollinson, S. et al. (2011). The most patterns of regional atrophy on MRI and regional
common type of FTLD-FUS (aFTLD-U) is associated cerebral blood flow change on SPECT in young onset
with a distinct clinical form of frontotemporal demen- patients with Alzheimer’s disease, frontotemporal
tia but is not related to mutations in the FUS gene. dementia and vascular dementia. Acta Neurologica
Acta Neuropathologica, 122: 99–110. Scandinavica, 105(4): 261–269.
Sreedharan, J., Blair, I.P., Tripathi, V.B. et al. (2008). TDP-43 Watts, G.D., Wymer, J., Kovach, M.J. et al. (2004). Inclusion
mutations in familial and sporadic amyotrophic lateral body myopathy associated with Paget disease of
sclerosis. Science, 319: 1668–72. bone and frontotemporal dementia is caused by
Suhonen, N.M., Kaivorinne, A.L., Moilanen, V. et al. mutant valosin-containing protein. Nature Genetics,
(2015). Slowly progressive frontotemporal lobar 36: 377–81.
66
Pick’s disease: Its relationship to progressive
aphasia, semantic dementia and
frontotemporal dementia

JOHN HODGES

made (Brun et al., 1994; Neary et al., 1998; McKhann et al.,


66.1 INTRODUCTION 2001; Kertesz et al., 2003b).
The aims of this chapter are to review the evolution of
Considerable advances continue to be made in our under- the terms used to describe this spectrum of disorders, to
standing of the group of neurodegenerative diseases that highlight recent advances and examine areas of continuing
present itself with focal cognitive deficits arising from cir- controversy. While my own bias is to prefer the term PiD for
cumscribed pathology of the frontal and/or temporal lobes, this group of disorders – which is more readily understood
most commonly referred to collectively as Pick’s disease by carers and parallels our use of the label Alzheimer’s dis-
(PiD) or frontotemporal dementia (FTD). However, the lit- ease (AD) – the current tide of opinion is to use FTD as an
erature on these conditions is filled with a confusing pleth- umbrella term for the overall clinical syndrome.
ora of terms, which can make these developments difficult
to follow for the non-expert. Central to this problem is a
lack of clarity as to the intended level of description (clinical
syndrome versus clinicopathological entity versus specific 66.2 WHAT DID PICK ACTUALLY
histological diagnosis) and a lack of concordance between DESCRIBE?
these levels. For example, while some labels denote a clini-
cal syndrome without specific histological implications In 1892, Arnold Pick, working in Prague, reported a
(e.g. progressive aphasia, semantic dementia or dementia 71-year-old man with progressive mental deterioration and
of frontal type), others denote specific neuropathological unusually severe aphasia, who at post-mortem had marked
entities (e.g. PiD or familial tauopathy), hybrid clinico- atrophy of the cortical gyri of the left temporal lobe (Pick,
pathological entities (e.g. FTD) or even specific genetic dis- 1892; Girling and Berrios, 1994). Pick wanted to draw atten-
orders (e.g. FTD with parkinsonism linked to chromosome tion to the fact that local accentuation of progressive brain
17). Recent differences in opinion over terminology are atrophy may lead to symptoms of local disturbance (in
well illustrated by the titles of the following books, all pub- this instance aphasia). He also made specific predictions
lished in the last decade: Pick’s Disease and Pick Complex regarding the role of the mid-temporal region of the left
(Kertesz and Munoz, 1998), Frontotemporal Dementia hemisphere in the representation of word meaning. In sub-
(Pasquier et al., 1996), Fronto-Temporal Lobar Degeneration: sequent papers, he described four further patients with left
Fronto-Temporal Dementia, Progressive Aphasia, Semantic temporal atrophy (Girling and Berrios, 1997) or frontotem-
Dementia (Snowden et al., 1996) Frontotemporal Dementia poral atrophy (Pick, 1901; Girling and Markova, 1995). It
Syndromes (Hodges, 2007). This lack of clarity is acknowl- was only in his 1906 publication that Pick turned his atten-
edged and efforts to rationalize terminology and achieve tion to bilateral frontal atrophy with resultant behavioural
clinicopathological and nosological consensus have been disturbance.

759
760 Dementia

Several points should be emphasized, which is as follows: labels to describe patients with the clinical syndrome of
progressive frontal or temporal lobe degeneration (Baldwin
●● Pick’s primary interest in these patients was their and Förstl, 1993).
language disorder, particularly, the clinicoanatomical
correlates of aphasia.
●● He did not claim to have discovered a new disease, 66.3 REDISCOVERING PID: FROM
merely novel phenomena arising from asymmetric
DEMENTIA OF THE FRONTAL
degeneration.
●● He did not describe distinct neuropathological changes
TYPE AND PROGRESSIVE
in his patients with focal atrophy. APHASIA TO FTD
●● Both the major syndromes now included under the
rubric of FTD (dementia of frontal type and semantic 66.3.1 DEMENTIA OF FRONTAL TYPE
dementia) were clearly reported by Pick.
A renaissance of interest in the focal dementias occurred
In view of these monumental contributions, it is sad that in the 1980s. Workers from Lund (Brun, 1987; Gustafson,
Pick has been relegated to a minor place in the modern ter- 1987) reported on a large series of patients with frontal lobe
minology of FTD. degeneration out of which only a small proportion of whom
The histological abnormalities associated with PiD were, had Pick cells and Pick bodies – the remainder had very
in fact, described a few years later by Alzheimer (1911) who similar findings but without specific inclusions (i.e. focal
recognized changes distinct from those found in the form lobar atrophy with severe neuronal loss and spongiosis) –
of cerebral degeneration later associated with his name. the Lund group preferred to adopt the term ‘frontal degen-
Alzheimer recognized both argyrophilic intracytoplasmic eration of non-Alzheimer type’. At approximately the same
inclusions (Pick bodies) and diffusely staining ballooned time, Neary et al. (1986) in Manchester began a series of
neurones (Pick cells) in association with focal lobar atro- important clinicopathological studies of patients with pre-
phy. A comprehensive review of 20 patients with the litera- senile dementia. They, likewise, found a high proportion
ture of aphasia due to focal lobar atrophy written soon after of cases with a progressive frontal lobe syndrome that had
Alzheimer’s description (Mingazzini, 1914), did not use the neither specific changes of AD (plaques and tangles) nor
label PiD, which was introduced by Gans some 8 years later specific Pick pathology. They introduced the term ‘demen-
(Gans, 1922). The term was then taken up by Onari and tia of frontal type’. Over the next few years other groups
Spatz (1926), but Carl Schneider (1927) is probably most described very similar cases under the labels ‘frontal lobe
responsible for its popularization. degeneration’ (Miller et al., 1991) and ‘dementia lacking
Unfortunately, however, Schneider concentrated on the distinct histological features’ (Knopman et al., 1990). My
frontal lobe component of the syndrome and thus began own preferred term for this group of patients is behavioural
the neglect of the temporal lobe syndromes. He distin- variant of FTD (bvFTD). (Davies et al., 2006; Kipps et al.,
guished three clinical phases, the first characterized by 2007; Rascovsky et al., 2007; Hornberger et al., 2008; Kipps
impaired judgement and behaviour, the second by local et al., 2009a).
symptoms and the third by generalized dementia. Many In the advanced stages of the disease, patients with this
papers describing PiD then appeared in the 1930s and disorder present no diagnostic difficulty. They have the triad
1940s, which again mainly focused on the frontal lobe of the following:
aspects of the disorder.
After World War II, interest in PiD faded, together with ●● A profound change in personality and behaviour
a general waning of interest in the cognitive aspects of ●● Neuropsychological impairments indicative of selective
neurology. The focus of interest in English language pub- or disproportionate frontal pathology
lications became the neuropathology and later the genet- ●● Appropriate changes on structural and/or functional
ics of PiD. This resulted in a gradual change in the criteria brain imaging
for PiD, which evolved to include the necessity for specific
pathological changes (i.e. focal atrophy with Pick cells and Diagnosing early cases is by no means easy and one of
or Pick bodies). Indeed, some authors went as far as to claim the challenges is to develop better methods of early, accurate
that AD and PiD were clinically indistinguishable in one’s detection (Gregory et al., 1999). Since patients are typically
lifetime (Katzman, 1986). In continental Europe, however, unaware of the insidious changes noted by others, we rely
there remained a strong interest in the clinical phenomena upon carer-based assessments. One undoubted advance in
of the dementias; PiD remained an in vivo diagnosis based the area has been therefore, the development of standard-
on a combination of clinical features suggestive of frontal ized carer interviews such as the Neuropsychiatric Inventory
and/or temporal lobe dysfunction and focal lobar atrophy (Cummings et al., 1994), the Cambridge Behavioural
(Mansvelt, 1954; Tissot et al., 1975). This controversy con- Inventory (Wedderburn et al., 2008) and the Frontal
tinues and has contributed to the adoption of the many Behaviour Inventory (Kertesz et al., 1997, 2000, 2003a). Recent
Pick’s disease: Its relationship to progressive aphasia, semantic dementia and frontotemporal dementia 761

work has focused on specific behaviours that can discriminate reported six patients with a long history of insidiously wors-
patients with bvFTD and semantic dementia from patients ening aphasia in the absence of signs of more generalized
with AD, such as disinhibition, loss of empathy, changes in cognitive failure. One of these patients underwent a brain
appetite, food preferences and eating habits (Ikeda et al., biopsy, which revealed non-specific histology without spe-
2002) or the presence of stereotypes (Nyatsanza et al., 2003). cific markers of either AD or PiD. Following Mesulam’s
An international effort has culminated in the publication seminal paper, approximately 100 patients with progressive
of consensus guidelines for the diagnosis of bvFTD, which aphasia were reported over the next 15 years (for reviews
appear to be robust (Rascovsky et al., 2011). see: Mesulam and Weintraub, 1992; Hodges and Patterson,
Many patients with bvFTD, who visit psychiatrists, 1996; Snowden et al., 1996; Garrard and Hodges, 1999).
acquire a label of functional psychiatric disorder (including From this literature, it became clear that, although the lan-
simple schizophrenia, depression and obsessive-compul- guage impairment in patients with progressive aphasia is
sive disorder) because despite gross behavioural changes heterogeneous, there are two identifiable and distinct apha-
other aspects of cognition are typically preserved (Ames sia syndromes: progressive non-fluent aphasia and progres-
et al., 1994; Gregory and Hodges, 1996; Miller et al., 1997). sive fluent aphasia. In the latter syndrome, speech remains
This is understandable in terms of the site of pathology fluent and well-articulated but becomes progressively devoid
in bvFTD, the orbital and mesial frontal lobes, which are of content words. The language and other non-verbal cogni-
critically involved in social judgement, motivation, risk tive deficits observed in these patients reflect a breakdown
assessment and compulsive behaviour (Cummings, 1999). in semantic memory that has led many authors to apply the
By contrast, dorsolateral frontal regions, which have been label of ‘semantic dementia’ (Snowden et al., 1989; Hodges
the focus of classic neuropsychological studies, are spared et al., 1992, 1994; Saffran and Schwartz, 1994; Hodges and
in the early stages of the disease. The development of Patterson, 1996).
quantifiable tasks sensitive to orbital and mesial frontal Although, the term ‘semantic dementia’ is recent, the
function has begun with paradigms involving probability syndrome has, as discussed above, been recognized under
and gambling (Rogers et al., 1998; Rahman et al., 1999), different labels for many years. Pick (1892) and Warrington
emotion processing (Keane et al., 2002; Piguet et al., 2011; (1975) were the first to clearly delineate the syndrome of
Kumfor et al., 2013) and ‘theory of mind’ (ToM) tasks, semantic memory impairment in recent times. Drawing on
such as detection of social faux pas (Gregory et al., 2002; the work of Tulving (1972, 1983), Warrington recognized
Torralva et al., 2007) and the discrimination of sincere that the progressive anomia in her patients was not sim-
from sarcastic statements (Kipps et al., 2009b; Rankin ply a linguistic deficit, but reflected a fundamental loss of
et al., 2009). semantic memory (or knowledge) about the items, which
An important recent development has been the identi- thereby affected naming, word comprehension and object
fication of a subgroup of patients with clinical features of recognition. Semantic memory is the term applied to the
bvFTD who show little or no progression, often over many component of long-term memory that contains the perma-
years of follow-up (Davies et al., 2006; Kipps et al., 2007, nent representation of our knowledge about things in the
2008). Such patients are almost universally men and can world and their interrelationship, facts and concepts as well
be distinguished on the basis of a number of factors: nor- as words and their meaning (Hodges et al., 1992; Garrard
mal performance on executive and social cognition tasks et al., 1997; Hodges and Patterson, 1997; Hodges et al., 1998).
(Hornberger et al., 2008; Kipps et al., 2009b), preserva- Cases of semantic dementia have also been recognized for
tion of activities of daily living (Mioshi and Hodges 2007; many years in Japan as cases of ‘gogi (word meaning) apha-
Mioshi et al., 2007), absence of brain atrophy on magnetic sia’ (Tanabe et al., 1992).
resonance imaging (MRI) (Davies et al., 2006; Kipps et al., Our original paper reported five cases with progres-
2009a) and normal fluorodeoxyglucose-positron emission sive loss of semantic memory and focal temporal atrophy
tomography (FDG-PET) (Kipps et al., 2009b). The aetiol- (Hodges et al., 1992). All five presented a fluent aphasia
ogy of the so-called bvFTD phenotype syndrome remains and all the characteristics of semantic memory loss: empty
unclear. A small proportion of such patients have the newly speech with word-finding difficulty and occasional seman-
discovered C9orf72 mutation (discussed in Section 66.4) tic paraphasias (mother for father), a severe reduction in
while others have decompensated personality disorders or the generation of exemplars on category fluency tests (in
functional psychiatric disorders but much work remains to which subjects are asked to produce as many examples as
be done in this area. possible from defined semantic categories, such as animals
or musical instruments, within 1 minute), impaired single
66.3.2 PROGRESSIVE APHASIA AND word comprehension on picture-pointing tests, a loss of
SEMANTIC DEMENTIA fine-grained attribution knowledge about a range of items
with preservation of broad superordinate information on
The other strand of the story concerns the rediscovery of the verbal and pictorial tests of knowledge. By contrast, other
syndrome of progressive aphasia in association with focal aspects of language competency (phonology and syntax)
left perisylvian or temporal lobe atrophy. In 1982, Mesulam were strikingly preserved. In contrast to AD, the patients
762 Dementia

also had good day-to-day (episodic) memory, although disease (MND) (Chare et al., 2014). Second is the evolution
more recently it has been shown that this sparing of auto- of the pattern of cognitive and behavioural changes over
biographical memory applies to fairly recent memories only time: although semantic dementia patients present with pro-
(Graham and Hodges, 1997; Hodges and Graham, 1998; gressive anomia and other linguistic deficits, on follow-up
Irish et al., 2011). They also showed no impairment on tests show emergence of the features that characterize the frontal
of immediate (working) memory, visually based problem- form of FTD (Hodges and Patterson, 1996; Edwards Lee et
solving or visuoperceptual abilities. Four out of the five al., 1997). Indeed, semantic dementia and bvFTD may share
patients showed severe and circumscribed temporal lobe many behavioural features even on presentation (Bozeat et al.,
atrophy on computed tomography (CT) or MRI scanning. 2000). Third, the fact that modern neuroimaging techniques
Since 1992, we have studied many such patients further and demonstrate subtle involvement of the orbitofrontal cortex in
have confirmed the association of the temporal lobe with the majority of cases presenting prominent temporal atrophy
focal atrophy – involving the temporal pole and inferolateral and semantic dementia (Mummery et al., 2000; Rosen et al.,
neocortex (particularly, the perirhinal cortex and fusiform 2002a; Williams et al., 2005) needs to be acknowledged.
gyrus). In many cases, the atrophy is strikingly asymmetric, The status of patients with the non-fluent form of pro-
involving the left side predominately (Hodges et al., 1998; gressive aphasia within the spectrum of FTD is more com-
Garrard and Hodges, 1999; Mummery et al., 1999; Galton plex. Clinically, such cases are clearly separable from cases
et al., 2001; Hodges and Patterson 2007). of semantic dementia. Speech is faltering and distorted
This raises the following question: what are the with frequent phonological substitutions and grammatical
cognitive and/or behavioural signatures of isolated right errors in such cases. Other non-language-based aspects of
temporal atrophy? In 1994, we reported a patient who cognition remain well preserved, as do activities of daily liv-
suffered from visual hallucination (VH), with progres- ing. Changes in behaviour and personality of the type that
sive prosopagnosia, followed by a specific loss of knowl- typify dementia of frontal type are seen in the later stages
edge about people (Evans et al., 1995). This VH patient of semantic dementia, but are rare after a number of years
was unable to identify even very famous people from their when global cognitive decline occurs (Green et al., 1990;
face or name, yet had intact general semantic and autobio- Hodges and Patterson, 1996). Some patients with progres-
graphical memory (Kitchener and Hodges, 1999). A more sive non-fluent aphasia have Alzheimer pathology at post-
recent study (Thompson et al., 2004) reported a dissocia- mortem, albeit with an atypical distribution; that is to say
tion of person-specific from general semantic knowledge marked involvement of perisylvian language areas but
in two patients with contrasting patterns of temporal atro- sparing of medial temporal structures (Greene et al., 1996;
phy. Subject MA, with predominantly left temporal atro- Galton et al., 2000; Knibb et al., 2006) occurs, while oth-
phy, showed impairment of general semantics with relative ers have tau-positive inclusion pathology with classic Pick
preservation of knowledge about people, while subject JP, bodies or more diffuse neuronal and glial tau inclusions.
with predominantly right temporal atrophy, showed the (Weintraub et al., 1990; Mesulam and Weintraub, 1992;
opposite pattern of impairments with severely impaired Knibb et al., 2006). Recent functional neuroimaging stud-
person-specific knowledge in the context of relatively pre- ies have implicated the anterior insula as the key abnormal
served knowledge about objects and animals. The group led region (Nestor et al., 2003).
by Miller have also drawn attention to the bizarre behav- A potentially important recent development has been
iours (including irritability, impulsiveness, alterations in the separation of non-fluent aphasic patients into two sub-
dress, limited and fixed ideas and decreased facial expres- groups. The first group was characterized by apraxia of
sion) exhibited by patients with predominantly right tem- speech and/or agrammatism in association with left interior
poral lobe atrophy (Edwards Lee et al., 1997; Miller et al., frontal lobe and insular atrophy and underlying FTD tau
1997). A recent review of 47 patients with semantic demen- pathology. The second group had severe anomia, dysfluency
tia identified distinct behavioural and cognitive profiles because of word-finding pauses, occasional phonological
associated with the left and right temporal variants of the errors and markedly impaired sentence repetition that have
disease (Thompson et al., 2003). Social awkwardness, job been termed logopenic progressive aphasia with emerging
loss, lack of insight and difficulty with person identifica- evidence of largely AD pathology at post-mortem or on
tion were all more likely to be associated with major right amyloid-based PET imaging (Gorno-Tempini et al., 2008;
temporal atrophy, whereas word-finding difficulties and Mesulam et al., 2008; Leyton et al., 2011).
reduced comprehension were all more likely to be associ-
ated with predominantly left-sided atrophy.
There are a number of compelling reasons to consider
semantic dementia as part of the same disease spectrum as
66.4 FAMILIAL FTD
bvFTD. The first is pathological: the majority of cases with
semantic dementia have deposition of TAR-DNA-binding The story of the chromosome 17 linkage is extraordinary
protein-43 (TDP-43), which constitutes the pathological in a number of ways. Families around the world with what
variants of FTLD is also found in a proportion of those with has become to be known as FTD with parkinsonism linked
behavioural presentations of FTD and in motor neurone to chromosome 17 (FTDP-17) (Spillantini et al., 1998a)
Pick’s disease: Its relationship to progressive aphasia, semantic dementia and frontotemporal dementia 763

had originally been reported under a range of headings region of chromosome 9 accounts for approximately a third
including: disinhibition–dementia–parkinsonism–amyotro- of familial FTD and may be a half of familial amyotrophic
phy complex (DDPAC) (Wilhelmsen et al., 1994), rapidly pro- lateral sclerosis (ALS) cases (DeJesus-Hermandez et al.,
gressive autosomal dominant parkinsonism and dementia 2011; Renton et al., 2011;Majounie et al., 2012). The major-
with pallido-ponto-nigral degeneration (Wszolek et al., 1992), ity of FTD cases have the behavioural variant. The clini-
familial progressive subclinical gliosis (Petersen et al., 1995), cal course may be very protracted with little in the way of
hereditary dysphasic disinhibition dementia (Morris et al., brain atrophy and a high rate of psychotic phenomena has
1984), hereditary FTD (Heutink et al., 1997), multiple system been reported in gene carriers (Devenney et al., 2014).
tauopathy with presenile dementia (Spillantini et al., 1997),
familial presenile dementia with psychosis (Sumi et al., 1992)
and PiD (Schenk, 1951). In 1996, a meeting of representatives
from all of the groups identifying linkage to chromosome 17 66.5 CORTICOBASAL DEGENERATION,
was held in Ann Arbor, MI (Foster et al., 1997). Comparison of
PROGRESSIVE SUPRANUCLEAR
clinical and pathological data revealed a great deal of similar-
ity between the families who all shared the characteristics of
PALSY AND FTD
predominately frontotemporal distribution of pathology with
marked behavioural changes. Extrapyramidal dysfunction In 1967, Rebeiz et al. described three patients with a neuro-
was present in most. In some families, psychotic symptoms degenerative illness affecting both cortex and basal ganglia
were a major feature and a number of them had amyotrophy. (Rebeiz et al., 1968). Each patient presented with a progres-
It was recognized at that time that some of the families shared sive asymmetrical akinetic-rigid syndrome and apraxia; on
the common pathology with microtubule-associated protein the basis of neuropathology findings. Rebeiz et al. called
(MAP) tau-positive inclusions. Progress in this field then the disorder ‘corticodentatonigral degeneration with neu-
became rapid. It was soon discovered that, most, if not all, ronal achromasia’, later renamed to corticobasal degenera-
families had tau inclusions with a distinctive morphological tion (CBD). Initially, CBD was conceptualized as a distinct
pattern leading to the coining of the term ‘familial tauopathy’ clinicopathological entity, but in the early 1990s it was
and the suggestion that the disease might reflect a mutation recognized that considerable clinical and pathological het-
in the tau gene known to be located in the 17q21–22 region erogeneity existed within the disorder. Clinically, cases of
(Spillantini et al., 1998a). Within 2 years of the Ann Arbor pathologically proven CBD were described presenting the
meeting, the first mutations were identified (Dumanchin clinical syndromes of FTD (Mathuranath et al., 2000) or
et al., 1998; Hutton et al., 1998; Poorkaj et al., 1998; Spillantini progressive aphasia (Lippa et al., 1991). Conversely, it was
et al., 1998b). Subsequent progress has been rapid: more than recognized that other pathologies might underlie the clini-
50 different tau mutations have been reported in association cal syndrome of CBD; in a review of 32 consecutive cases of
with familial FTD, differing according to their positions in clinically diagnosed CBD (Boeve et al., 1999), the underly-
the tau gene, their effects on tau messenger RNA (mRNA) ing pathological diagnosis was CBD in 18 cases, AD in 3
and protein and the type of tau pathology they cause (Ingram cases, PiD in 2, progressive supranuclear palsy (PSP) in 6
and Spillantini, 2002). and dementia lacking specific histology in another 2 of the
Not all cases of familial FTD are caused by tau muta- total cases. The pathological heterogeneity has been recog-
tions. In one large Danish family with autosomal dominant nized in subsequent studies (Ling et al., 2010). A distinction
FTD, linkage has been established to chromosome 3 (Brown has been drawn by some authors between the corticobasal
et al., 1995; Brown, 1998) and, in some families with FTD- syndrome (the constellation of clinical features character-
MND, linkage has been established to chromosome 9. In istically) and CBD (the histopathological disorder itself).
a series of 22 cases of familial FTD only, 11 (50%) had tau There is growing awareness of the overlap between the
mutations (Morris et al., 2001). clinical syndrome of CBD and progressive non-fluent apha-
In 2006, another major breakthrough established muta- sia (PNFA): many patients with PNFA develop apraxia and
tions of the progranulin gene also on chromosome 17, which parkinsonian features and conversely, many CBD patients
results in dominantly inherited FTD with ubiquitin-posi- have subtle language production deficits that worsen as the
tive tau-negative pathology (Baker et al., 2006). Mutations disease progresses (Graham et al., 2003).
of the progranulin gene account for a proportion of familial The related disorder, PSP, originally described by Steele,
cases not caused by tau gene mutations (Gass et al., 2006). In Richardson and Olszewski in 1964 is classically character-
the same year, mutations of progranulin have been linked ized by progressive axial rigidity, bradykinesia, vertical
to abnormalities of TDP-43 identified as the pathologi- gaze palsy and dysarthria. Pathologically, tau-positive neu-
cal protein in the majority of those with FTLD with MND rofibrillary tangles and threads are found in the substantia
type inclusions but without MND (FTLD-U) and in MND nigra, the subthalamic nucleus and the dentate nucleus. It
(Neumann et al., 2006). is increasingly recognized that pathological PSP may pres-
The most exciting recent development has been the dis- ent itself with the clinical features of CBD or even progres-
covery of the C9orf72 gene mutation. This abnormality, sive aphasia or a frontal lobe syndrome. Conversely, the
a pathological hexanucleotide repeat on the non-coding PSP phenotype may result from tau gene mutations (Morris
764 Dementia

et al., 2003; Soliveri et al., 2003). Both CBD and PSP share tau gene mutations (FTDP-17) and diffuse tau-positive
an identified genetic risk factor, the H1 extended haplotype neuronal and astrocytic immunoreactivity, those charac-
of the tau gene and the tau deposits in both these diseases terized by tau-positive astrocytic plaques and ballooned
consist of the four-repeat isoforms of tau. achromatic neurones (CBD) and those with tau-positive
argyrophilic grain disease (AGD) are included in this cri-
teria. Second, cases with TDP-43-positive inclusions in the
dentate gyrus and brain stem motor nuclei which include
66.6 FTD WITH MND cases with associated MND also include those with pro-
granulin and C9orf72 gene mutations, which have been
Although, MND has traditionally been regarded as a dis- subclassified according to the neuropathological appear-
order which spares higher cognitive abilities, it has become ance. Third, FTD with fused-in sarcoma (FUS)-positive
clear since early reports from Japan (Mitsuyama and inclusions which constitutes a very small proportion of
Takamiya, 1979) that the rate of dementia in MND is sig- cases is included in this category.
nificantly greater than expected. Indeed, 10% to 20% of The question as to whether distinct pathological sub-
patients with MND show features of FTD (Rakowicz and types map onto distinct clinical syndromes is of great
Hodges, 1998; Lillo and Hodges, 2009) and a much higher interest. Relatively few clinicopathological series have
proportion have more subtle behavioural changes (Lillo been reported till date (Rascovsky et al., 2002; Rosen et al.,
et al., 2011). Neuroimaging in MND cases with cognitive 2002b; Hodges et al., 2003; Josephs et al., 2006; Mesulam
impairment demonstrates widespread frontal and temporal et al., 2008; Chare et al., 2014). To summarize, it appears
atrophy. Conversely, a significant minority of patients with that the majority of patients with semantic dementia have
FTD develop features of MND (Neary et al., 1990; Caselli TDP-43-positive pathology with relatively few cases hav-
et al., 1993; Rakowicz and Hodges, 1998; Bak and Hodges, ing tau-positive inclusions. The converse is true of PNFA,
1999; Strong et al., 2003; Burrell et al., 2011). Such patients which is typically associated with tau-positive forms of FTD
present cognitive symptoms, either with bvFTD or with while those with the logopenic variant of progressive apha-
progressive aphasia, which then progresses rapidly, followed sia show Alzheimer pathology. Pathology in bvFTD is least
by the emergence of bulbar features and mild limb amyotro- predictable with approximately a half having tau-positive
phy. Such cases have a characteristic pattern of histological FTD and the other half having TDP-43-positive FTD.
change with TDP-43-positive inclusions in cortical regions
and the dentate gyrus. One interesting observation is that
FTD with MND (FTD-MND) patients almost invariably
have disproportionately greater impairment of verb rather
66.8 HOW COMMON IS FTD?
than noun processing, which affects both production and
comprehension (Bak et al., 2001) and have a high rate of psy- Historically, it has long been recognized that FTD is less
chotic symptoms (Lillo et al., 2010; Devenney et al., 2014). common than AD. Until recently, however, most estimates
The overlap has been consolidated by the discovery of the of the prevalence of FTD were based on reports from spe-
C9or72 mutation, which is present in a high proportion of cialist units where FTD tended to be over-represented as a
cases with the overlap syndrome as well as those with famil- diagnosis. This has changed with the publication of a num-
ial FTD and MND. ber of community-based studies that give a clearer picture
of the true prevalence of FTD.
Ratnavalli et al. (2002) conducted a prevalence survey
aiming to ascertain all cases of early-onset dementia (age of
66.7 CORRELATION BETWEEN
onset less than 65 years) in a particular U.K. region with a
CLINICAL SYNDROMES AND
total population of 326,000. The overall prevalence for early-
NEUROPATHOLOGY onset dementia was 81 per 100,000 patients aged 45–64
years, with equivalent prevalence of AD and FTD among
The definitive diagnosis of FTD depends upon neuro- 15 per 100,000 patients. Of 185 cases seen by the study
pathological examination. Unlike other dementia syn- group, 23 (12%) fulfilled the Lund– Manchester criteria for
dromes, notably AD, FTD encompasses considerable FTD, suggesting that, although FTD is by no means rare, it
pathological heterogeneity. There have been considerable still represents only a minority of cases of dementia in the
developments in the past few years. Three broad subdivi- under-65 age group. (For comparison, AD was diagnosed in
sions have been recognized depending on the profile of 34%, vascular dementia in 18%, alcohol-related dementia in
immunohistochemical staining and the pattern of intra- 10% and dementia with Lewy bodies in 7%.)
cellular inclusions (Mackenzie et al., 2010; Hodges et al., Similar figures were obtained by Harvey et al. (2003),
2011; Josephs et al. 2011). First of all, patients with tau- who surveyed three London boroughs with a total popula-
positive pathology, which in turn comprise a number of tion of 567,500 people and calculated an overall prevalence
subvariants including those with classic argyrophilic, for early-onset dementia among 98 per 100,000 patients
tau-positive, intraneuronal Pick bodies, those with MAP aged 45–64 years.
Pick’s disease: Its relationship to progressive aphasia, semantic dementia and frontotemporal dementia 765

The prevalence of FTD in the over-65-year olds is largely Burrell, J., Kiernan, M.C., Vucic, S. and Hodges, J.R. (2011).
unknown. Recent prospective investigation of 451 patients Motor neuron dysfunction in FTD. Brain: A Journal of
who were 85-year olds in Sweden, found that 86 (19%) ful- Neurology, 134: 2582–2594.
filled the criteria for a frontal lobe syndrome and 14 (3%) Caselli, R.J., Windebank, A.J., Petersen, R.C. et al. (1993).
fulfilled the Lund–Manchester criteria for the frontal variant Rapidly progressive aphasic dementia and motor neu-
of frontotemporal dementia (fvFTD) (Gislason et al., 2003). ron disease. Annals of Neurology, 33: 200–207.
Chare, L., Hodges, J.R., Leyton, C.E. et al. (2014). New cri-
teria for frontotemporal dementia syndromes: Clinical
REFERENCES and pathological diagnostic implications. Journal of
Neurology, Neurosurgery, and Psychiatry, 85: 865–870.
Alzheimer, A. (1911). Über eigenartige krankheitsfälle des Cummings, J.L. (1999). Principles of neuropsychiatry:
späteren Alters (About peculiar cases of disease in Towards a neuropsychiatric epistemology. Neurocase,
old age). Zeitschrift für die Gesamte Neurologie und 5: 181–188.
Psychiatrie, 4: 356–385. Cummings, J.L., Mega, M., Gray, K. et al. (1994). The
Ames, D, Cummings, J.L., Wirshing, W.C. et al. (1994). neuropsychiatric inventory: Comprehensive assess-
Repetitive and compulsive behavior in frontal lobe ment of psychopathology in dementia. Neurology,
degenerations. Journal of Neuropsychiatry and Clinical 44: 2308–2314.
Neurosciences, 6: 100–113. Davies, R.R., Kipps, C.M., Mitchell, J. et al. (2006).
Bak, T. and Hodges, J.R. (1999). Cognition, language Progression in frontotemporal dementia: Identifying
and behaviour in motor neurone disease: Evidence a benign behavioral variant by magnetic resonance
of frontotemporal dementia. Dementia and Geriatric imaging. Archives of Neurology, 63: 1627–1631.
Cognitive Disorders, 10: 29–32. DeJesus-Hernandez, M., Mackenzie, I.R., Boeve, B.F. et al.
Bak, T.H., O’Donovan, D.G., Xuereb, J.H. et al. (2001). (2011). Expanded GGGGCC hexanucleotide repeat in
Selective impairment of verb processing associated noncoding region of C9ORF72 causes chromosome
with pathological changes in the Brodman areas 44 9p-linked FTD and ALS. Neuron, 72: 245–256.
and 45 in the motor neurone disease-dementia-aphasia Devenney, E., Hornberger, M., Irish, M. et al. (2014).
syndrome. Brain: A Journal of Neurology, 124 (Pt 1): Frontotemporal dementia associated with the
103–120. C9ORF72 mutation – A unique clinical profile. JAMA
Baker, M., Mackenzie, I.R., Pickering-Brown, S.M. et al. Neurology, 71: 331–339.
(2006). Mutations in progranulin cause tau-negative Dumanchin, C., Camuzat, A., Campion, D. et al. (1998).
frontotemporal dementia linked to chromosome 17. Segregation of a missense mutation in the micro-
Nature, 442 (7105): 916–919. tubule-associated protein tau gene with familial
Baldwin, B. and Förstl, H. (1993). Pick’s disease – 101 years frontotemporal dementia and parkinsonism. Human
on still there but in need of reform. British Journal of Molecular Genetics, 7: 1825–1829.
Psychiatry, 163: 100–105. Edwards Lee, T., Miller, B., Benson, F. et al. (1997). The
Boeve, B.F., Maraganore, D.M., Parisi, J.E. et al. (1999). temporal variant of frontotemporal dementia. Brain:
Pathologic heterogeneity in clinically diagnosed corti- A Journal of Neurology, 120: 1027–1040.
cobasal degeneration. Neurology, 53: 795–800. Evans, J.J., Heggs, A.J., Antoun, N. et al. (1995).
Bozeat, S., Gregory, C.A., Lambon Ralph, M.A. and Progressive prosopagnosia associated with selective
Hodges, J.R. (2000). Which neuropsychiatric and right temporal lobe atrophy: A new syndrome? Brain:
behavioural features distinguish frontal and temporal A Journal of Neurology, 118: 1–13.
variants of frontotemporal dementia from Alzheimer’s Foster, N.L., Wilhelmsen, K., Sima, A.A.F. et al. (1997).
disease? Journal of Neurology, Neurosurgery, and Frontotemporal dementia and parkinsonism linked to
Psychiatry, 69: 178–186. chromosome 17: A consensus conference. Annals of
Brown, J. (1998). Chromosome 3-linked frontotempo- Neurology, 41: 706–715.
ral dementia. Cellular and Molecular Life Sciences, Galton, C.J., Patterson, K., Graham, K.S. et al. (2001).
54: 925–927. Differing patterns of temporal atrophy in Alzheimer’s dis-
Brown, J., Ashworth, A., Gydesen, S. et al. (1995). Familial ease and semantic dementia. Neurology, 57: 216–225.
nonspecific dementia maps to chromosome 3. Human Galton, C.J., Patterson, K., Xuereb, J.H., Hodges,
Molecular Genetics, 4: 1625–1628. J.R et al. (2000). Atypical and typical presentations
Brun, A. (1987). Frontal lobe degeneration of non- of Alzheimer’s disease: A clinical, neuropsychologi-
Alzheimer’s type. I. Neuropathology. Archives of cal, neuroimaging and pathological study of 13 cases.
Gerontology and Geriatrics, 6: 209–233. Brain: A Journal of Neurology, 123: 484–498.
Brun, A., Englund, B., Gustafson, L. et al. (1994). Clinical Gans, A. (1922). Betrachtungen über art und ausbreitung
and neuropathological criteria for frontotemporal des krankhaften prozesses in einem fall von picksher
dementia. Journal of Neurology, Neurosurgery, and atrophie des stirhirns. Zeitschrift für die Gesamte
Psychiatry, 57: 416–418. Neurologie und Psychiatrie, 80: 10–28.
766 Dementia

Garrard, P. and Hodges, J.R. (1999) Semantic dementia: Gregory, C.A., Serra-Mestres, J. and Hodges, J.R. (1999).
Implications for the neural basis of language and Early diagnosis of the frontal variant of frontotemporal
meaning. Aphasiology, 13: 609–623. dementia: How sensitive are standard neuroimag-
Garrard, P., Perry, R. and Hodges, J.R. (1997) Disorders ing and neuropsychologic tests? Neuropsychiatry,
of semantic memory. Journal of Neurology, Neuropsychology and Behavioural Neurology,
Neurosurgery, and Psychiatry, 62: 431–435. 12: 128–135.
Gass, J., Cannon, A., Boeve, B. et al. (2006). Mutations Gustafson, L. (1987). Frontal lobe degeneration of non-
in progranulin are a major cause of ubiquitin-positive Alzheimer’s type II: Clinical picture and differential
frontotemporal lobar degeneration. Human Molecular diagnosis. Archives of Gerontology and Geriatrics,
Genetics, 15: 2988–3001. 6: 209–223.
Girling, D.M. and Berrios, G.E. (1994). On the relationship Harvey, R.J., Skelton-Robinson, M. and Rossor, M.N. (2003).
between senile cerebral atrophy and aphasia. History The prevalence and causes of dementia in people
of Psychiatry, 8: 542–547 (translation of Pick, A. [1892]. under the age of 65 years. Journal of Neurology,
Über die beziehungen der senilen atrophie zur aphasie. Neurosurgery, and Psychiatry, 74: 1206–1209.
Prager Medizinische Wochenschrift, 17: 165–167). Heutink, P., Stevens, M., Rizzu, P. et al. (1997). Hereditary
Girling, D.M. and Berrios, G.E. (1997). On the symptom- frontotemporal dementia is linked to chromosome
atology of left-sided temporal lobe atrophy. History 17q21-q22: A genetic and clinicopathological study of
of Psychiatry, 8: 149–159 (translation of Pick, A. [1904]. three Dutch families. Annals of Neurology, 41: 150–159.
Zur symptomatologie der linksseitigen schäfenlappe- Hodges, J.R. (2007). Frontotemporal Dementia
natrophie. Monatschrift für Psychiatrie und Neurologie, Syndromes, Vol 1. 334. Cambridge, United Kingdom:
16: 378–88). Cambridge University Press.
Girling, D.M. and Markova, I.S. (1995). Senile atrophy as Hodges, J.R., Davies, R., Xuereb, J. et al. (2003). Survival
the basis for focal symptoms. History of Psychiatry, 6: in frontotemporal dementia. Neurology, 61: 349–354.
533–537 (translation of Pick, A. [1901]. Senile hirnatro- Hodges, J.R., Garrard, P., Patterson, K. (1998). Semantic
phie als grundlage für herderescheinungen. Wiener dementia. In Kertesz, A. and Munoz, D.G. (eds.), Pick’s
Klinische Wochenschrift, 14: 403–404). Disease and Pick Complex. New York, NY: Wiley-Liss
Gislason, T.B., Sjogren, M., Larsson, L. and Skoog, Inc., pp. 83–104.
I. (2003). The prevalence of frontal variant frontotem- Hodges, J.R. and Graham, K.S. (1998). A reversal of the
poral dementia and the frontal lobe syndrome in a temporal gradient for famous person knowledge in
population based sample of 85 year olds. Journal of semantic dementia: Implications for the neural orga-
Neurology, Neurosurgery, and Psychiatry, 74: 867–871. nization of long-term memory. Neuropsychologia,
Gorno-Tempini, M.L., Brambati, S.M., Ginex, V. et al. 36: 803–825.
(2008). The logopenic/phonological variant of primary Hodges, J.R. and Patterson, K. (1996). Nonfluent progres-
progressive aphasia. Neurology, 71: 1227–1234. sive aphasia and semantic dementia: A comparative
Graham, N.L., Bak, T.H. and Hodges, J.R. (2003). neuropsychological study. Journal of the International
Corticobasal degeneration as a cognitive disorder. Neuropsychological Society, 2 (6): 511–524.
Movement Disorders, 18: 1224–1232. Hodges, J.R. and Patterson, K.E. (1997). Semantic memory
Graham, K.S. and Hodges, J.R. (1997). Differentiating the disorders. Trends in Cognitive Science, 1: 67–72.
roles of the hippocampal complex and the neocor- Hodges, J.R., Patterson, K., Oxbury, S. et al. (1992).
tex in long-term memory storage: Evidence from the Semantic dementia progressive fluent aphasia with
study of semantic dementia and Alzheimer’s disease. temporal lobe atrophy. Brain: A Journal of Neurology,
Neuropsychology, 11: 77–89. 115: 1783–1806.
Green, J., Morris, J.C., Sandson, J. et al. (1990). Hodges, J.R, Patterson, K. and Tyler, L.K. (1994). Loss of
Progressive aphasia: A precursor of global dementia? semantic memory: Implications for the modularity of
Neurology, 40: 423–429. mind. Cognitive Neuropsychology, 11: 505–542.
Greene, J.D.W., Patterson, K., Xuereb, J. and Hodges, Hornberger, M., Piguet, O., Kipps, C., Hodges, J.R. et al.
J.R. (1996) Alzheimer disease and nonfluent progres- (2008). Executive function in progressive and nonpro-
sive aphasia. Archives of Neurology, 53: 1072–1078. gressive behavioral variant frontotemporal dementia.
Gregory, C.A. and Hodges, J.R. (1996). Frontotemporal Neurology, 71: 1481–1488.
dementia use of consensus criteria and prevalence of Hutton, M., Lendon, C.L., Rizzu, P. et al. (1998). Association
psychiatric features. Neuropsychiatry, Neuropsychology of missense and 5'-splice-site mutations in tau with the
and Behavioural Neurology, 9: 145–153. inherited dementia FTDP-17. Nature, 18: 702–705.
Gregory, C., Lough, S., Stone, V. et al. (2002). Theory of Ikeda, M., Brown, J., Holland, A.J. et al. (2002). Changes
mind in patients with frontal variant frontotemporal in appetite, food preference, and eating habits in
dementia and Alzheimer’s disease: Theoretical and frontotemporal dementia and Alzheimer’s disease.
practical implications. Brain: A Journal of Neurology, Journal of Neurology, Neurosurgery, and Psychiatry,
125: 752–764. 73: 371–376.
Pick’s disease: Its relationship to progressive aphasia, semantic dementia and frontotemporal dementia 767

Ingram, E.M. and Spillantini, M.G. (2002). Tau gene muta- Kitchener, E. and Hodges, J.R. (1999). Impaired knowl-
tions: Dissecting the pathogenesis of FTDP-17. Trends edge of famous people and events and intact
in Molecular Medicine, 8 (12): 555–562. autobiographical knowledge in a case of progres-
Irish, M., Hornberger, M., Lah, S. et al. (2011). Profiles of sive right temporal lobe degeneration: Implications
recent autobiographical memory retrieval in seman- for the organization of remote memory. Cognitive
tic dementia, behavioural-variant frontotemporal Neuropsychology, 16: 589–607.
dementia, and Alzheimer’s Disease. Neuropsychologia, Knibb, J.A., Xuereb, J.H., Patterson, P. et al. (2006).
49: 2694–2702. Clinical and pathological characterization of progres-
Josephs, K.A., Duffy, J.R., Strand, E.A. et al. (2006). sive aphasia. Annals of Neurology, 59: 156–165.
Clinicopathological and imaging correlates of progres- Knopman, D.S., Mastri, A.R., Frey, W.H. et al. (1990).
sive aphasia and apraxia of speech. Brain: A Journal of Dementia lacking distinctive histological features:
Neurology, 129: 1385–1398. A common non-Alzheimer degenerative disease.
Josephs, K.A., Hodges, J.R., Snowden, J. et al. (2011). Neurology, 40: 251–256.
Neuropathological background of phenotypi- Kumfor, F., Irish, M., Hodges, J.R. et al. (2013). Discrete
cal variability in frontotemporal dementia. Acta neural correlates for the recognition of basic emotions
Neuropathology, 122: 137–153. in frontotemporal dementia. PLoS ONE, 8: e67457.
Katzman, R. (1986). Differential diagnosis of dementing Leyton, C.E., Villemagne, V.L., Savage, S. et al. (2011).
illnesses. Neurologic Clinics, 4: 329–340. Subtypes of progressive aphasia: Application of the
Keane, J., Calder, A.J., Hodges, J.R. and Young, A.W. International Consensus Criteria and validation using
(2002). Face and emotion processing in frontal variant β-amyloid imaging. Brain: A Journal of Neurology,
frontotemporal dementia. Neuropsychologia, 40 (6): 134: 3030–3043.
655–665. Lillo, P., Garcin, B., Bak, T. et al. (2010). Neurobehavioral
Kertesz, A. (2003). Pick’s complex and FTDP-17. features in frontotemporal dementia with amyo-
Movement Disorders, 18 (Suppl. 6): S57–S62. trophic lateral sclerosis. Archives of Neurology,
Kertesz, A., Davidson, W. and Fox, H. (1997). Frontal behav- 67: 826–830.
ioral inventory: Diagnostic criteria for frontal lobe demen- Lillo, P., Hodges, J.R. (2009). Frontotemporal demen-
tia. Canadian Journal of Neurological Sciences, 24: 29–36. tia and motor neuron disease: Overlapping
Kertesz, A., Davidson, W., McCabe, P. and Munoz, clinico-­pathological disorders. Journal of Clinical
D. (2003a). Behavioral quantitation is more sensitive than Neuroscience, 16: 1131-1135.
cognitive testing in frontotemporal dementia. Alzheimer Lillo, P., Mioshi, E., Zoing, M. et al. (2011). How common
Disease and Associated Disorders, 17: 223–229. are behavioral changes in amyotrophic lateral sclero-
Kertesz, A. and Munoz, D.G. (1998). Pick’s Disease and sis? Amyotrophic Lateral Sclerosis, 12: 45–51.
Pick Complex. New York, NY: Wiley-Liss Inc. Ling, H., O’ Sullivan, S.S., Holton, J.L. et al. (2010). Does
Kertesz, A., Nadkarni, N., Davidson, W. and Thomas, corticobasal degeneration exist? A clinicopatho-
A.W. (2000). The frontal behavioral inventory in the differ- logical re-evaluation. Brain: A Journal of Neurology,
ential diagnosis of frontotemporal dementia. Journal of 133: 2045–2057.
the International Neuropsychological Society, 6: 460–468. Lippa, C.F., Cohen, R., Smith, T.W. and Drachman,
Kertesz, A., Munoz, D.G. and Hillis, A. (2003b). Preferred D.A. (1991). Primary progressive aphasia with focal
terminology. Annals of Neurology, 54 (Suppl. 5): S3–S6. neuronal achromasia. Neurology, 41: 882–886.
Kipps, C.M., Hodges, J.R., Fryer, T.D. and Nestor, Mackenzie, I.R., Rademakers, R. and Neumann, M. (2010).
P.J. (2009a). Combined magnetic resonance imag- TDP-43 and FUS in amyotrophic lateral sclerosis and
ing and positron emission tomography brain imaging frontotemporal dementia. The Lancet Neurology,
in behavioural variant frontotemporal degeneration: 9 (10): 995–1007.
Refining the clinical phenotype. Brain: A Journal of Majounie, E., Renton, A.E., Mok, A. et al. (2012).
Neurology, 132: 2566–2578. Frequency of the C9orf72 hexanucleotide repeat
Kipps, C.M., Nestor, P.J., Acosta-Cabronero, J. et al. expansion in patients with amyotrophic lateral scle-
(2009b). Understanding social dysfunction in the rosis and frontotemporal dementia: A cross-sectional
behavioural variant of frontotemporal dementia: study. The Lancet Neurology, 11: 323–330.
The role of emotion and sarcasm processing. Brain: Mansvelt, J.V. (1954). Pick’s disease: A syndrome of lobar
A Journal of Neurology, 132: 592–603. cerebral atrophy, its clinico-anatomical and histopath-
Kipps, C.M., Nestor, P.J., Dawson, C.E. et al. (2008) ological types. Thèse. Utrecht University, Utrecht, The
Measuring progression in frontotemporal dementia: Netherlands.
Implications for therapeutic interventions. Neurology, Mathuranath, P.S., Xuereb, J.H., Bak, T. and Hodges,
70: 2046–2052. J.R. (2000). Corticobasal ganglionic degeneration and/
Kipps, C.M., Nestor, P.J., Fryer, T.D., Hodges, J.R. et al. or frontotemporal dementia? A report of two overlap
(2007). Behavioural variant frontotemporal dementia: cases and review of literature. Journal of Neurology,
Not all it seems? Neurocase, 13: 237–247. Neurosurgery, and Psychiatry, 68: 304–312.
768 Dementia

McKhann, G.M., Albert, M.S., Grossman, M. et al. Neary, D., Snowden, J.S., Gustafson, L. et al. (1998).
(2001). Clinical and pathological diagnosis of fron- Frontotemporal lobar degeneration: A consen-
totemporal dementia report of the Work Group on sus on clinical diagnostic criteria. Neurology,
Frontotemporal Dementia and Pick’s Disease. Archives 51: 1546–1554.
of Neurology, 58: 1803–1809. Neary, D., Snowdon, J.S., Mann, D.M.A. et al. (1990).
Mesulam, M.M. (1982). Slowly progressive aphasia Frontal lobe dementia and motor neuron disease.
without generalised dementia. Annals of Neurology, Journal of Neurology, Neurosurgery, and Psychiatry, 53:
11: 592–598. 23–32.
Mesulam, M.M. and Weintraub, S. (1992). Primary pro- Nestor, P.J., Graham, N.L., Fryer, T.D. et al. (2003).
gressive aphasia. In Boller, F. (ed.), Heterogeneity of Progressive non-fluent aphasia is associated with
Alzheimer’s Disease. Berlin, Germany: Springer-Verlag, hypometabolism centred on the left anterior insula.
pp. 43–66. Brain: A Journal of Neurology, 126: 2406–2418.
Mesulam, M., Wicklund, A., Johnson, N. et al. (2008). Neumann, M., Sampathu, D.M., Kwong, L.K. et al. (2006).
Alzheimer and frontotemporal pathology in subjects Ubiquitinated TDP-43 in frontotemporal lobar degen-
of primary progressive aphasia. Annals of Neurology, eration and amyotrophic lateral sclerosis. Science,
63: 709–719. 314: 130–133.
Miller, B.L., Cummings, J.L., Villanueva-Meyer, J. et al. (1991). Nyatsanza, S., Shetty, T., Gregory, C. et al. (2003). A study
Frontal lobe degeneration: Clinical, neuropsychological, of stereotypic behaviours in Alzheimer’s disease and
and SPECT characteristics. Neurology, 41: 1374–1382. frontal and temporal variant frontotemporal dementia.
Miller, B.L., Darby, A., Benson, D.F. et al. (1997). Journal of Neurology, Neurosurgery, and Psychiatry,
Aggressive, socially disruptive and antisocial behav- 74: 1398–1402.
iour associated with frontotemporal dementia. British Onari, K. and Spatz, H. (1926). Anatomische beitrage zur
Journal of Psychiatry, 170: 150–155. lehre von der Pickschen umschriebenen grosshirnrin-
Mingazzini, G. (1914). On aphasia due to atrophy of the denatrophie (Piscksche Krankheit). Zeitschrift für die
cerebral convolutions. Brain: A Journal of Neurology, Gesamte Neurologie und Psychiatrie, 101: 470–511.
36: 493–524. Pasquier, F., Lebert, F. and Scheltens, P. (1996).
Mioshi, E. and Hodges, J.R. (2007). Activities of daily living Frontotemporal Dementia. The Netherlands: ICG
in frontotemporal dementia and Alzheimer disease – Publications.
Reply. Neurology, 69: 2110–2110. Petersen, R.B., Tabaton, M., Chen, S.G. et al. (1995).
Mioshi, E., Kipps, C.M., Dawson, K. et al. (2007). Activities Familial progressive subcortical gliosis: Presence of
of daily living in frontotemporal dementia and prions and linkage to chromosome 17. Neurology,
Alzheimer disease. Neurology, 68: 2077–2084. 45: 1062–1067.
Mitsuyama, Y. and Takamiya, S. (1979). Presenile dementia Pick, A. (1892). Über die beziehungen der senilen atro-
with motor neuron in Japan. Archives of Neurology, phie zur aphasie. Prager Medizinische Wochenschrift,
36: 592–593. 17: 165–167.
Morris, J.C., Cole, M., Banker, B.Q., Wright, D et al. (1984). Pick, A. (1901). Senile hirnatrophie als Grundlage für
Hereditary dysphasic dementia and the Pick-Alzheimer hernderscheinungen. Wiener Klinische Wochenschrift,
spectrum. Annals of Neurology, 16: 455–466. 14: 403–404.
Morris, H.R., Khan, M.N., Janssen, J.C. et al. (2001). The Pick, A. (1906). Über einen weiteren symptomenkom-
genetic and pathological classification of familial plex im Rahmen der dementia senilis, bedingt durch
frontotemporal dementia. Archives of Neurology, umschriebene starkere hirnatrophie. Monatschrift für
58: 1813–1816. Psychiatrie und Neurologie, 19: 97–108.
Morris, H.R., Osaki, Y., Holton, J. et al. (2003). Tau exon 10 Piguet, O., Hornberger, M., Mioshi, E. et al. (2011).
1 16 mutation FTDP-17 presenting clinically as sporadic Behavioural-variant frontotemporal dementia:
young onset PSP. Neurology, 61: 102–104. Diagnosis, clinical staging and management. The
Mummery, C.J., Patterson, K., Price, C.J. et al. (2000). Lancet Neurology, 10: 162–172.
A voxel based morphometry study of semantic Poorkaj, P., Bird, T.D., Wijsman, E. et al. (1998). Tau is a
dementia. The relationship between temporal lobe candidate gene for chromosome 17 frontotemporal
atrophy and semantic dementia. Annals of Neurology, dementia. Annals of Neurology, 43: 815–825.
47: 36–45. Rahman, S., Sahakian, B.J., Hodges, J.R. et al. (1999).
Mummery, C.J., Patterson, K., Wise, R.J.S. et al. (1999). Specific cognitive deficits in mild frontal variant fron-
Disrupted temporal lobe connections in semantic totemporal dementia. Brain: A Journal of Neurology,
dementia. Brain: A Journal of Neurology, 122: 61–73. 122: 1469–1493.
Neary, D., Snowden, J.S., Bowen, D.M. et al. (1986). Rakowicz, Z. and Hodges, J.R. (1998). Dementia and apha-
Neuropsychological syndromes in presenile demen- sia in motor neurone disease: An under recognised
tia due to cerebral atrophy. Journal of Neurology, association. Journal of Neurology, Neurosurgery, and
Neurosurgery, and Psychiatry, 49: 163–174. Psychiatry, 65: 881–889.
Pick’s disease: Its relationship to progressive aphasia, semantic dementia and frontotemporal dementia 769

Rankin, K.P., Salazar, A., Gorno-Tempini, M.L. et al. (2009). Soliveri, P., Rossi, G., Monza, D. et al. (2003). A case of
Detecting sarcasm from paralinguistic cues: Anatomic dementia parkinsonism resembling progressive supra-
and cognitive correlates in neurodegenerative disease. nuclear palsy due to mutation in the tau protein gene.
Neuroimage, 47: 2005–2015. Archives of Neurology, 60: 1454–1456.
Rascovsky, K.J., Hodges, J.R., Kipps, C.M. et al. (2007). Spillantini, M.G., Bird, T.D. and Ghetti, B. (1998a).
Diagnostic criteria for the behavioral variant of fron- Frontotemporal dementia and parkinsonism linked to
totemporal dementia (bvFTD): Current limitations and chromosome 17. A new group of tauopathies. Brain
future directions. Alzheimer Associated Disorders, Pathology, 8: 387–402.
21: S14–S18. Spillantini, M.G., Goedert, M., Crowther, R.A. et al.
Rascovsky, K., Hodges, J.R., Knopman, D. et al. (2011). (1997). Familial multiple system tauopathy with pre-
Sensitivity of revised diagnostic criteria for the senile dementia: A disease with abundant neuronal
behavioural variant of frontotemporal dementia. Brain: and glial tau filaments. Proceedings of the National
A Journal of Neurology, 134: 2456–2477. Academy of Sciences of the United States of America,
Rascovsky, K, Salmon, D.P., Ho, G.J. et al. (2002). 94 (8): 4113–4118.
Cognitive profiles differ in autopsy-confirmed Spillantini, M.G., Murrell, J.R., Goedert, M. et al. (1998b).
frontotemporal dementia and AD. Neurology, Mutation in the tau gene in familial multiple system
58: 1801–1808. tauopathy with presenile dementia. Proceedings of the
Ratnavalli, E., Brayne, C., Dawson, K. et al. (2002). The National Academy of Sciences of the United States of
prevalence of frontotemporal dementia. Neurology, America, 95: 7737–7741.
58: 1615–1621. Steele, J.C., Richardson, J.C. and Olszewski, J. (1964).
Rebeiz, J.J., Kolodny, E.H. and Richardson, E.P. (1968). Progressive supranuclear palsy. A heterogeneous
Corticodentatonigral degeneration with neuronal degeneration involving the brain stem, basal ganglia
achromasia. Archives of Neurology, 18: 20–33. and cerebellum with vertical gaze and pseudobul-
Renton, A.E., Majounie, E., Waite, A. et al. (2011). bar palsy, nuchal dystonia and dementia. Archives of
A hexanucleotide repeat expansion in C9orf72 is the Neurology, 10: 333–359.
cause of chromosome 9p21-linked ALS-FTD. Neuron, Strong, M.J., Lomen-Hoerth, C., Caselli, R.J. et al. (2003).
72: 257–268. Cognitive impairment, frontotemporal dementia, and
Rogers, R.D., Sahakian, B.J., Hodges, J.R. et al. (1998). the motor neuron diseases. Annals of Neurology,
Dissociating executive mechanisms of task control fol- 54 (Suppl. 5): S20–S23.
lowing frontal lobe damage and Parkinson’s disease. Sumi, S.M., Bird, T.D., Nochlin, D. et al. (1992). Familial
Brain: A Journal of Neurology, 121: 815–842. presenile dementia with psychosis associated with
Rosen, H.J., Gorno-Tempini, M.L., Goldman, W.P. et al. cortical neurofibrillary tangles and degeneration of the
(2002a). Patterns of brain atrophy in frontotempo- amygdala. Neurology, 42: 120–127.
ral dementia and semantic dementia. Neurology, Tanabe, H., Ikeda, M., Nakagawa, Y. et al. (1992). Gogi
58: 198–208. (word meaning) aphasia and semantic memory for
Rosen, H.J., Hartikainen, K.M., Jagust, W. et al. (2002b). words. Higher Brain Function Research, 12: 153–169.
Utility of clinical criteria in differentiating frontotem- Thompson, S.A., Graham, K.S., Williams, G. et al. (2004).
poral lobar degeneration (FTLD) from AD. Neurology, Dissociating person-specific from general semantic
58: 1608–1615. knowledge: Roles of the left and right temporal lobes.
Saffran, E.M. and Schwartz, M.F. (1994). Of cabbages and Neuropsychologia, 42: 359–370.
things: Semantic memory from a neuropsychologi- Thompson, S.A., Patterson, K. and Hodges, J.R. (2003).
cal perspective – A tutorial review. In Umilta, C. and Left/right asymmetry of atrophy in semantic demen-
Moscovitch, M. (eds.), Attention and Performance XV. tia: Behavioral-cognitive implications. Neurology,
Hove and London: Lawrence Erlbaum, pp. 507–536. 61 (9): 1196–1203.
Schenk, V.W.S. (1951). Maladie de Pick: Etude anatomo- Tissot, R., Constantanidis, J. and Richard, J. (1975).
clinique de 8 cas. Annales Medico-Psychologiques La Maladie de Pick. Paris, France: Masson.
(Paris), 109: 574–587. Torralva, T., Kipps, C.M., Hodges, J.R. et al. (2007). The
Schneider, C. (1927). Über Picksche krankheit. relationship between affective decision-making
Monatschrift für Psychologie und Neurologie, and theory of mind in the frontal variant of fronto-­
65: 230–275. temporal dementia. Neuropsychologia, 45:342–349.
Snowden, J.S., Goulding, P.J. and Neary, D. (1989). Tulving, E. (1972). Episodic and semantic memory. In
Semantic dementia: A form of circumscribed cerebral Tulving, E. and Donaldson, W. (eds.), Organization of
atrophy. Behavioural Neurology, 2: 167–182. Memory. New York, NY: Academic Press, pp. 381–403.
Snowden, J.S., Neary, D. and Mann, D.M.A. (1996). Tulving, E. (1983). Elements of Episodic Memory. New
Fronto-temporal Lobar Degeneration: Fronto-temporal York, NY: Oxford University Press.
Dementia, Progressive Aphasia, Semantic Dementia.
New York, NY: Churchill Livingstone.
770 Dementia

Warrington, E.K. (1975). Selective impairment of seman- Wilhelmsen, K.C., Lynch, T., Pavlou, E. et al. (1994).
tic memory. Quarterly Journal of Experimental Localization of disinhibition-dementia-parkinsonism-
Psychology, 27: 635–657. amyotrophy complex to 17q21–22. American Journal
Wedderburn, C., Wear, H., Brown, B. et al. (2008). of Human Genetics, 55: 1159–1165.
The utility of the Cambridge Behavioural Williams, G.B., Nestor, P.J. and Hodges, J.R. (2005).
Inventory in neurodegenerative disease. Journal The neural correlates of semantic and behavioural
of Neurology, Neurosurgery, and Psychiatry, deficits in frontotemporal dementia. Neuroimage,
79: 500–503. 24: 1042–1051.
Weintraub, S., Rubin, N.P. and Mesulam, M-M. (1990). Wszolek, Z.K., Pfeiffer, R.F., Bhatt, M.H. et al. (1992).
Primary progressive aphasia: Longitudinal course, Rapidly progressive autosomal dominant parkinsonism
profile, and language features. Archives of Neurology, and dementia with pallido-ponto-nigral degeneration.
47: 1329–1335. Annals of Neurology, 32: 312–320.
67
The genetics and molecular pathology of
frontotemporal lobar degeneration

DAVID M.A. MANN

the early identification of the ‘classic’ inclusion bodies (Pick


67.1 INTRODUCTION bodies) and swollen neurones (Pick cells) (Pick, 1892), and
the characteristic, though non-specific, features of micro-
The term frontotemporal lobar degeneration (FTLD) is used vacuolation of outer cortical laminae marking the site of the
here to describe a clinically and pathologically heteroge- principal molecular changes (Knopman et al., 1990).
neous group of non-Alzheimer forms of dementia arising The recognition in about 40% of patients of a previous
from degeneration of the frontal and temporal lobes. The family history of a similar disorder consistent with autosomal
prototypical, and most common, clinical syndrome is fron- dominant inheritance (Stevens et al., 1998; Rosso et al., 2003)
totemporal dementia (FTD), manifesting as behavioural paved the way for the breakthroughs in knowledge and under-
and personality changes, involving disinhibition, stereotypy, standing of the molecular pathology of the disorder that have
unsocial acts and language disorder, leading to apathy, mut- come through the wealth of genetic investigations performed
ism and late neurological (frontal release or extrapyramidal) over the past decade. By 1997, genetic linkage had identified a
signs (Neary et al., 1998, 2005, 2007). However, the spectrum locus on the long arm of chromosome 17 (17q21–22) in some
of illness is wider, involving cases where movement disorder, families, and because parkinsonism usually accompanied the
with relatively mild dementia, is the major disabling feature dementia, these cases came to be snappily described as ‘fronto-
(Cordes et al., 1992; Wszolek et al., 1992; Lynch et al., 1994) temporal dementia and parkinsonism linked to chromosome
or others with the linguistic disorders of semantic dementia 17’ (FTDP-17) (Foster et al., 1997). In 1998, mutational events
(SD) and progressive non-fluent aphasia (PNFA) (Snowden in the tau gene, MAPT, were identified (Hutton et al., 1998;
et al., 1992; Neary et al., 1998, 2005, 2007). FTD can be Poorkaj et al., 1998; Spillantini et al., 1998). The prevalence of
accompanied by clinical motor neurone disease (MND) in MAPT mutations within FTLD varies between 6% and 18%
about 10% of cases giving the syndrome of frontotemporal depending on the population studied (Dumanchin et al., 1998;
dementia with motor neurone disease (FTD1MND) (Neary Rizzu et al., 1999; Houlden et al., 1999a; Kowalska et al., 2001;
et al., 1998, 2005, 2007). Onset of illness is usually before Poorkaj et al., 2001; Binetti et al., 2003; Rosso et al., 2003).
65 years of age, with duration being between 6 and 9 years, Nonetheless, despite exhaustive genetic analyses, there
though up to 15 years is not uncommon (Neary et al., 1998; remained a number of linked families in whom no muta-
Rosso et al., 2003; Neary et al., 2005, 2007). tions in MAPT could be found. This paradox was resolved
Outward inspection of the brain in FTLD gives few clues in 2006 when mutations within the progranulin gene
to pathogenesis, but can be a useful index of clinical pre- (PGRN), located just 1.8 cm from MAPT, were identified
sentation. Hence, FTD is associated with bilateral atrophy (Baker et al., 2006; Cruts et al., 2006). Collectively, MAPT
of the frontal and anterior temporal lobes and SD is asso- and PGRN mutations likely account for no more than 20%
ciated with bitemporal lobe atrophy, often asymmetric, of all cases of FTLD and less than 50% of all autosomal-
but usually favouring the left side. PNFA is associated with dominant inherited forms; further causative mutations and
marked asymmetry, most often of the left cerebral hemi- genetic risk factors remain to be defined or confirmed.
sphere (Neary et al., 2005, 2007). Routine histology has con- Over the past decade, numerous immunohistochemical
tributed little to our understanding of the disorder, beyond studies have identified aggregated or accumulated proteins

771
772 Dementia

within the brain in neurodegenerative diseases not only to produce an excess of 4R tau, and hence, the insoluble tau
improve diagnostic power and criteria but also to under- aggregates are composed predominantly of 4R tau isoforms
stand better the basic pathology and devise experimental (Clark et al., 1998; Hong et al., 1998; Hasegawa et al., 1999;
approaches to enhance knowledge of pathogenesis. About Pickering-Brown et al., 2002; De Silva et al., 2006), appear-
45% of FTLD cases display insoluble tau proteins in their ing as neurofibrillary tangle-like structures within large
brains in the form of intraneuronal neurofibrillary tangles and smaller pyramidal cells of cortical layers III and V and
or Pick bodies; about half are associated with MAPT muta- prominent within glial cells in the deep white matter, globus
tions (Taniguchi et al., 2004; Shi et al., 2005). However, a pallidus and internal capsule (Pickering-Brown et al., 2002).
tau-negative, but ubiquitin-positive pathology, known as Neurones within subcortical structures, such as corpus stri-
FTLD-U, accounts for the other 55% of cases (Lipton et al., atum, medial thalamus, nucleus basalis of Meynert, dorsal
2004; Taniguchi et al., 2004; Mott et al., 2005; Shi et al., raphe, pontine and dentate nuclei, locus caeruleus and sub-
2005; Forman et al., 2006a); in most, the ubiquitinated tar- stantia nigra can likewise be affected, but less severely so
get protein is the transactive response (TAR) DNA-binding (Pickering-Brown et al., 2002).
protein, TDP-43.

67.3 
PGRN AND TDP-43 PATHOLOGY
67.2 THE TAU GENE (MAPT) AND TAU
PATHOLOGY IN FTLD
67.3.1 PGRN MUTATIONS IN FTLD
67.2.1 THE STRUCTURE OF MAPT AND MAPT mutations account for only 10% of FTLD cases at
THE FUNCTION OF TAU PROTEINS most (Rizzu et al., 1999; Houlden et al., 1999a; Poorkaj et al.,
2001; Rosso et al., 2003), although the frequency may reach
The tau gene (MAPT) has 15 coding regions (exons), and 20% when only cases consistent with autosomal dominance
transcripts are alternatively spliced to produce six different are considered (Rizzu et al., 1999). Indeed, following the
isoforms ranging from 352 to 441 amino acids in length. identification of MAPT mutations, there remained at least
Tau is normally located in axons and regulates microtu- five chromosome 17-linked families for which no mutational
bule assembly/disassembly and axonal transport of proteins event in MAPT could be determined. Moreover, no insoluble
and organelles. All six isoforms play a role in maintenance aggregates of tau were seen biochemically or histopatho-
of microtubular structure. If one or more fails, or if there is a logically (Foster et al., 1997; Rosso et al., 2001; Rademakers
stoichiometric imbalance in the different variants, microtu- et al., 2002; Froelich Fabre et al., 2003), although ubiquitin-
bule formation will become more difficult or the stability of positive inclusions were variably present (Rosso et al., 2001;
microtubules formed will become compromised. Any excess Rademakers et al., 2002; Froelich Fabre et al., 2003). Such
of tau (of any isoform composition) can be bundled into cases became known as FTLD-ubiquitinated (FTLD-U). In
indigestible residues (neurofibrillary tangles or Pick bodies) 2006, causative mutations in these families were identified
that choke the cell, but may also induce neurotoxicity. (Baker et al., 2006; Cruts et al., 2006). Many subsequent stud-
ies identified mutations in PGRN, with 66 different muta-
67.2.2 MAPT MUTATIONS IN FTLD tions within 199 families recorded to date (see AD and FTD
mutation database: wwww.molgen.ua.ac.be/FTDmutations).
To date, about 44 MAPT mutations in around 131 families PGRN mutations in FTLD include missense mutations
have been identified (see Alzheimer’s disease [AD] and FTD generating premature stop codons, insertion or deletion
mutation database: wwww.molgen.ua.ac.be/FTDmutations). mutations resulting in frameshifts, or changes within ini-
Typically, patients with MAPT mutations display FTD with tiation codons precluding transcription (Baker et al., 2006;
early signs of disinhibition, loss of initiative, stereotypic Cruts et al., 2006). Some are located within or close to splice
behaviours and linguistic changes of a semantic nature donor sites, resulting in splicing out of particular exons
(Pickering-Brown et al., 2002), although other patients dis- (Baker et al., 2006; Cruts et al., 2006; Masellis et al., 2006;
play a phenotype resembling progressive supranuclear palsy Le Ber et al., 2007). Missense mutations in exon 0 lead to
(PSP) or corticobasal degeneration (CBD) (Bugiani et al., nuclear degradation, and mutations that affect the Kozak
1999; Wszolek et al., 2001). sequence prevent translation (Cruts et al., 2006; Le Ber et al.,
2007). One particular mutation, Ala9Asp, affects the hydro-
67.2.3 PATHOLOGY AND BIOCHEMISTRY phobic core of the signal peptide sequence disrupting the
normal insertion of the protein into the endoplasmic reticu-
All cases with MAPT mutations are characterized by the lum leading to a trapping of the (transcribed and translated)
deposition of insoluble, aggregated tau proteins within neu- mutated protein within the Golgi apparatus (Mukherjee
rones and glial cells in the cerebral cortex and other brain et al., 2006). Whole deletions of PGRN occur, but only
regions. Nevertheless, there are two histological patterns: rarely (Gijselinck et al., 2008; Rovelet-Lecrux et al., 2008).
those mutations in and around the stem loop structure Nonetheless, all mutations, irrespective of type, ultimately
The genetics and molecular pathology of frontotemporal lobar degeneration 773

generate the same functional effect – a null allele – with at changes in FTLD are not reflected by changes in biochem-
least 50% loss of translated protein giving haploinsufficiency. istry or structure of PGRN, but are witnessed as brain
Most transcripts are immediately destroyed through non- accumulations of ubiquitinated protein devoid of PGRN,
sense-mediated decay, and it is unlikely that any, or perhaps principally in the cerebral cortex and hippocampus, as
only a small proportion, are translated into mutated proteins, neuronal cytoplasmic inclusion (NCI) bodies or dystro-
thereby explaining the lack of mutated PGRN protein within phic neurites (DNs) (Baker et al., 2006; Cruts et al., 2006).
the ubiquitinated lesions (Baker et al., 2006). The biological Neuronal intranuclear inclusions (NIIs) of a ‘cat’s eye’ or
effects would, therefore, seem to be ones of a ‘loss of function ‘lentiform’ appearance have been described (Baker et al.,
effect’, although the brain accumulation of abnormal ubiqui- 2006; Cruts et al., 2006; Pickering-Brown et al., 2006;
tinated proteins may also confer a neurotoxic ‘gain of func- Snowden et al., 2006; Mackenzie et al., 2006b; Snowden
tion’, or some combination of the two is possible. et al., 2007; Pickering-Brown et al., 2008), although these
In contrast to MAPT, PGRN mutations may not be fully occur in FTLD-U cases where no PGRN mutation has been
penetrant. In one family (DR8), several unaffected car- determined (Davidson et al., 2007). The ubiquitinated NCI
riers have been reported to live well beyond the obligate and DN contain TDP-43 (Arai et al., 2006; Neumann et al.,
onset age for the mutation, one as late as 81 years (Cruts 2006). TDP-43 is a 43-kDa nuclear protein, 414 amino
et al., 2006). In R493X mutation, there is delayed onset age acids long, first identified as a binding partner to the TAR
in patients carrying the rare rs9897528 on the wild-type DNA element of the human immunodeficiency virus. The
PGRN allele (Rademakers et al., 2007), suggesting genetic TDP-43 gene (TARDBP), located on chromosome 1p36.2,
variability on the wild-type allele might influence the age- contains six exons and is ubiquitously expressed. It has
related disease penetrance of PGRN mutations. Bruni et al. two highly conserved RNA recognition motifs (RRM1 and
(2007) also reported a potential influence of the wild-type RRM2) and a C-terminal, glycine-rich tail, which medi-
allele upon age-associated disease penetrance. Common ates protein–protein interactions, including interactions
polymorphisms on the normal allele may impact on disease with other heterogeneous ribonuclear protein (hnRNP)
expression through (further) lowering of PGRN protein. family members. Under physiological conditions, TDP-43
Conversely, MAPT haplotype or apolipoprotein E (ApoE) is mostly present within the nucleus although low levels
genotype does not influence either age at onset or disease occur within the cytoplasm (Ayala et al., 2008; Buratti
penetrance (Cruts et al., 2006; Gass et al., 2006; Bruni et al., and Baralle, 2008; Thorpe et al., 2008; Turner et al., 2008;
2007; Rademakers et al., 2007). Winton et al., 2008). It is believed that TDP-43 shuttles
continuously between nucleus and cytoplasm, a process
67.3.2 THE STRUCTURE AND FUNCTION regulated by nuclear localization and nuclear export signal
OF PROGRANULIN motifs. Ultrastructural studies show TDP-43 is enriched
within euchromatin domains, specifically within peri-
PGRN contains 13 exons and encodes a full-length protein chromatin fibrils (Casafont et al., 2009), which are nuclear
(predicted molecular weight of 68 kDa) secreted as a glyco- sites of transcription and transcriptional splicing. TDP-43
sylated 85-kDa precursor that can be cleaved by elastase- may, therefore, function as a transcription repressor or as
like activity into a series of 6-kDa cysteine-rich fragments an initiator of exon skipping in the alternative splicing of
called granulins (GRNs) (Zhu et al., 2002; He et al., 2003). messenger RNA (mRNA) (Wang et al., 2004; Buratti and
Both PGRN and the GRNs are biologically active with roles Baralle, 2008).
in tissue remodelling processes (Ahmed et al., 2007; van Most cases of FTLD-U, however, are not associated
Swieten and Heutink, 2008). In peripheral tissues, PGRN with PGRN mutations, although the majority of these still
plays a role in development, wound repair and inflamma- display TDP-43 pathological changes. There are qualita-
tion, by activating signalling cascades that control cell cycle tive differences in the relative burdens of TDP-43 immu-
progression and cell motility (He and Bateman, 2003). In noreactive NCI, DN and NII across the FTLD-U cases,
the brain, PGRN is expressed in both neurones and microg- and four major histological types have been indepen-
lia. Reduced levels of this secreted mitogenic factor induce dently described (Sampathu et al., 2006; Mackenzie et al.,
neurodegeneration in FTLD, indicating an important role 2006a), now unified under a single classification (Cairns
in neuronal survival. As with VCP and CHMP2B mutations, et al., 2007a; Mackenzie et al., 2009). In this, type 1 histol-
loss of PGRN may be associated with defects in protein deg- ogy describes cases with a preponderance of DN within
radation through cytoplasmic organelles (Ahmed et al., neocortical regions with rounded, solid (Pick body-like)
2007). Suppression of PGRN expression results in caspase- NCI within the hippocampus dentate gyrus (DG) gran-
dependent cleavage of TDP-43, with cellular accumulation ule cells. Type 2 histology describes cases with a pre-
of C-terminal fragments (Zhang et al., 2007). ponderance of neuronal NCI and few, if any, DN within
the neocortex, with more granular NCI within the hip-
67.3.3 PATHOLOGY OF PGRN MUTATIONS pocampus. In type 3 histology, numerous NCI and DN
are present within the neocortex, and in many cases len-
Consistent with observations that PGRN mutations do not tiform NII are seen. Granular NCI are again seen within
result in the translation of mutated proteins, pathological the hippocampus.
774 Dementia

These histological variations are not without clinical tau-negative, TDP-43-negative FTLD cases, the ubiquiti-
significance. Type 1 histology is common in SD, whereas nated inclusion bodies are immunoreactive to a protein
type 2 histology describes FTD1MND. Type 3 histology is called fused-in sarcoma (FUS) (also known as translocation
present in PGRN mutation, but not exclusively so, and can in liposarcoma [TLS]), and may be part of a newly identified
describe either a ‘straightforward’ FTD presentation or ‘family’ of proteinopathies, termed ‘FUSopathies’ (Munoz
often, PNFA (Mackenzie et al., 2006b; Davidson et al., 2007; et al., 2009; Neumann et al., 2009a, 2009b). In the context
Josephs et al., 2009). In addition to these diagnostically of FTLD, these should be known as FTLD-FUS (Mackenzie
characteristic changes in cerebral cortex and hippocampus, et al., 2009).
similar changes are widespread throughout other neocorti-
cal regions, including insular and anterior parietal cortex 67.4.3 PATHOGENETIC CONSIDERATIONS
and cingulate gyrus; posterior parietal and occipital cortex
are rarely affected. Non-cortical regions, such as amygdala TDP-43 and FUS are both transcription factors involved
and corpus striatum are frequently and severely affected, in the transport of mRNA to dendritic spines for local
although thalamus, substantia nigra and inferior olives are ­translation in relation to synaptic activity. This process
variably and mildly involved (Brandmeir et al., 2008; Geser requires the involvement of several types of RNA-containing
et al., 2008a; Davidson et al., 2009; Josephs et al., 2009). ­granules – ribonuclear protein particles (RNP), process-
Because of the regional variations in distribution of TDP-43 ing bodies (PBs) and stress granules (SGs) (Bramham and
pathological changes throughout the neuraxis, these histo- Wells, 2007). However, while TDP-43 appears to be associ-
logical subtypes might represent distinct clinicopathological ated with PB, FUS relates to SG (Bramham and Wells, 2007;
entities with separate pathogenesis (Josephs et al., 2009). Wang et al., 2008). PB are associated with mRNA decay
When TDP-43 NCI are present in the cell, TDP-43 processes, and decapping enzyme 1 protein – a marker
immunostaining is absent from the nucleus, possibly hav- for PB – is absent from NCI in basophilic inclusion body
ing been sequestered into NCI. Perturbation of trafficking disease (BIBD) (Fujita et al., 2008), explaining the lack of
of TDP-43 between nucleus and cytoplasm may be a func- TDP-43 in such inclusions. In contrast, NCI in BIBD con-
tional consequence of its incorporation into these cyto- tain mRNA-binding proteins poly(A) binding protein 1 and
plasmic aggregates (Winton et al., 2008), although it is still T-cell intracellular antigen 1, indicating their association
unclear whether disease is due to a toxic gain of function with SG rather than RNP or PB (Fujita et al., 2008). Hence,
relating to these pathological aggregates, or from loss of TDP-43 and FUS play different, although complementary,
normal nuclear functions, or some combination of the two. roles in the processing of RNA.
While for purposes of classification it is logical to clump
aFTLD-U, NIFID and BIBD into the broad-brush category of
FTLD-FUS, this might hide mechanistic differences. The dis-
67.4 OTHER TAU AND TDP-43 orders clearly differ in neuropathology – the NCI in NIFID
NEGATIVE FTLD CASES and aFTLD-U do not contain RNA, vermiform NII are com-
monplace in NIFID and aFTLD-U but rare or absent in
67.4.1 PATHOLOGY BIBD, loss of lower motor neurones, giving MND phenotype,
is frequent in BIBD but less common in aFTLD-U (Munoz
Although about 95% of FTLD cases can be accounted by et al., 2009) – all features pointing to distinctions in patho-
tauopathy or TDP-43 proteinopathy, there still remains a genesis. The challenge will be to understand how changes in
small proportion of cases where alternative target proteins the metabolism of TDP-43 and FUS dictate patterns of neu-
are involved or remain undetermined. rodegeneration and clinical expression of disease.
In some of these, ubiquitinated, Pick body-like inclu-
sions are present in neurones of frontal and temporal cor-
tex, and basal ganglia. These are negative for tau, TDP-43
and α-synuclein but (variably) immunoreactive for all 67.5 OTHER TDP-43 PROTEINOPATHIES
class IV intermediate filaments (IFs), light, medium and
heavy neurofilament subunits (Bigio et al., 2003; Cairns TDP-43 pathological changes colocalize within the ubiqui-
et al., 2003; Josephs et al., 2003; Cairns et al., 2004a) and tinated NCI in various other neurodegenerative disorders.
α-internexin (Cairns et al., 2004b). Consequently, the dis- Such changes occur in 20%–30% patients with AD
order became known as neuronal intermediate filament (Amador-Ortiz et al., 2007; Hu et al., 2008; Uryu et al.,
inclusion disease (NIFID) and was classed as FTLD-IF 2008) or dementia with Lewy bodies (DLB) (Nakashima-
(Mackenzie et al., 2009). Yasuda et al., 2007; Arai et al., 2009), in some cases of CBD
(Uryu et al., 2008), PSP and in others with Parkinson’s dis-
67.4.2 FUSOPATHIES ease (Uryu et al., 2008) and parkinsonism–dementia com-
plex of Guam (Guam PDC) (Hasegawa et al., 2007; Geser
Very recent work (Munoz et al., 2009; Neumann et al., et al., 2008b), in argyrophilic grain disease, Perry syndrome
2009a, 2009b) has shown that in the majority of these (Farrer et al., 2009) and in familial British dementia (FBI)
The genetics and molecular pathology of frontotemporal lobar degeneration 775

(Schwab et al., 2009). TDP-43 protein has not been (consis- NCI within the hippocampal DG granule cells (Holm et al.,
tently) found in tangles in AD (Amador-Ortiz et al., 2007; 2007). Such cases are thus presently classified as FTLD-
Uryu et al., 2008), in Pick bodies (Davidson et al., 2007; UPS (Mackenzie et al., 2009). In some family members, a
Uryu et al., 2008, but see Arai et al., 2006; Freeman et al., few tau tangles are seen, without senile (amyloid) plaques
2008) or in the ubiquitinated inclusions of FTD-3 (Holm (Yancopoulou et al., 2003), but less commonly than is usual
et al., 2007). Elsewhere, TDP-43 pathological changes for cases of FTLD with MAPT mutations. To date, only four
occur in Rosenthal fibres and granular eosinophilic bod- families have been identified with this particular muta-
ies in l­ow-grade tumours and reactive brain tissue (but not tion (see AD and FTD mutation database: www.molgen.
in anoxic or ischaemic lesions) (Lee et al., 2008), indicat- ua.ac.be/FTDmutations) and the significance of this par-
ing pathological alterations in the structure or function of ticular genetic change to the broader pathogenesis of FTLD
TDP-43 may occur across a wide range of neurodegenera- remains unknown.
tive diseases and other disorders, and might result from a
variety of mechanistic disturbances. 67.6.3 OTHER GENETIC RISK FACTORS
FOR FTLD
Although it is well established that ApoE e4 allele is a risk
67.6 OTHER GENETIC CHANGES factor for late-onset sporadic and familial AD (Corder et al.,
1993), whether this plays a role in FTLD is still unclear.
67.6.1 CHROMOSOME 9 There have been many claims that ApoE e4 allele frequency
is elevated in FTLD (Czech et al., 1994; Frisoni et al., 1994;
Linkage to chromosome 9p in several families clinically Farrer et al., 1995; Schneider et al., 1995; Stevens et al., 1997;
sharing an FTD and MND phenotype has been claimed Fabre et al., 2001), although other (usually larger) studies
(Hosler et al., 2000; Morita et al., 2006; Vance et al., 2006), generally have not substantiated this in either Caucasian
but not confirmed (Ostojic et al., 2003). A sequence varia- (Minthon et al., 1997; Geschwind et al., 1998; Pickering-
tion in the intraflagellar transport 74 gene (IFT74) was Brown et al., 2001; Riemenschneider et al., 2002; Short
reported to segregate with disease in one family, but similar et al., 2002; Verpillat et al., 2002; Binetti et al., 2003) or
variations were not found in other linked families (Momeni non-Caucasian (Kowalska et al., 2001) populations. Indeed,
et al., 2006; Xiao et al., 2007). Nonetheless, the pathology a meta-analysis involving most of these studies (Verpillat
in such cases that have been reported is that of FTLD-TDP, et al., 2002) found no overall association between ApoE
type 2 (Cairns et al., 2007b), consistent with expectations e4 allele frequency and FTLD (but see Srinivasan et al.,
given the clinical phenotype. 2006). In patients with MAPT mutations, ApoE e4 allele fre-
UBAP1 encodes a protein of 502 residues, predicted to quency is not different from controls (Houlden et al., 1999b;
have a molecular weight of 55 kDa and originally cloned Pickering-Brown et al., 2002). Nonetheless, Srinivasan et al.
from a tumour suppressor locus (Qian et al., 2001). While (2006) reported that ApoE e4 allele frequency might be
little is known of the actual function of the protein, the gene selectively increased in males with FTLD, thereby doubling
is a likely member of the ubiquitin-activated enzymes fam- their chances of developing disease. It is also possible that
ily, which includes proteins with connections to ubiquitin clinical subgroups under FTLD umbrella might be differ-
and the ubiquitination pathway, suggesting a link between entially affected. Short et al. (2002) reported ApoE e4 allele
the ubiquitin proteasome system (UPS) and this condition. frequency to be increased in SD, compared to FTD and PA,
although Pickering-Brown et al. (2001) found this to be
67.6.2 CHROMOSOME 3 normal in FTD and SD, but tended to be increased in PA.
However, in both studies the number of patients with PA
Linkage to chromosome 3p11–12 has been reported in a and SD was relatively small and the present variability in
Danish family (Brown et al., 1995) showing FTD with fron- findings could be a reflection of this.
totemporal atrophy, neuronal loss and gliosis (Gydesen In contrast to AD, possession of ApoE e4 allele does not
et al., 2002). The gene responsible for disease in this pedigree appear to affect age at onset of disease (or duration of ill-
(Skibinski et al., 2005), and in three smaller families with ness) in sporadic FTLD (Pickering-Brown et al., 2001; Short
FTD1MND (Parkinson et al., 2006), has been identified as et al., 2002; Srinivasan et al., 2006) or in cases with MAPT
CHMP2B, which encodes the endosomal sorting complex mutations (Houlden et al., 1999b; Pickering-Brown et al.,
required for transport III (ESCRTIII complex subunit). 2002; Riemenschneider et al., 2002). Hence it seems unlikely
CHMP2B is part of the chromatin-modifying protein/ that ApoE e4 allele acts as a risk factor or disease modifier
charged multivesicular body protein family and is involved in FTLD generally, but could play a role in other clinical
in the degradation of surface receptor proteins and the for- subtypes, such as SD or PA.
mation of endocytotic multivesicular bodies, and forms Results from small series of FTLD patients suggest ApoE
a component of the UPS. The neuropathology is that of e2 allele could act as a risk factor (Lehmann et al., 2000;
FTLD-U. The ubiquitinated inclusions contain p62 pro- Verpillat et al., 2002), but again this remains to be substan-
tein, but not TDP-43 or FUS, occurring mostly as granular tiated (Kowalska et al., 2001; Pickering-Brown et al., 2001;
776 Dementia

Riemenschneider et al., 2002; Binetti et al., 2003). Meta- ascertained in 13 genealogically unrelated FTLD families
analysis, nevertheless, suggested increased ApoE e2 allele (Gass et al., 2006; Huey et al., 2006; Pickering-Brown et al.,
frequency. 2006; Spina et al., 2007). Patients have a wide onset age
range (44–69 years) and variable clinical diagnoses includ-
ing FTD, PNFA, CBD and AD (Rademakers et al., 2007).
Haplotype analysis has suggested two separate founding
67.7 FOUNDER EFFECTS events. A single common founder, most likely originating
in the United Kingdom approximately 300 years ago, impli-
67.7.1 MAPT cated 27 present day families. As with MAPT 116 mutation,
patients with R493X founder haplotype have been observed
P301L is the most common MAPT mutation found to date, in countries of British migration, including the United
being present in many families worldwide (Clark et al., States and Australia. Significantly, neither the PGRN R493X
1998; Dumanchin et al., 1998; Hutton et al., 1998; Mirra nor the MAPT 116 mutations have been identified in FTLD
et al., 1999; Rizzu et al., 1999; Houlden et al., 1999a; Binetti series from continental Europe (including the Netherlands,
et al., 2003; Rosso et al., 2003). Four of five families with Belgium, Poland, France, Italy and Spain) consistent with
P301L mutation shared the less common H2 haplotype on the view that the likely founders were from the United
the disease gene (Houlden et al. 1999a). These four families Kingdom, the appearance of patients in other countries fol-
were of Dutch origin (Rizzu et al., 1999) and might, there- lowing patterns of British migration. Founder effects have
fore, stem from a founder within this population (Rosso also been suggested for Gln415X (Pickering-Brown et al.,
et al., 2003). Two other families with P301L mutation on 2008) and Cys31LeufsX35 (Mackenzie et al., 2006b) muta-
the same H2 haplotype background (Sobrido et al., 2003) tions, both again relating to British stock.
indicate this same founder probably underlies other North
American/French Canadian families with French/Dutch
ancestry (Bird et al., 1999). However, P301L mutation has
been reported in other North American families with no 67.8 BIOMARKERS FOR FTLD
known Dutch/French or French Canadian ancestry (Mirra
et al., 1999) raising the possibility of a separate founder. Although clinicopathological correlations have sug-
Japanese families with P301L mutation do not share the gested that certain clinical FTLD subtypes are strongly
same extended H2 haplotype, again implying separate associated with one particular kind of histology, or even
founders. However, despite sharing H1 haplotype, several histological subtype, these associations are not perfect,
other MAPT polymorphisms, unique to Japanese individu- particularly for FTD that can be associated with all known
als, differed between two Japanese subjects with P301L histological forms of FTLD (including FUS pathology),
mutation suggesting separate founders. and for PNFA where tauopathy (Pick histology) can some-
Haplotype analysis (Pickering-Brown et al., 2004) has times be present. While the determination of genotype
shown a likely founder effect for British (Pickering- (i.e. identification of MAPT/PGRN/ CHMP2B/VCP muta-
Brown et al., 2002), North American (Yamaoka et al., 1996; tions) can unequivocally point towards the relevant histol-
Hulette et al., 1999) and Australian families (Dark, 1997; ogy, such cases are still relatively few and the full range of
Hutton et al., 1998) with 116 splice site mutation. Subsequent genetic involvement is unclear. Conversely, although par-
analysis (Colombo et al., 2009) dates this particular muta- ticular clinical phenotypes are frequently associated with
tion back to around 1300 ad within North Wales, a rural and particular mutational events (for example certain MAPT
relatively remote region, some 23 generations ago. It is likely mutations usually present with FTD, often with linguistic
that the founder lived in this part of the United Kingdom changes [Pickering-Brown et al., 2008], and PGRN muta-
with ‘spread’ of the mutation into more industrialized regions tions with FTD or PNFA phenotype [Snowden et al., 2006;
during the Industrial Revolution, further spreading to other Snowden et al., 2007; Pickering-Brown et al., 2008]), the
parts of the world, particularly the Commonwealth countries mutation present in familial autosomal dominantly inher-
through emigration in the eighteenth to twentieth centuries. ited FTLD cannot be precisely inferred without gene anal-
The identification of this large extended pedigree may be use- ysis. Hence, a straightforward laboratory test, based on
ful for identifying d
­ isease-modifying loci. easily accessible body fluids or tissues, that would identify
All other MAPT mutations appear to exist within sin- patients with FTLD from other neurodegenerative disor-
gle families, or single family members, and may represent ders, or perhaps more importantly discriminate cases with
mutational events that have remained ‘private’ to that fam- underlying FTLD-tau from those with FTLD-TDP without
ily or to have ‘spread’ less extensively to other countries. the requirement of complex genetic investigations, would
be a major adjunct to diagnostic precision. It could also act
67.7.2 PGRN as a predictive test, having great practical value in direct-
ing therapeutic strategies aimed at preventing or remov-
R493X is the most commonly reported PGRN mutation to ing tau or TDP-43 pathological changes from the brain
date, explaining the disease in 2.2% of FTLD cases, and in FTLD and other TDP-43 proteinopathies. The recent
The genetics and molecular pathology of frontotemporal lobar degeneration 777

identification of key molecular changes in the pathogen- Arai, T., Mackenzie, I.R.A., Hasegawa, M. et al. (2009).
esis of FTLD (and MND) should open the way to progress Phosphorylated TDP-43 in Alzheimer’s disease and
in these respects. dementia with Lewy bodies. Acta Neuropathologica,
117: 125–136.
Ayala, Y.M., Misteli, T. and Baralle, F.E. (2008). TDP-43
regulates retinoblastoma protein phosphorylation
67.9 CONCLUSIONS through the repression of cyclindependent kinase 6
expression. Proceedings of the National Academy
The past 3 years has seen a sea change in appreciation of of Sciences of the United States of America, 105:
the genetics and molecular pathology of FTLD. The twin 3785–3789.
discoveries of the presence of gene mutations in progranu- Baker, M., Mackenzie, I.R.A., Pickering-Brown, S.M. et al.
lin as a cause of FTLD, and the identification of the ubiq- (2006). Mutations in progranulin cause tau-negative
uitinated proteins, TDP-43 and FUS in the pathological frontotemporal dementia linked to chromosome 17.
lesions of non-tauopathy forms of FTLD, has opened new Nature, 442: 916–919.
vistas into the causes and effects of those tissue changes Bigio, E.H., Lipton, A.M., White, C.L. et al. (2003).
that are the pathological markers of disease, even if they Frontotemporal and motor neurone degeneration with
may not be the actual damaging agents. However, what neurofilament inclusion bodies: Additional evidence
we have seen so far, and what meagre clues have gained, is for overlap between FTD and ALS. Neuropathology
likely to be only the tip of the iceberg. There is still a whole and Applied Neurobiology, 29: 239–253.
world of basic neurobiology relating to PGRN, TDP-43 and Binetti, G., Nicosia, F., Benussi, L. et al. (2003). Prevalence of
FUS waiting to be discovered, and with that it will become TAU mutations in an Italian clinical series of familial fron-
increasingly clear as to the roles these molecules play not totemporal patients. Neuroscience Letters, 338: 85–87.
only in FTLD, or even in other neurodegenerative disor- Bird, T.D., Nochlin, D., Poorkaj, P. et al. (1999). A clinical
ders such as AD, but perhaps most importantly in terms pathological comparison of 3 families with frontotem-
of how they might maintain a watchful surveillance over poral dementia and identical mutations in the tau gene
the health and well-being of the nervous system ensuring (P301L). Brain: A Journal of Neurology, 122: 741–756.
proper brain function. These data will not be won easily. Bramham, C.R. and Wells, D.G. (2007). Dendritic mRNA:
It has been nearly two decades since the discovery of the Transport, translation and function. Nature Reviews in
amyloid precursor protein (APP), yet a definitive role for Neuroscience, 8: 776–789.
this protein in the brain is still elusive. Likewise, the prion Brandmeir, N.J., Geser, F., Kwong, L.K. et al. (2008).
protein has long been known, yet its precise brain func- Severe subcortical TDP-43 pathology in sporadic
tion remains unclear. PGRN, like APP and prion protein, frontotemporal lobar degeneration with motor neuron
may subserve a variety of roles concerned with regulating disease. Acta Neuropathologica, 115: 123–131.
the fundamental health, environment and infrastructure Brown, J., Ashworth, A., Gydesen, S. et al. (1995). Familial
of the brain. Although AD and prion disease are patho- non-specific dementia maps to chromosome 3. Human
logically characterized by accumulations of aggregated Molecular Genetics, 4: 1625–1628.
proteins, with presumed neurotoxic effects, it may be loss Bruni, A.C., Momeni, P., Bernardi, L. et al. (2007).
of the parent protein that is vital. The loss of function effect Heterogeneity within a large kindred with fronto-
of PGRN mutations in FTLD is instructive in this latter temporal dementia: A novel progranulin mutation.
context. Neurology, 69: 140–147.
Bugiani, O., Murrell, J.R., Giaccone, G. et al. (1999).
Frontotemporal dementia and corticobasal degenera-
REFERENCES tion in a family with a P301S mutation in tau. Journal
of Neuropathology and Experimental Neurology, 58:
Ahmed, Z., Mackenzie, I.R., Hutton, M.L. and Dickson, 667–677.
D.W. (2007). Progranulin in frontotemporal lobar Buratti, E. and Baralle, F.E. (2008). Multiple roles of TDP-
degeneration and neuroinflammation. Journal of 43 in gene expression, splicing regulation, and human
Neuroinflammation, 4: 7. disease. Frontiers in Bioscience, 13: 867–878.
Amador-Ortiz, C., Lin, W.-L., Ahmed, Z. et al. (2007). Cairns, N.J., Bigio, E.H., Mackenzie, I.R.A. et al. (2007a).
TDP-43 immunoreactivity in hippocampal sclerosis Neuropathologic diagnostic and nosologic criteria for
and Alzheimer’s disease. Annals of Neurology, 61: frontotemporal lobar degeneration: Consensus of the
435–445. consortium for frontotemporal lobar degeneration.
Arai, T., Hasegawa, M., Akiyama, H. et al. (2006). TDP- Acta Neuropathologica, 114: 5–22.
43 is a component of ubiquitin-positive tau-negative Cairns, N.J., Grossman, M., Arnold, S.E. et al. (2004a).
inclusions in frontotemporal lobar degeneration and Clinical and neuropathologic variation in neuronal
amyotrophic lateral sclerosis. Biochemical Biophysical intermediate filament inclusion disease. Neurology, 63:
Research Communications, 351: 602–611. 1376–1384.
778 Dementia

Cairns, N.J., Neumann, M., Bigio, E.H. et al. (2007b). De Silva, R., Lashley, T., Strand, K. et al. (2006). An
TDP-43 in familial and sporadic frontotemporal lobar immunohistochemical study of cases of sporadic and
degeneration with ubiquitin inclusions. The American inherited frontotemporal lobar degeneration using
Journal of Pathology, 171: 227–240. 3R- and 4R-specific tau monoclonal antibodies. Acta
Cairns, N.J., Perry, R.H., Jaros, E. et al. (2003). Patients Neuropathologica, 111: 329–340.
with a novel neurofilamentopathy: Dementia with Dumanchin, C., Camuzat, C., Campion, D. et al. (1998).
neurofilament inclusions. Neuroscience Letters, 341: Segregation of a missense mutation in the micro-
177–180. tubule-associated protein tau gene with familial
Cairns, N.J., Zhuckareva, V., Uryu, K. et al. (2004b). frontotemporal dementia with parkinsonism. Human
Alpha-internexin is present in the pathological inclu- Molecular Genetics, 7: 1825–1829.
sions of neuronal intermediate filament inclusion Fabre, S.F., Forsell, C., Viitanen, M. et al. (2001). Clinic-
disease. The American Journal of Pathology, 164: based cases with frontotemporal dementia show
2153–2161. increased cerebrospinal fluid tau and high apolipo-
Casafont, I., Bengoechea, R., Tapia, O. et al. (2009). TDP- protein E e4 frequency, but no tau gene mutations.
43 localises in mRNA transcription and processing Experimental Neurology, 168: 413–418.
sites in mammalian neurones. Journal of Structural Farrer, L.A., Abraham, C.R., Volicer, L. et al. (1995). Allele
Biology, 167: 235–241. e4 of apolipoprotein E shows a dose effect on age
Clark, L.N., Poorkaj, P., Wszolek, Z. et al. (1998). at onset of Pick disease. Experimental Neurology,
Pathogenic implications of mutations in the tau gene 136: 162–170.
in pallido-ponto-nigral degeneration and related neu- Farrer, M.J., Hulihan, M.M., Kachergus, J.M. et al. (2009).
rodegenerative disorders linked to chromosome 17. DCTN1 mutations in Perry syndrome. Nature Genetics,
Proceedings of the National Academy of Sciences of 41: 163–165.
the United States of America, 95: 13103–13107. Forman, M.S., Farmer, J., Johnson, J.K. et al. (2006a).
Colombo, R., Tavian, D., Baker, M.C. et al. (2009). Recent Frontotemporal dementia: Clinicopathological correla-
origin and spread of a common Welsh MAPT splice tions. Annals of Neurology, 59: 952–962.
mutation causing frontotemporal lobar degeneration. Foster, N.L., Wilhelmsen, K., Sima, A.A.F. et al. (1997).
Neurogenetics, 10: 313–318. Frontotemporal dementia and Parkinsonism linked to
Corder, E.H., Saunders, A.M., Strittmatter, W.J. et al. chromosome 17: A consensus conference. Annals of
(1993). Gene dose of apolipoprotein E type 4 allele Neurology, 41: 706–715.
and the risk of Alzheimer’s disease in late onset fami- Freeman, S.F., Spires-Jones, T., Hyman, B.T. et al. (2008).
lies. Science, 261: 921–923. TAR-DNA binding protein 43 in Pick disease. Journal
Cordes, M., Wszolek, W.K., Calne, D.B. et al. (1992). of Neuropathology and Experimental Neurology, 67:
Magnetic resonance imaging studies in rapidly 62–67.
progressive autosomal dominant Parkinsonism and Frisoni, G.B., Calabresi, L., Geroldi, C. et al. (1994).
dementia with pallido-ponto-nigral degeneration. Apolipoprotein E e4 allele in Alzheimer’s disease and
Neurodegeneration, 1: 217–224. vascular dementia. Dementia, 5: 240–242.
Cruts, M., Gijselinck, I., van der Zee, J. et al. (2006). Null Froelich Fabre, S., Axelman, P., Almkvist, A. et al. (2003).
mutations in progranulin cause ubiquitin-positive fron- Extended investigation of tau and mutation screen-
totemporal dementia linked to chromosome 17q21. ing of other candidate genes on chromosome 17q21
Nature, 442: 920–924. in a Swedish FTDP-17 family. American Journal of
Czech, C., Forstl, H., Monning, U. et al. (1994). ApoE Medical Genetics. Part B, Neuropsychiatric Genetics:
e4 in clinically diagnosed Alzheimer’s disease, fron- The Official Publication of the International Society of
tal lobe degeneration and non-demented controls. Psychiatric Genetics, 121B: 112–118.
Neurobiology of Ageing, 15 (Suppl. 1): S132. Fujita, K., Ito, H., Nakano, S. et al. (2008).
Dark, F. (1997). A family with autosomal dominant, non- Immunohistochemical identification of messenger
Alzheimer’s presenile dementia. Australian/New RNA-related proteins in basophilic inclusions of
Zealand Journal of Psychiatry, 31: 139–144. adult-onset atypical motor neuron disease. Acta
Davidson, Y., Amin, H., Kelley, T. et al. (2009). TDP-43 in Neuropathologica, 116: 439–445.
ubiquitinated inclusions in the inferior olives in fronto- Gass, J., Cannon, A., Mackenzie, I.R. et al. (2006).
temporal lobar degeneration and in other neurodegen- Mutations in progranulin are a major cause of
erative diseases: A degenerative process distinct from ­ubiquitin-positive frontotemporal lobar degeneration.
normal ageing. Acta Neuropathologica, 118: 359–369. Human Molecular Genetics, 55: 2988–3001.
Davidson, Y., Kelley, T., Mackenzie, I.R. et al. (2007). Geschwind, D., Karrim, J., Nelson, S.F. and Miller, B.
Ubiquitinated pathological lesions in frontotemporal (1998). The Apolipoprotein E epsilon4 allele is not a
lobar degeneration contain the TAR DNA-binding pro- significant risk factor for frontotemporal dementia.
tein, TDP-43. Acta Neuropathologica, 113: 521–533. Annals of Neurology, 44: 134–138.
The genetics and molecular pathology of frontotemporal lobar degeneration 779

Geser, F., Brandmeir, N.J., Kwong, L.K. et al. (2008a). Hulette, C.M., Pericak-Vance, M.A., Roses, A.D. et al.
Evidence of multisystem disorder in whole-brain map (1999). Neuropathological features of frontotemporal
of pathological TDP-43 in amyotrophic lateral sclero- dementia with parkinsonism linked to chromosome
sis. Archives of Neurology, 65: 636–641. 17q21-22(FTDP-17). Journal of Neuropathology and
Geser, F., Winton, M.J., Kwong, L.W. et al. (2008b). Experimental Neurology, 58: 859–866.
Pathological TDP-43 in parkinsonism–dementia Hutton, M., Lendon, C.L., Rizzu, P. et al. (1998).
complex and amyotrophic lateral sclerosis. Acta Association of missense and 50-splice-site mutations
Neuropathologica, 115: 133–146. in tau with the inherited dementia FTDP-17. Nature,
Gijselinck, I., van der Zee, J., Engelborghs, S. et al. (2008). 393: 702–705.
Progranulin locus deletion in frontotemporal demen- Josephs, K.A., Holton, J.L., Rossor, M.N. et al. (2003).
tia. Human Mutation, 29: 53–58. Neurofilament inclusion body disease: A new pro-
Gydesen, S., Brown, J.M., Brun, A. et al. (2002). teinopathy? Brain: A Journal of Neurology, 126:
Chromosome 3 linked frontotemporal dementia (FTD- 2291–2303.
3). Neurology, 59: 1585–1594. Josephs, K.A., Stroh, A., Dugger, B. and Dickson, D.W.
Hasegawa, M., Arai, T., Akiyama, H. et al. (2007). TDP-43 is (2009). Evaluation of subcortical pathology and
deposited in the Guam parkinsonism–dementia complex clinical correlations in FTLD-U subtypes. Acta
brains. Brain: A Journal of Neurology, 130: 1386–1394. Neuropathologica, 118: 349–358.
Hasegawa, M., Smith, M.J., Iijima, M. et al. (1999). FTDP-17 Knopman, D.S., Mastri, A.R., Frey, W.H. et al. (1990).
mutations N279K and S305N in tau produce increased Dementia lacking distinctive histologic features:
splicing of exon 10. FEBS Letters, 443: 93–96. A common non-Alzheimer degenerative dementia.
He, Z. and Bateman, A. (2003). Progranulin (granulin- Neurology, 40: 251–256.
epithelin precursor, PC-cell-derived growth factor, Kowalska, A., Asada, T., Arima, K. et al. (2001). Genetic
acrogranin) mediates tissue repair and tumorigenesis. analysis in patients with familial and sporadic fron-
Journal of Molecular Medicine, 81: 600–612. totemporal dementia: Two tau mutations in only
He, Z., Ong, C.H., Halper, J. and Bateman, A. (2003). familial cases and no association with apolipoprotein
Progranulin is a mediator of the wound response. e4. Dementia and Geriatric Cognitive Disorders, 12:
Nature Medicine, 9: 225–229. 387–392.
Holm, I.E., Englund, E., Mackenzie, I.R.A. et al. (2007). Le Ber, I., van der Zee, J., Hannequin, D. et al. (2007).
A reassessment of the neuropathology of frontotempo- Progranulin null mutations in both sporadic and famil-
ral dementia linked to chromosome 3 (FTD-3). Journal ial frontotemporal dementia. Human Mutation, 28:
of Neuropathology and Experimental Neurology, 66: 846–855.
884–891. Lee, E.B., Lee, V.M.Y., Trojanowski, J.Q. and Neumann, M.
Hong, M., Zhukareva, V., Vogelsberg-Ragaglia, V. et al. (2008). TDP-43 immunoreactivity in anoxic, ischaemic
(1998). Mutation-specific functional impairments in and neoplastic lesions of the central nervous system.
distinct tau isoforms of hereditary FTDP-17. Science, Acta Neuropathologica, 115: 305–310.
282: 1914–1917. Lehmann, D.J., Smith, A.D., Combrinck, M. et al. (2000).
Hosler, B., Siddique, T., Sapp, P.C. et al. (2000). Linkage of Apolipoprotein E e2 may be a risk factor for sporadic
familial amyotrophic lateral sclerosis with frontotempo- frontotemporal dementia. Journal of Neurology,
ral dementia to chromosome 9q21-22. Journal of the Neurosurgery and Psychiatry, 69: 404–405.
American Medical Association, 284: 1664–1669. Lipton, A.M., White, C.L. III., Bigio, E.H. (2004).
Houlden, H., Baker, M., Adamson, J. et al. (1999a). Frontotemporal lobar degeneration with motor neuron
Frequency of tau mutations in three series of non- disease-type inclusions predominates in 76 cases of
Alzheimer’s degenerative dementia. Annals of frontotemporal degeneration. Acta Neuropathologica,
Neurology, 46: 243–248. 108: 379–385.
Houlden, H., Rizzu, P., Stevens, M. et al. (1999b). Lynch, T., Sano, M., Marder, K.S. et al. (1994). Clinical
Apolipoprotein E genotype does not affect age at characteristics of a family with chromosome 17-linked
onset of dementia in families with defined tau muta- disinhibition–dementia–parkinsonism–amyotrophy
tions. Neuroscience Letters, 260: 193–195. complex. Neurology, 44: 187–194.
Hu, W.T., Josephs, K.A., Knopman, D.S. et al. (2008). Mackenzie, I.R.A., Baker, M., Pickering-Brown, S. et al.
Temporal lobar predominance of TDP-43 neuronal (2006a). The neuropathology of frontotemporal lobar
cytoplasmic inclusions in Alzheimer’s disease. Acta degeneration caused by mutations in the progranulin
Neuropathologica, 116: 215–220. gene. Brain: A Journal of Neurology, 129: 3081–3090.
Huey, E.D., Grafman, J., Wassermann, E.M. et al. (2006). Mackenzie, I.R.A., Neumann, M., Bigio, E. et al. (2009).
Characteristics of frontotemporal dementia patients Nomenclature for neuropathologic subtypes of fronto-
with a Progranulin mutation. Annals of Neurology, 60: temporal lobar degeneration: Consensus recommen-
374–380. dations. Acta Neuropathologica, 117: 15–18.
780 Dementia

Mackenzie, I.R.A., Shi, J., Shaw, C.L. et al. (2006b). Neumann, M., Sampathu, D.M., Kwong, L.K. et al. (2006).
Dementia lacking distinctive histology (DLDH) revis- Ubiquitinated TDP-43 in frontotemporal lobar degen-
ited. Acta Neuropathologica, 112: 551–559. eration and amyotrophic lateral sclerosis. Science, 314:
Masellis, M., Momeni, P., Meschino, W. et al. (2006). Novel 130–133.
splicing mutation in the progranulin gene causing Ostojic, J., Axelman, K., Lannfelt, L. and Froelich-Fabre,
familial corticobasal syndrome. Brain: A Journal of S. (2003). No evidence of linkage to chromosome
Neurology, 129: 3115–3123. ­9q21-22 in a Swedish family with frontotempo-
Minthon, L., Hesse, C., Sjogren, M. et al. (1997). The apo- ral dementia and amyotrophic lateral sclerosis.
lipoprotein E e4 allele frequency is normal in fronto- Neuroscience Letters, 340: 245–247.
temporal dementia, but correlates with age at onset of Parkinson, N., Ince, P.G., Smith, M.O. et al. (2006). ALS
disease. Neuroscience Letters, 226: 65–67. phenotypes with mutations in CHMP2B (charged mul-
Mirra, S.S., Murrell, J.R., Gearing, M. et al. (1999). Tau tivesicular body 2B). Neurology, 67: 1074–1077.
pathology in a family with dementia and a P301L Pick, A. (1892). Uber die Bejiehungen des senilen
mutation in tau. Journal of Neuropathology and Hirnatrophie zur Aphasie. Prager Medizinische
Experimental Neurology, 58: 335–345. Wochenschrift, 17: 165–167.
Momeni, P., Schymick, J., Jain, S. et al. (2006). Analysis of Pickering-Brown, S., Baker, M., Bird, T. et al. (2004).
IFT74 as a candidate gene for chromosome 9p-linked Evidence of a founder effect in families with fron-
ALS-FTD. BMC Neurology, 6: 44. totemporal dementia that harbor the tau +16 splice
Morita, M., Al-Chalabi, A., Andersen, P.M. et al. (2006). mutation. American Journal of Medical Genetics. Part
A locus on chromosome 9p confers susceptibility to B, Neuropsychiatric Genetics: The Official Publication
ALS and frontotemporal dementia. Neurology, 66: of the International Society of Psychiatric Genetics,
839–844. 125B: 79–82.
Mott, R.T., Dickson, D.W., Trojanowski, J.Q. et al. (2005). Pickering-Brown, S.M., Baker, M., Gass, J. et al. (2006).
Neuropathologic, biochemical, and molecular charac- Mutations in progranulin explain atypical phenotypes
terization of the frontotemporal dementias. Journal with variants in MAPT. Brain: A Journal of Neurology,
of Neuropathology and Experimental Neurology, 64: 129: 3124–3126.
420–428. Pickering-Brown, S.M., Owen, F., Snowden, J.S. et al.
Mukherjee, O., Pastor, P., Cairns, N.J. et al. (2006). HDDD2 (2001). Apolipoprotein E E4 allele has no effect on age
is a familial frontotemporal lobar degeneration with at onset or duration of disease in cases of frontotem-
ubiquitin-positive, tau-negative inclusions caused by a poral dementia with Pick- or microvacuolar-type histol-
missense mutation in the signal peptide of progranu- ogy. Experimental Neurology, 163: 452–456.
lin. Annals of Neurology, 60: 314–322. Pickering-Brown, S.M., Richardson, A.M.T., Snowden,
Munoz, D., Neumann, M., Kusaka, H. et al. (2009). FUS J.S. et al. (2002). Inherited frontotemporal dementia in
pathology in basophilic inclusion body disease (BIBD). nine British families associated with intronic mutations
Acta Neuropathologica, 118: 617–627. in the tau gene. Brain: A Journal of Neurology, 125:
Nakashima-Yasuda, H., Uryu, K., Robinson, J. et al. (2007). 732–751.
Co-morbidity of TDP-43 proteinopathy in Lewy body Pickering-Brown, S., Rollinson, S., Du Plessis, D. et al.
related diseases. Acta Neuropathologica, 114: 221–229. (2008). Frequency and clinical characteristics
Neary, D., Snowden, J.S., Gustafson, L. et al. (1998). of p
­ rogranulin (PGRN) mutation carriers in the
Frontotemporal lobar degeneration: A consensus on Manchester frontotemporal lobar degeneration
clinical diagnostic criteria. Neurology, 51: 1546–1554. cohort: Comparison to patients with tau (MAPT) and
Neary, D., Snowden, J.S. and Mann, D.M.A. (2005). no known mutation. Brain: A Journal of Neurology,
Frontotemporal dementia. The Lancet Neurology, 4: 131: 721–731.
771–779. Poorkaj, P., Bird, T., Wijsman, E. et al. (1998). Tau is a
Neary, D., Snowden, J.S. and Mann, D.M.A. (2007). candidate gene for chromosome 17 frontotemporal
Frontotemporal lobar degeneration: Clinical and patho- dementia. Annals of Neurology, 43: 815–825.
logical relationships. Acta Neuropathologica, 114: 31–38. Poorkaj, P., Grossman, M., Steinbart, E. et al. (2001).
Neumann, M., Rademakers, R., Roeber, S. et al. (2009a). Frequency of tau gene mutations in familial and spo-
A new subtype of frontotemporal lobar degeneration radic cases of non-Alzheimer dementia. Archives of
with FUS pathology. Brain: A Journal of Neurology, Neurology, 58: 383–387.
132: 2922–2931. Qian, J., Yang, J., Zhang, X. et al. (2001). Isolation and
Neumann, M., Roeber, S., Kretzschmar, H. and Mackenzie, characterization of a novel cDNA, UBAP1, derived
I.R.A. (2009b). Abundant FUS pathology in neuronal from the tumor suppressor locus in human chromo-
intermediate filament inclusion disease. Acta some 9p21-22. Journal of Cancer Research and Clinical
Neuropathologica, 118: 605–616. Oncology, 127: 613–618.
The genetics and molecular pathology of frontotemporal lobar degeneration 781

Rademakers, R., Baker, M., Gass, J. et al. (2007). An inter- Snowden, J.S., Neary, D., Mann, D.M.A. et al. (1992).
national initiative to study phenotypic variability asso- Progressive language disorder due to lobar atrophy.
ciated with progranulin haploinsufficiency in patients Annals of Neurology, 31: 174–183.
with the common 1477C→ T (Arg493X) mutation. The Snowden, J.S., Pickering-Brown, S.M., Du Plessis, D.
Lancet Neurology, 6: 857–868. et al. (2007). Progressive anomia revisited: Focal
Rademakers, R., Cruts, M., Dermaut, B. et al. (2002). Tau- degeneration associated with progranulin gene muta-
negative frontal lobe dementia at 17q21: Significant tion. Neurocase, 13: 366–377.
fine mapping of the candidate region to a 4.8-cm Snowden, J.S., Pickering-Brown, S.M., Mackenzie, I.R.
interval. Molecular Psychiatry, 7: 1064–1074. et al. (2006). Progranulin gene mutations associated
Riemenschneider, M., Diehl, J., Muller, U. et al. (2002). with frontotemporal dementia and progressive apha-
Apolipoprotein E polymorphism in German patients sia. Brain: A Journal of Neurology, 129: 3091–3102.
with frontotemporal degeneration. Journal of Sobrido, M.-J., Miller, B.L., Havlioglu, N. et al. (2003).
Neurology, Neurosurgery and Psychiatry, 72: 639–643. Novel tau polymorphisms, tau haplotypes and splic-
Rizzu, P., Van Swieten, J.C., Joosse, M. et al. (1999). High ing in familial and sporadic frontotemporal dementia.
prevalence of mutations in the microtubule-associated Archives of Neurology, 60: 698–702.
protein tau in a population study of frontotemporal Spillantini, M.G., Murrell, J.R., Goedert, M. et al. (1998).
dementia in the Netherlands. American Journal of Mutation in the tau gene in familial multiple system
Human Genetics, 64: 414–421. tauopathy with presenile dementia. Proceedings of the
Rosso, S.M., Donker Kaat, L., Baks, T. et al. (2003). National Academy of Sciences of the United States of
Frontotemporal dementia in the Netherlands: Patient America, 95: 7737–7741.
characteristics and prevalence estimates from a popu- Spina, S., Murrell, J.R., Huey, E.D. et al. (2007).
lation based study. Brain: A Journal of Neurology, 126: Clinicopathologic features of frontotemporal dementia
2016–2022. with Progranulin sequence variation. Neurology, 68:
Rosso, S.M., Kamphorst, W., de Graaf, B. et al. (2001). 820–827.
Familial frontotemporal dementia with ubiquitin posi- Srinivasan, R., Davidson, Y., Gibbons, L. et al. (2006). The
tive inclusions is linked to chromosome 17q21-22. apolipoprotein E e4 allele selectively increases the
Brain: A Journal of Neurology, 124: 1948–1957. risk of frontotemporal lobar degeneration in males.
Rovelet-Lecrux, A., Deramecourt, V., Legallic, S. et al. Journal of Neurology, Neurosurgery and Psychiatry, 77:
(2008). Deletion of the progranulin gene in patients 154–158.
with frontotemporal lobar degeneration or Parkinson Stevens, M., van Duijn, C.M., de Knijff, P. et al. (1997).
disease. Neurobiology of Disease, 31: 41–45. Apolipoprotein E gene and sporadic frontal lobe
Sampathu, D.M., Neumann, M., Kwong, L.K. et al. (2006). dementia. Neurology, 48: 1526–1529.
Pathological heterogeneity of frontotemporal lobar Stevens, M., van Duijn, C.M., Kamporst, W. et al. (1998).
degeneration with ubiquitin-positive inclusions Familial aggregation in frontotemporal dementia.
delineated by ubiquitin immunohistochemistry and Neurology, 50: 1541–1545.
novel monoclonal antibodies. The American Journal of Taniguchi, S., McDonagh, A.M., Pickering-Brown, S.M.
Pathology, 169: 1343–1352. et al. (2004). The neuropathology of frontotemporal
Schneider, J.A., Gearing, M., Robbins, R.S. et al. (1995). lobar degeneration with respect to the cytologi-
Apolipoprotein E genotype in diverse neurodegenera- cal and biochemical characteristics of tau protein.
tive disorders. Annals of Neurology, 38: 131–135. Neuropathology and Applied Neurobiology, 30: 1–18.
Schwab, C., Arai, T., Hasegawa, M. et al. (2009). TDP- Thorpe, J.R., Tang, H., Atherton, J. and Cairns, N.J.
43 pathology in familial British dementia. Acta (2008). Fine structural analysis of the neuronal inclu-
Neuropathologica, 118: 303–311. sions in frontotemporal lobar degeneration with TDP-
Shi, J., Shaw, C.L., Richardson, A.M.T. et al. (2005). 43 proteinopathy. Journal of Neural Transmission, 115:
Histopathological changes underlying frontotemporal 1661–1671.
lobar degeneration with clinicopathological correla- Turner, B.J., Baumer, D., Parkinson, N.J. et al. (2008).
tion. Acta Neuropathologica, 110: 501–512. TDP-43 expression in mouse models of amyotrophic
Short, R.A., Graff-Radford, N.R., Adamson, J. et al. (2002). lateral sclerosis and spinal muscular atrophy. BMC
Differences in tau and apolipoprotein E polymorphism Neuroscience, 9: 104–114.
frequencies in sporadic frontotemporal lobar degener- Uryu, K., Nakashima-Yasuda, H., Forman, M.S. et al.
ation syndromes. Archives of Neurology, 59: 611–615. (2008). Concomitant TAR-DNA-binding protein
Skibinski, G., Parkinson, N.J., Brown, J.M. et al. (2005). 43 pathology is present in Alzheimer disease and
Mutations in the endosomal ESCRTIII-complex sub- corticobasal degeneration but not in other tauopa-
unit CHMP2B in frontotemporal dementia. Nature thies. Journal of Neuropathology and Experimental
Genetics, 37: 806–808. Neurology, 67: 555–564.
782 Dementia

Vance, C., Al-Chalabi, A., Ruddy, D. et al. (2006). Wszolek, Z.K., Pfeiffer, R.F., Bhatt, M.H. et al. (1992).
Familial amyotrophic lateral sclerosis with fronto- Rapidly progressive autosomal dominant Parkinsonism
temporal dementia is linked to a locus on chromo- and dementia with pallido-ponto-nigral degeneration.
some 9p13.2-21.3. Brain: A Journal of Neurology, Annals of Neurology, 32: 312–320.
129: 868–876. Wszolek, Z.K., Tsuboi, Y., Uitti, R.J. et al. (2001).
Van Swieten, J.C. and Heutink, P. (2008). Mutations in Progressive supranuclear palsy as a disease phenotype
progranulin (GRN) within the spectrum of clinical and caused by S305S tau gene mutation. Brain: A Journal
pathological phenotypes of frontotemporal dementia. of Neurology, 124: 1666–1670.
The Lancet Neurology, 7: 965–974. Xiao, S., Sato, C., Kawarai, T. et al. (2007). Genetic studies
Verpillat, P., Camuzat, A., Hannequin, D. et al. (2002). of GRN and IFT74 in amyotrophic lateral sclerosis.
Association between the extended tau haplotype and Neurobiology of Aging, 29: 1279–1282.
frontotemporal dementia. Archives of Neurology, 59: Yamaoka, L.H., Welsh-Bohmer, K.A., Hulette, C.M. et al.
935–939. (1996). Linkage of frontotemporal dementia to chro-
Wang, H.Y., Wang, I.F., Bose, J. and Shen, C.K. (2004). mosome 17: Clinical and neuropathological charac-
Structural diversity and functional implications terization of phenotype. American Journal of Human
of the eukaryotic TDP gene family. Genomics, Genetics, 59: 1306–1312.
83: 130–139. Yancopoulou, D., Crowther, A., Chakrabarti, L. et al.
Wang, I.F., Wu, L.S., Chang, H.Y. and Shen, C.K. (2008). (2003). Tau protein in frontotemporal dementia linked
TDP-43, the signature protein of FTLD-U, is a neuronal to chromosome 3 (FTD-3). Journal of Neuropathology
activity-responsive factor. Journal of Neurochemistry, and Experimental Neurology, 62: 878–882.
105: 797–806. Zhang, Y.J., Xu, Y.F., Dickey, C.A. et al. (2007). Progranulin
Winton, M.J., Igaz, I.M., Wong, M.M. et al. (2008). mediates caspase-dependent cleavage of TAR DNA
Disturbance of nuclear and cytoplasmic TAR binding protein-43. Journal of Neuroscience, 27:
­DNA-binding protein (TDP-43) induces disease-like 10530–10534.
redistribution, sequestration, and aggregate for- Zhu, J., Nathan, C., Jin, W. et al. (2002). Conversion of
mation. The Journal of Biological Chemistry, 283: proepithelin to epithelins: Roles of SLPI and elastase in
13302–13309. host defense and wound repair. Cell, 111: 867–878.
68
Semantic dementia

JULIE S. SNOWDEN AND MATTHEW JONES

to be heterogeneous (Mesulam, 1982; Tyrrell et al., 1990a;


68.1 INTRODUCTION Snowden, et al., 1992). While some people present a non-
fluent, effortful and agrammatic form, others exhibit a flu-
Semantic dementia (SD) is a disorder of conceptual knowl- ent aphasia, characterized by impaired naming and word
edge resulting from degeneration of the anterior temporal comprehension. The label ‘semantic variant of PPA’ or
lobes. It is one of the canonical clinical syndromes, along ‘svPPA’ (Gorno-Tempini et al., 2011) has been applied to this
with the behavioural variant of frontotemporal demen- latter form. Close examination of such patients frequently
tia (bvFTD) disorder and expressive language disorder of reveals that the semantic disorder extends beyond the ver-
progressive non-fluent aphasia (PNFA), associated with a bal domain (Adlam et al., 2006) in keeping with SD. The
non-Alzheimer, frontotemporal lobar degeneration (FTLD) aphasia classification reflects the prominence of the lan-
pathology (Neary et al., 1998; Josephs et al., 2011). People guage symptoms. The terms SD and svPPA are sometimes
with SD lose the ability to name and understand words, to used interchangeably (e.g. Libon et al., 2013).
recognize faces and to understand the significance of objects. Cases of progressive prosopagnosia (Tyrrell et al., 1990b;
Although relatively rare, recognition of this disorder is Evans et al., 1995) are likely to represent right hemisphere
important because of its distinct implications for manage- presentations of SD (Josephs et al., 2008).
ment. Moreover, in recent years it has attracted considerable
academic interest because of its potential to shed light on the
organization and neural basis of semantic memory. From a
neurobiological perspective, a central interest is the relation- 68.3 DIAGNOSTIC CRITERIA
ship between SD and other clinical presentations of FTLD.
Clinical diagnostic criteria for SD developed in 1998 (Neary
et al., 1998) are shown in Table 68.1. Table 68.2 shows cri-
68.2 BACKGROUND teria defined under the label svPPA (Gorno-Tempini et al.,
2011). The SD criteria recognizes that face and object rec-
The term ‘semantic dementia’ was introduced in 1989 ognition difficulties may be prominent early symptoms,
(Snowden et al., 1989) to encapsulate the multimodal loss whereas the svPPA criteria, being part of more general cri-
of knowledge that typifies this syndrome. Prior to this, there teria for PPA, require that language difficulties are the most
were seminal reports in the literature of patients with cir- prominent deficit. The SD criteria incorporate behavioural
cumscribed impairments of semantic memory associated features as supportive diagnostic features, whereas the
with neurodegenerative disease, who would nowadays be svPPA criteria do not. The SD criteria distinguish between
classified as having SD (Warrington, 1975; Schwartz et al., core and supportive features, whereas the svPPA criteria
1979). Moreover, in the classical neurological and psychi- specify the number of features required to make a diagnosis
atric literature, there are patient descriptions (Pick, 1892; and distinguish different levels of diagnostic certainty (clin-
Rosenfeld, 1909) that appear prototypical of SD (Snowden, ical only, imaging-supported, pathologically confirmed).
2001). It is clear that SD has long existed. Perhaps, the most fundamental difference, however, relates
SD is sometimes described within the context of primary to the underlying intention of the criteria. The SD criteria
progressive aphasia (PPA) (see Chapter 69). Since the earli- are intended to predict an FTLD pathology, hence the inclu-
est descriptions (Mesulam, 1982), PPA has been recognized sion of features such as behavioural characteristics and the
783
784 Dementia

Table 68.1 Clinical diagnostic features of semantic dementia


I. Core diagnostic features
a. Insidious onset and gradual progression
b. Language disorder characterized by the following:
i. Progressive, fluent, empty spontaneous speech
ii. Loss of word meaning, manifested by impaired naming and comprehension
iii. Semantic paraphasias
c. Perceptual disorder characterized by the following:
i. Prosopagnosia: impaired recognition of identity of familiar faces
ii. Associative agnosia: impaired recognition of object identity
d. Preserved perceptual matching and drawing reproduction
e. Preserved single word repetition
f. Preserved ability to read aloud and write orthographically-regular words to dictation
II. Supportive diagnostic features
a. Speech and language
i. Press of speech
ii. Idiosyncratic word usage
iii. Absence of phonemic paraphasias
iv. Surface dyslexia and dysgraphia
v. Preserved calculation
b. Behaviour
i. Loss of sympathy and empathy
ii. Narrowed preoccupations
iii. Parsimony
c. Physical signs
i. Absent or late primitive reflexes
ii. Akinesia, rigidity and tremor
d. Neuropsychological testing
i. Profound semantic loss manifests in failure of word comprehension and naming and/or face and object
recognition
ii. Preserved phonology and syntax, elementary perceptual processing, spatial skills and day-to-day memorizing
e. Electrophysiology
i. Normal
f. Brain imaging (structural and/or functional)
i. Predominant anterior temporal abnormality (symmetric or asymmetric)
Source: Neary, D. et al., Neurology, 51, 1546–1554, 1998. Reproduced with permission from Wolters Kluwer Health Publishers.

preservation of calculation skills known to be helpful in socio-economic factors which affect incidence. Accurate
distinguishing FTLD from AD. The svPPA criteria by con- incidence and prevalence data are scarce. In an analy-
trast are designed to identify a specific language phenotype sis of patient referrals to a specialist dementia clinic in
associated with neurodegenerative disease, but make no Manchester, patients with prototypical, ‘pure’ SD accounted
assumptions or predictions regarding the precise nature of for approximately 10% of cases of FTLD, which in turn
that neurodegenerative pathology. accounted for about 25% of cases of primary degenerative
dementia presenting itself before the age of 65.

68.4 DEMOGRAPHIC FEATURES
68.5 PRESENTING COMPLAINTS
SD affects men and women equally. The mean age at onset is
around 60 years (Hodges et al., 2010), ranging from 40 to 80 The presenting complaint is typically of a difficulty ‘with
years. The median survival has been estimated at 12.8 years words’ or ‘remembering things’. Careful history-taking
(Hodges et al., 2010), although there is wide variation with reveals that the problem in ‘remembering’ is not one of
durations as short as 3 years and as long as 16 years. Most memory loss in the traditional sense. Patients are able to
cases are sporadic, a positive family history being less com- remember day-to-day events, keep track of time, remem-
mon than in other forms of FTLD (Goldman et al., 2005; ber appointments, find their way around without being lost
Hodges et al., 2010). There are no known geographical or and retain a degree of functional independence that would
Semantic dementia 785

Table 68.2 Clinical diagnosis of semantic variant PPA


I.
a. Both of the following core features must be present in the patient:
i. Impaired confrontation naming
ii. Impaired single-word comprehension
b. At least three of the following other diagnostic features must also be present:
i. Impaired object knowledge, particularly for low frequency or low-familiarity items
ii. Surface dyslexia or dysgraphia
iii. Spared repetition
iv. Spared speech production (grammar and motor speech)
II. Imaging-supported semantic variant PPA diagnosis
a. Both of the following criteria must be present in the patient:
i. Clinical diagnosis of semantic variant PPA
ii. Imaging must show one or more of the following results:
A. Predominant anterior temporal lobe atrophy
B. Predominant anterior temporal hypoperfusion or hypometabolism on SPECT or PET
III. Semantic variant PPA with definite pathology
a. Clinical diagnosis (criterion 1 below) and either criterion 2 or 3 must be present as well:
i. Clinical diagnosis of semantic variant PPA
ii. Histopathologic evidence of a specific neurodegenerative pathology (e.g. FTLD-tau, FTLD-TDP, AD, others)
iii. Presence of a known pathogenic mutation
Source: Gorno-Tempini, M.L. et al., Neurology, 76, 1006–1014, 2011. Reproduced with permission from Wolters Kluwer Health publishers.
Notes: To meet svPPA criteria, patients must first meet overarching criteria for PPA. These specify that language difficulties should be the
most prominent problem. Prominent behavioural disturbance is a basis for exclusion.
Abbreviations: PPA, primary progressive aphasia; SPECT, single photon emission computed tomography; PET, positron emission topogra-
phy; FTLD-tau, frontotemporal lobar degeneration-tau; FTLD-TDP, FTLD variant with TAR-DNA-binding protein 43 (TDP-43); AD,
Alzheimer’s disease.

be incompatible with a classical amnesia. The problem lies precisely the same time each day. They may have a prefer-
in remembering what words mean, who people are and what ence for order and align objects, straighten out folds in the
objects are for. The presenting symptoms are most often in curtains and plump up the cushions as soon as a person
the verbal domain, manifested by patients’ difficulty in the rises from an armchair.
understanding and use of words, but occasionally they are People with SD may show hypersensitivity to sensory
in the visual realm, manifested by a difficulty in recognizing stimuli (Snowden et al., 2001; Mahoney et al., 2011) and
faces. Relatives may also report a failure of object recogni- overact to light touch or to ostensibly innocuous environ-
tion: for example, an inability to recognize fruits and veg- mental sounds. They may be oblivious to danger, probably
etables in the supermarket. Medical referral is sometimes linked to their loss of conceptual understanding of the
precipitated by an accompanying behavioural alteration. world. They commonly show narrowed food preferences
and favour sweet flavour in foods.
Although many of these ‘temporal’ behaviours overlap
with those found in the predominantly ‘frontal’ disorder
bvFTD, there are qualitative differences (Snowden et al.,
68.6 BEHAVIOURAL CHANGES
2001). These are summarized in Table 68.3.

Patients become self-centred, lacking in empathy and


inflexible. Their behavioural repertoire becomes narrowed
and they frequently become preoccupied with one or two 68.6.1 INSIGHT
activities (e.g. painting, solving jigsaw puzzles or sudoku
puzzles), which they pursue relentlessly and at which they Patients are aware of difficulties, yet frequently provide
are adept (Green and Patterson, 2009). They may become trivial explanations for them, such as being ‘out of prac-
mentally preoccupied by particular themes (e.g. the with- tice with talking’ by virtue of being alone all day. Signs
drawal of their driving licence), which they repeat inces- of frustration are rare. A likely reason is the absence of
santly to the exasperation of family members. Behaviour an internal model of prior conceptual knowledge: patients
acquires a stereotypical quality. Patients may adopt a fixed have no comparator by which to judge the magnitude of
routine, clock-watch and carry out specific activities at what they have lost.
786 Dementia

Table 68.3 Behavioural characteristics of semantic dementia and bvFTD

Characteristics Semantic dementia bvFTD


Affect Warm Blunted
Social behaviour Exaggerated sociability Reduced sociability
Interest and motivation Narrowed interests General loss of interest
Task performance Productive and goal-directed Unproductive, no goal-direction
Attention Focused, good persistence Inattentive, poor persistence
Wandering and pacing Uncommon Common
Repetitive behaviours Complex routines Simple motor stereotypes
Clockwatches Unconcerned by time
Compulsive quality No compulsive quality
Eating Food fads, more selective Gluttony, more indiscrimination
Response to sensory stimuli Exaggerated Diminished
Abbreviation: bvFTD, behavioural variant of frontotemporal dementia.

a tendency for vocabulary relating to patients’ daily life to


68.7 NEUROLOGICAL SIGNS be better preserved than non-personally relevant vocabu-
lary (Snowden et al., 1994, 1995). Word comprehension and
Patients are generally free from neurological signs until naming assessment should, therefore, probe vocabulary that
late in the disease. Akinesia and rigidity and frontal release does not relate directly to the patient’s daily routine. Loss
phenomena may emerge in advanced disease. Very rarely of knowledge about a word’s meaning may not be absolute.
SD may be accompanied by motor neurone disease (Kim The patient may, for example, know that a lemon is a food
et al., 2009; Ostberg and Bogdanovic, 2011). Myoclonus is rather than an animal, but not know how it differs from a
absent. banana. Assessment needs to probe knowledge of attributes
as well as broad category knowledge. Potential methods of
assessing knowledge are shown in Figure 68.1.
Language comprehension problems arise principally at
68.8 COGNITIVE CHARACTERISTICS the single word level. Comprehension of syntax is relatively
well preserved. Reading and writing is similarly preserved
68.8.1 LANGUAGE for words with regular spellings. However, patients read
phonetically rather than meaningfully, so produce ‘regular-
Patients speak fluently and effortlessly, with normal artic- ization’ errors (e.g. ‘glove’ pronounced to rhyme with ‘rove’
ulation and grammar and without phonological (sound- and ‘stove’; ‘pint’ to rhyme with ‘mint’), consistent with sur-
based) errors, giving the superficial impression of normal face dyslexia (Patterson et al., 1985). Parallel errors occur in
language. Indeed, patients are often garrulous (Table 68.4). writing (e.g. ‘caught’ written as ‘cort’).
Nevertheless, there is typically a reduction in content words. With progression of the disease, speech content is
Specific terms may be used over inclusively: e.g. ‘container’ increasingly empty and stereotypical. Conversational rep-
to refer to objects that have no capacity to ‘contain’; ‘twist- ertoire becomes progressively reduced until only a few ste-
ing’ used to refer to actions, where no twisting motion is reotypical words or phrases remain.
involved.
The semantic impairment, which may be scarcely appar-
ent at clinical interview, becomes strikingly evident on for- 68.8.2 FACE AND OBJECT RECOGNITION
mal testing of naming and word comprehension. Patients
often score close to floor level on standard picture naming Problems in recognizing familiar faces occur early in the
tasks and derive no benefit from phonemic cues. Semantic course of disease. Difficulties are most profound for imper-
errors (e.g. ‘dog’ for tiger) may occur, reflecting loss of con- sonal faces, such as those of celebrities and least marked for
ceptual discrimination between related concepts. Word family and friends with whom the patient maintains daily
comprehension is also affected. Expressions of lack of under- contact. Perceptual discrimination of faces (i.e. whether two
standing of words patients were previously familiar with (e.g. faces are the same or different) is preserved.
‘Tiger, tiger, what’s a tiger? I don’t know what that is’) are a Object recognition difficulties invariably emerge with
revealing demonstration of the loss of word meaning. There disease progression. Like words, loss of understanding is
are known influences on naming performance. Common not all or none. Patients may recognize a lemon as edible
words are more likely to be named than low frequency terms but not know whether it tastes sweet or sour, or if it needs
(Lambon Ralph et al., 1998) and items typical of a category to be cooked. Moreover, they may recognize and use appro-
better than atypical items (Woollams et al., 2008). There is priately their own belongings while failing to recognize
Semantic dementia 787

Table 68.4 Characteristics of language disorder in semantic dementia


Spontaneous speech Fluent, effortless, often garrulous
Empty content, reduced nominal terms
Semantic paraphasias
Substitution of generic terms
Verbal stereotypes
Preserved use of syntax
Preserved phonology
Normal articulation and prosody
Comprehension Impaired word understanding
Good understanding of syntax
Repetition Relatively preserved
Naming Profound anomia
Semantic errors present (e.g. ‘dog’ for elephant)
No benefit from phonemic cues
Reading Fluent reading aloud
Accurate reading of regular words
Regularization of irregular words (surface dyslexia)
Impaired comprehension of written material
Writing Fluent, effortless execution
Regularization of irregular words
(e.g. ‘cort’ for caught) (surface dysgraphia)

Word-picture match

Which is the lion?

Knowledge of habitat Hot country or cold country?

Where would you find this? In the farmland or in the wild?

In Africa or in America?

Knowledge of attributes Friendly/tame or


Lion
unfriendly/dangerous?
How does this behave?

What noise does it make? Roars or barks?

Figure 68.1 Methods of assessing semantic knowledge.

other examples of those same objects or pictures of those discrimination and matching tasks that are not dependent
objects (Snowden et al., 1994; Bozeat et al., 2002) belong- upon recognition of object meaning. They may draw nor-
ing to others. This latter feature helps to explain the clinical mally, sometimes to a strikingly proficient level (Figure 68.2).
observation that patients function reasonably well within
the narrow confines of their familiar environment and rep- 68.8.3 RECOGNITION FROM TOUCH,
ertoire of daily activities at a time when abstract semantic TASTE, SMELL AND SOUND
knowledge is profoundly impaired. Eventually, even highly
familiar objects may no longer be recognized. Primary sensory abilities are well preserved, so patients
Non-semantic aspects of object perception are well pre- have no difficulty detecting the presence of tactile, gusta-
served. Patients have no difficulty carrying out perceptual tory, olfactory and auditory stimuli and discriminating
788 Dementia

Figure 68.2 Painting by semantic dementia patient.

whether two stimuli are the same or different. However, 68.8.6 MEMORY


the disorder of meaning extends to all sensory modali-
ties (Snowden et al., 1996; Bozeat et al., 2000; Hodges and A feature that is striking on clinical grounds and that helps to
Patterson, 2007; Luzzi et al., 2007; Piwnica-Worms et al., distinguish SD from other forms of dementia is the apparent
2010). Patients may have difficulty recognizing the identity preservation of patients’ current autobiographical memory
of textures, tastes, smells and non-verbal sounds such as the (Warrington, 1975; Snowden et al., 1996). Patients remember
ringing of a telephone or the sound of thunder despite per- appointments and personally relevant events and keep track of
ceiving those stimuli normally. time. They negotiate the environment without being lost. If they
moved to a new home, they have no difficulty learning their
68.8.4 NUMERACY new surroundings. In contrast, they show gross breakdown in
impersonal, factual (semantic) knowledge, including knowl-
Understanding of number concepts and the ability to edge about public figures and important world events. The
carry out arithmetical procedures often appear remarkably preservation of autobiographical memory is not easily ­captured
well preserved (Cappelletti et al., 2001, 2002; Crutch and by standard memory tests, which typically involves lists of
Warrington, 2002), perhaps explaining why SD patients com- words, faces or line drawings that may have little ­meaning for
monly enjoy number puzzles such as sudoku and television the patient. Moreover, routine bedside questions, designed to
quiz shows involving numbers. Nevertheless, number con- probe orientation, such as ‘What town are we in?’ demand nam-
cepts are not entirely normal (Julien et al., 2008). Eventually, ing skills and thus may be compromised in SD. Time orienta-
arithmetical skills become compromised and patients cease tion scores are typically superior to those of place o
­ rientation,
to understand the meaning of spoken and written numerals. reflecting better preservation of number-related vocabulary.
Testing of memory needs to include test m ­ aterials that are
68.8.5 SPATIAL SKILLS meaningful to the patients and questions regarding the patient’s
daily life. Episodic memory, assessed in a ­naturalistic setting,
Spatial and navigational skills are generally excellent. has been shown to be well preserved (Adlam et al., 2009).
Patients are able to find their way without becoming lost and
may use spatial cues to compensate for object recognition
difficulties (for example, by recalling the spatial location of 68.8.7 EXECUTIVE SKILLS
food items on a supermarket shelf). On neuropsychologi-
Executive skills, such as rule abstraction, mental set-
cal testing, patients typically perform well on traditional
shifting, response inhibition and problem solving, that are
spatial tests such as line orientation (Benton et al., 1978),
heavily dependent on frontal lobe function, can be remark-
dot counting, position discrimination and cube estima-
ably well preserved in SD. Nevertheless, semantic break-
tion (Warrington and James, 1991), although in late stage
down inevitably impacts task performance when stimuli
disease performance may be compromised secondarily by
have no meaning for the patient.
their semantic disorder (e.g. failure to recall the names of
numbers). Even in advanced stage of the disease, patients
negotiate their environment skilfully, may recall the loca-
tion of their chair or bed on a hospital ward, localize and
manipulate objects in the environment with ease and spa- 68.9 BRAIN IMAGING
tially align objects perfectly. By contrast to the essential
preservation of spatial skills, patients may exhibit a ‘topo- Magnetic resonance imaging (MRI) shows cerebral atrophy,
graphical agnosia’ reflecting a failure to recognize buildings particularly marked in the anterior temporal lobes, and affect-
and other locations. ing inferior more than superior temporal gyri (Hodges et al.,
Semantic dementia 789

1992; Mummery et al., 2000; Chan et al., 2001; Rohrer et al., speculated that such cases have a less pure semantic disor-
2008, 2009). Although bilateral, the temporal lobe atrophy is der. Tau pathology has been linked (Pickering-Brown et al.,
often asymmetric (see Figure 68.3), the left or right predomi- 2002) to a mixed clinical picture in which bvFTD is com-
nance being associated with clinical presentations emphasiz- bined with semantic breakdown.
ing word meaning or face recognition disorder, respectively. In some series (Hodges et al., 2010), a minority of
With the progression of the disease, atrophy spreads within patients have been reported to show Alzheimer pathology.
both hemispheres (Rohrer et al., 2008; Brambati et al., 2009), Such findings exemplify the fact that semantic impairment
although asymmetries may manifest even at end stage. may be a prominent feature in some Alzheimer patients and
Parallel hypometabolic changes in the temporal lobes are so can be mistaken for SD. Such patients typically exhibit a
found on positron emission tomography (PET) imaging classical amnesia and early calculation problems, in addi-
(Diehl et al., 2004; Desgranges et al., 2007). tion to their semantic impairment.
Studies of white matter connectivity have shown extensive
loss of connectivity of ventral pathways, including uncinate
and inferior longitudinal fasciculus, with relative sparing of
frontoparietal pathways (Agosta et al., 2010; Acosta-Cabronero 68.11 MOLECULAR GENETICS
et al., 2011), suggesting breakdown of a wider semantic net-
work extending beyond the anterior temporal lobes. Genetic mutations in the MAPT, GRN and C9orf72 genes
that are associated with bvFTD and PNFA (see Chapter
67) are typically absent in pure SD, in keeping with the
low familial incidence. A mixed syndrome of semantic loss
68.10 NEUROPATHOLOGY combined with bvFTD is a feature of +16 tau mutations
(Pickering-Brown et al., 2002). Semantic loss may occa-
The striking feature on macroscopic examination is atrophy sionally be a prominent symptom of C9orf72 mutations
of the temporal lobes. Atrophy is bilateral, but often asym- (Galimberti et al., 2013; Abbate et al., 2014).
metrical and affects particularly anterior and inferior parts
of the temporal lobes. Frontal cortex is affected to a lesser
degree. There is relative sparing of superior temporal gyrus,
parietal and occipital cortex. 68.12 RELATIONSHIP TO BVFTD AND
In FTLD, the histopathological changes are not uniform PNFA
(see Chapter 67). A primary distinction is made between
tau- and TAR DNA-binding protein 43 (TDP-43) patholo- The established association between SD, bvFTD and
gies (Mackenzie et al., 2010), the latter being distinguished PNFA, comes from several sources: (1) the existence of
into three subtypes based on the relative preponderance of mixed/overlapping clinical syndromes, (2) the poten-
neuronal cytoplasmic and intranuclear inclusions and dys- tial association of each syndrome with motor neurone
trophic neurites (Mackenzie et al., 2011). Pure SD cases are disease (Neary et al., 1990; Caselli et al., 1993; Kim et al.,
strongly associated with TDP-43, type C pathology (Josephs 2009), and (3) shared pathologies. The latter point requires
et al., 2009, 2011; Snowden et al., 2011) characterized by a elaboration. Until recently, the assumption was that syn-
preponderance of dystrophic neurites within cerebral cor- dromic differences reflected differences in the anatomi-
tex. Occasionally, an association with tau pathology has cal distribution within the anterior hemispheres of a
been reported (Hodges et al., 2010), although it might be common underlying pathology. Nevertheless, refinement of

(a) (b)

Figure 68.3 Magnetic resonance imaging (MRI) scans of two patients with semantic dementia showing severe temporal
lobe atrophy. In one case (a) atrophy is more marked on the left side and in the other case (b) on the right.
790 Dementia

histopathological characterization points to distinct patho- ●● In SD speech production is fluent and effortless. In AD,
logical as well as clinical phenotypes and a closer mapping it is typically hesitant, suggesting a difficulty in retriev-
between clinical phenotype and underlying histopathology ing words and sentences may be unfinished reflecting
than previously thought (Josephs et al., 2009; Snowden et al., loss of train of thought. There may be phonologi-
2011). SD appears uniquely associated with TDP-43 type C cal errors and in later stages a stuttering quality that
pathology, whereas PNFA is strongly linked to TDP-43 type accompanies speech is usually evident.
A or tau pathology and FTD with motor neurone disease to ●● In SD, comprehension is most impaired for words and
TDP-43 type B. Such findings suggest fundamental differ- better preserved for syntax, whereas in AD, the reverse
ences in the neurobiology of SD and other forms of FTLD. is typically the case.
●● Numerical skills are well preserved in SD, whereas cal-
culation problems are affected early in AD disease.
●● Spatial abilities are well preserved in SD, whereas AD
68.13 DIFFERENTIAL DIAGNOSIS patients commonly show difficulty orientating clothing
when dressing and topographical disorientation within
SD is poorly recognized for understandable reasons (see a familiar environment.
Table 68.5). It is most likely to be confused with Alzheimer’s ●● Patients with both SD and AD may have object recog-
disease (AD), reflecting the fact that presenting symp- nition problems. In SD, the difficulty lies in assigning
toms in both are commonly of ‘memory’ problems and meaning to objects that are perceived normally (associa-
patients’ fluent, effortless conversational speech might, at a tive visual agnosia). They can copy drawings of objects
clinical interview, mask the underlying semantic disorder. that they cannot recognize. In AD, perceptual break-
Nevertheless, the ‘typical’ clinical profiles of SD and AD are down lies at the level of achieving an integrated percept
very different (see Table 68.6) as outlined below: (apperceptive agnosia). Patients may interpret elements
of a figure instead of the overall outline (e.g. a pair of
●● In SD, ‘memory loss’ refers to loss of impersonal, con- scissors identified as a bowl on the basis of the circular
ceptual knowledge about the world, whereas personal, handle). They have difficulty copying line drawings,
autobiographical memories remain relatively well pre- their copies often being fragmented with loss of the
served. In AD the converse is true. spatial relationships between elements of the figure.

Table 68.5 Factors affecting poor recognition of semantic dementia


• Rare disorder
• Historical reports of ‘memory’ disorder
• Semantic disorder may not be evident during clinical interview
• Semantic impairments may be misinterpreted as classical amnesia
• Need explicit tests of word comprehension and naming, face and object recognition

Table 68.6 Clinical differences between semantic dementia and Alzheimer’s disease

Features Semantic dementia Alzheimer’s disease


Age of onset Most common in middle age Most common among elderly
Language Selective semantic disorder No selective semantic disorder
Impaired single word comprehension Impaired sentence comprehension
Perception Associative agnosia Apperceptive agnosia
Calculation Preserved early in course Impaired early in course
Spatial orientation Preserved Impaired early in course
Constructional skills Preserved Impaired early in course
Memory Autobiographical memory preserved Autobiographical memory impaired
Neurological signs Myoclonus absent Myoclonus may be present
Electroencephalography Normal Slowing of wave forms
Structural brain imaging Focal anterior-inferior temporal lobe atrophy Focal anterior temporal atrophy rare
typical
Hippocampi relatively preserved Hippocampi atrophic
Marked hemispheric asymmetry common Marked asymmetry rare
Functional brain imaging Anterior hemisphere abnormalities Posterior hemisphere abnormalities
Marked hemispheric asymmetry common Marked asymmetry rare
Semantic dementia 791

Notwithstanding these distinct profiles, differentia-


tion is not invariably clear-cut. Semantic impairment may 68.15 THEORETICAL ISSUES
be present in AD (Hodges and Patterson, 1995; Rogers
and Friedman, 2008; Libon et al., 2013) and in some AD SD attracts considerable theoretical interest from neuropsy-
patients it is a prominent feature (Alladi et al., 2007). chologists. Its circumscribed nature means that the disor-
Diagnostic differentiation in such cases is likely to lie der represents a natural model for understanding semantic
in the degree of accompanying amnesia and pattern of memory. It is now widely accepted that concepts depend
behavioural change. on a distributed brain network, modality-specific concep-
tual features being represented by distinct sensory and
motor brain regions (Pulvermüller et al., 2009; Kiefer and
Pulvermüller, 2012). Nevertheless, the profound and per-
68.14 TREATMENT AND vasive nature of conceptual loss in SD provides compelling
MANAGEMENT grounds for the view that the anterior temporal lobes have
a special role to play. The precise nature of that privileged
Pharmacological therapies for AD do not have a rational role remains contentious. For some, the anterior temporal
role in SD, because there is no evidence of abnormalities in lobes constitute a supramodal or amodal hub where infor-
the cholinergic system (Francis et al., 1993). In patients with mation is distilled and represented in a non-modal form
problematic behavioural changes, symptomatic treatment (Patterson et al., 2007; Hoffman et al., 2012). A challenge
may be warranted. There is anecdotal evidence that repeti- to this ‘amodal’ position is the fact that understanding of
tive, stereotypical behaviours may respond to selective sero- names (verbal information) and faces (visual information)
tonin reuptake inhibitors (SSRIs). may be affected differentially in SD, depending on whether
With regard to practical management, it is of relevance the left or right temporal lobe is most affected (Snowden
that concepts (of words, object meaning etc.), relevant to et al., 2004, 2012). Such findings, it is argued (Snowden
the patient’s daily life appear to be better retained than et al., 2012), would favour the notion of the anterior tem-
those which have no personal relevance (Snowden et al., poral lobes as modality-specific convergence zones rather
1994, 1995; Westmacott et al., 2003). It has been argued than a unitary amodal hub. Other authors (Mion et al.,
that this feature results from patients’ preserved auto- 2010) have accommodated hemispheric differences within
biographical memory: concepts, which would otherwise the ‘amodal’ framework by attributing those differences
be lost in their abstract sense, have some meaning to to differential strength of connectivity of the amodal hub
the patient if they are framed within the context of the to modality specific brain regions within the two hemi-
patient’s ongoing daily experience. A word, in isolation, spheres. Such debate exemplifies the importance of SD in
may have no meaning to the patient, yet meaning may be informing, understanding and constraining theories of the
conveyed by the context of a complete sentence. An object neural basis of conceptual knowledge.
might not be recognized out of context, yet recognized
within the context of its usual location. By implication,
input from speech therapists is more likely to be beneficial
68.16 CONCLUSIONS
if this takes place in the patient’s own surroundings using
as referents patients’ own belongings, than in the abstract
setting of a clinical consultation room using standard SD is a striking disorder, clinically distinct from AD and
pictorial materials. Moreover, the findings highlight the other forms of dementia. Patients exhibit unique pat-
importance of maintaining a familiar routine in patient terns of cognitive and behavioural symptomatology, which
management. demand novel approaches to treatment and management.
There is clinical evidence in SD that some learning is pos- Neuropsychological studies are likely to improve under-
sible. Patients may, for example, effectively learn the names standing of the cognitive characteristics of this disorder.
of new acquaintances or of prescribed medicines. They may Meanwhile, basic scientific advances ought to shed light on
also succeed in relearning lost vocabulary (Snowden and the underlying aetiology of this and other clinical manifesta-
Neary, 2002; Henry et al., 2008; Jokel and Anderson, 2012; tions of FTLD.
Savage et al., 2013; Hoffman et al., 2015). However, reten-
tion depends on continued rehearsal and is more robust REFERENCES
when words are linked to patients’ ongoing daily experi-
ences (Snowden and Neary, 2002). An alternative therapeu- Abbate, C., Arosio, B., Galimberti, D. et al. (2014).
tic approach involves the use of compensatory strategies Phenotypic variability associated with the C9orf72
of enactment to bypass verbal communication difficulties hexanucleotide repeat expansion: A sporadic case of
(Kindell et al., 2013). These findings demonstrate the poten- frontotemporal lobar degeneration with prodromal
tial value of speech and language therapy intervention in the hyposmia and predominant semantic deficits. Journal
management of SD. of Alzheimer’s Disease, 40 (4): 849–855.
792 Dementia

Acosta-Cabronero, J., Patterson, K., Fryer, T.D. et al. (2011). Evans, J.J., Heggs, A.J., Antoun, N. and Hodges, J.R.
Atrophy, hypometabolism and white matter abnormali- (1995). Progressive prosopagnosia associated with
ties in semantic dementia tell a coherent story. Brain: selective right temporal lobe atrophy: A new syn-
A Journal of Neurology, 134 (Pt 7): 2025–2035. drome? Brain: A Journal of Neurology, 118: 1–13.
Adlam, A.L., Patterson, K. and Hodges, J.R. (2009). Francis, P.T., Holmes, C., Webster, M.-T. et al. (1993).
‘I remember it as if it were yesterday’: Memory for Preliminary neurochemical findings in non-Alzheimer
recent events in patients with semantic dementia. dementia due to lobar atrophy. Dementia, 4: 172–177.
Neuropsychologia, 47 (5): 1344–1351. Galimberti, D., Fenoglio, C., Serpente, M. et al. (2013).
Adlam, A.L., Patterson, K., Rogers, Y.Y. et al. (2006). Autosomal dominant frontotemporal lobar degen-
Semantic dementia and fluent primary progressive eration due to the C9orf72 hexanucleotide repeat
aphasia: Two sides of the same coin? Brain: A Journal expansion: Late-onset psychotic clinical presentation.
of Neurology, 129: 3066–3080. Biological Psychiatry, 74 (5): 384–391.
Agosta, F., Henry, R.G., Migliaccio, R. et al. (2010). Goldman, J.S., Farmer, J.M., Wood, E.M. et al. (2005).
Language networks in semantic dementia. Brain: Comparison of family histories in FTLD subtypes and
A Journal of Neurology, 133: 286–299. related tauopathies. Neurology, 65: 1817–1819.
Alladi, S., Xuereb, J., Bak, T. et al. (2007). Focal cortical Gorno-Tempini, M.L., Hillis, A.E., Weintraub, S. et al.
presentations of Alzheimer’s disease. Brain: A Journal (2011). Classification of primary progressive aphasia
of Neurology, 130: 2636–2645. and its variants. Neurology, 76: 1006–1014.
Benton, A.L., Varney, N.R. and Hamsher, K.D. (1978). Green, H.A. and Patterson, K. (2009). Jigsaws –
Visuo-spatial judgement: A clinical test. Archives of A preserved ability in semantic dementia.
Neurology, 35: 364–367. Neuropsychologia, 47: 569–576.
Bozeat, S., Lambon Ralph, M.A., Patterson, K. and Henry, M.L., Beeson, P.M. and Rapcsak, S.Z. (2008).
Hodges, J. (2002). The influence of personal familiar- Treatment for anomia in semantic dementia. Seminars
ity and context on object use in semantic dementia. in Speech and Language, 29: 60–70.
Neurocase, 8: 127–134. Hodges, J.R., Michell, J., Dawson, K. et al. (2010).
Bozeat, S., Lambon Ralph, M.A., Patterson, K. et al. Semantic dementia: Demography, familial factors and
(2000). Non-verbal semantic impairment in semantic survival in a consecutive series of 100 cases. Brain:
dementia. Neuropsychologia, 38: 1207–1215. A Journal of Neurology, 133: 300–306.
Brambati, S.M., Rankin, K.P., Narvid, J. et al. (2009). Hodges, J.R. and Patterson, K. (1995). Is semantic memory
Atrophy progression in semantic dementia with asym- consistently impaired early in the course of Alzheimer’s
metric temporal involvement: A tensor-based mor- disease? Neuroanatomical and diagnostic implications.
phometry study. Neurobiology of Aging, 30: 103–111. Neuropsychologia, 33: 441–459.
Cappelletti, M., Butterworth, B. and Kopelman, M. (2001). Hodges, J.R. and Patterson, K. (2007). Semantic dementia:
Spared numerical abilities in a case of semantic A unique clinicopathological syndrome. The Lancet
dementia. Neuropsychologia, 39: 1224–1239. Neurology, 6: 1004–1014.
Cappelletti, M., Kopelman, M. and Butterworth, B. (2002). Hodges, J.R., Patterson, K., Oxbury, S. and Funnell,
Why semantic dementia drives you to the dogs (but E. (1992). Semantic dementia. Progressive fluent
not to the horses): A theoretical account. Cognitive aphasia with temporal lobe atrophy. Brain: A Journal of
Neuropsychology, 19: 483–503. Neurology, 115: 1783–1806.
Caselli, R.J., Windebank, A.J., Petersen, R.C. et al. (1993). Hoffman, P., Clarke, N., Jones, R.W. and Noonan, K.A.
Rapidly progressive aphasic dementia with motor neu- (2015). Vocabulary relearning in semantic dementia:
ron disease. Annals of Neurology, 33: 200–207. Positive and negative consequences of increasing vari-
Chan, D., Fox, N.C., Scahill, R.I. et al. (2001). Patterns ability in the learning experience. Neuropsychologia,
of temporal lobe atrophy in semantic dementia 76: 240–253.
and Alzheimer’s disease. Annals of Neurology, Hoffman, P., Jones, R.W. and Lambon-Ralph, M.A.
49: 433–442. (2012). The degraded concept representation system
Crutch, S.J. and Warrington, E.K. (2002). Preserved cal- in semantic dementia: Damage to pan-modal hub,
culation skills in a case of semantic dementia. Cortex, then visual spoke. Brain: A Journal of Neurology,
38: 389–399. 135: 3770–3780.
Desgranges, B., Matuszewski, V., Piolino, P. et al. (2007). Jokel, R. and Anderson, N.D. (2012). Quest for the best:
Anatomical and functional alterations in seman- efforts of errorless and active encoding on word re-
tic dementia: A voxel-based MRI and PET study. learning in semantic dementia. Neuropsychological
Neurobiology of Aging, 28 (12): 1904–1913. Rehabilitation, 22: 187–214.
Diehl, J., Grimmer, T., Drzezga, A. et al. (2004). Cerebral Josephs, K.A., Hodges, J.H., Snowden, J.S. et al.
metabolic patterns at early states of frontotempo- (2011). Neuropathological background of pheno-
ral dementia and semantic dementia: A PET study. typical variability in frontotemporal dementia. Acta
Neurobiology of Aging, 25: 1051–1056. Neuropathologica, 122: 137–153.
Semantic dementia 793

Josephs, K.A., Stroh, A., Dugger, B. and Dickson, Neary, D., Snowden, J.S., Gustafson, L. et al. (1998).
D.W. (2009). Evaluation of subcortical pathology Frontotemporal lobar degeneration. A consensus on
and clinical correlations in FTLD-U subtypes. Acta clinical diagnostic criteria. Neurology, 51: 1546–1554.
Neuropathologica, 118: 349–358. Neary, D., Snowden, J.S., Mann, D.M.A. et al. (1990).
Josephs, K.A., Whitwell, J.L., Vemuri, P. et al. (2008). Frontal lobe dementia and motor neuron disease.
The anatomic correlate of prosopagnosia in semantic Journal of Neurology, Neurosurgery, and Psychiatry,
dementia. Neurology, 71: 1628–1633. 53: 23–32.
Julien, C.L., Thompson, J.C., Neary, D. and Snowden, J.S. Ostberg, P. and Bogdanovic, N. (2011). Semantic dementia
(2008). Arithmetic knowledge in semantic demen- with lower motor neuron disease showing FTLD-TDP
tia: Is it invariably preserved? Neuropsychologia, type 3 pathology (sensu Mackenzie). Neuropathology,
46: 2732–2744. 31: 271–279.
Kiefer, M. and Pulvermüller, F. (2012). Conceptual rep- Patterson, K.E., Marshall, J.C. and Coltheart, M. (1985).
resentations in mind and brain: Theoretical develop- Surface Dyslexia: Neuropsychological and Cognitive
ments, current evidence and future directions. Cortex, Studies of Phonological Reading. Hove, London:
48 (7): 805–825. Erlbaum.
Kim, S.H., Seo, S.W., Go, S.M. et al. (2009). Semantic Patterson, K., Nestor, P.J. and Rogers, T.T. (2007). Where
dementia combined with motor neuron disease. do you know what you know? The representation
Journal of Clinical Neuroscience, 16: 1683–1685. of semantic knowledge in the human brain. Nature
Kindell, J., Sage, K., Keady, J. and Wilkinson, R. (2013). Reviews: Neuroscience, 8 (12): 976–987.
Adapting to conversation with semantic dementia: Pick A. (1892). Uber die Beziehungen der senilen hirnatro-
Using enactment as a compensatory strategy in every- phie zur aphasie. Prager Medizinische Wochenschrift,
day social situations. International Journal of Language 17: 165–167.
and Communication Disorders, 48: 497–507. Pickering-Brown, S.M., Richardson, A.M., Snowden, J.S.
Lambon Ralph, M.A., Graham, K.S., Ellis, A.W. and et al. (2002). Inherited frontotemporal dementia in
Hodges, J.R. (1998). Naming in semantic dementia – nine British families associated with intronic muta-
What matters? Neuropsychologia, 36: 775–784. tions in the tau gene. Brain: A Journal of Neurology,
Libon, D.J., Rascovsky, K., Powers, J. et al. (2013). 125: 732–751.
Comparative semantic profiles in semantic demen- Piwnica-Worms, K.E., Omar, R., Hailstone, J.C. and
tia and Alzheimer’s disease. Brain: A Journal of Warren, J.D. (2010). Flavour processing in semantic
Neurology, 136: 2497–2509. dementia. Cortex, 46: 761–768.
Luzzi, S., Snowden, J.S., Neary, D. et al. (2007). Distinct Pulvermüller, F., Cooper-Pye, E., Dine, C. et al. (2009).
patterns of olfactory impairment in Alzheimer’s dis- The word processing deficit in semantic dementia: All
ease, semantic dementia, frontotemporal dementia categories are equal, but some categories are more
and corticobasal degeneration. Neuropsychologia, equal than others. Journal of Cognitive Neuroscience,
45: 1823–1831. 22: 2027–2041.
Mackenzie, I.R.A., Neumann, M., Baborie, A. et al. (2011). Rogers, S.L. and Friedman, R.B. (2008). The underlying
A harmonized classification system for FTLD-TDP mechanisms of semantic memory loss in Alzheimer’s
pathology. Acta Neuropathologica, 122: 111–113. disease and semantic dementia. Neuropsychologia,
Mackenzie, I.R., Neumann, M., Bigio, E.H. et al. (2010). 46: 12–21.
Nomenclature for neuropathologic subtypes of fronto- Rohrer, J.D., McNaught, E., Foster, J. et al. (2008).
temporal lobar degeneration: Consensus recommen- Tracking progression in frontotemporal lobar degen-
dations: An update. Acta Neuropathologica, 119: 1–4. eration: Serial MRI in semantic dementia. Neurology,
Mahoney, C.J., Rohrer, J.D., Goll, J.C. et al. (2011). 71: 1445–1451.
Structural neuroanatomy and tinnitus and hyper- Rohrer, J.D., Warren, J.D., Modat, M. et al. (2009).
acusis in semantic dementia. Journal of Neurology, Patterns of cortical thinning in the language variants
Neurosurgery, and Psychiatry, 82: 1274–1278. of frontotemporal lobar degeneration. Neurology,
Mesulam, M. (1982). Slowly progressive aphasia with- 72: 1562–1569.
out generalised dementia. Annals of Neurology, Rosenfeld, M. (1909). Die partielle grosshirnatro-
11: 592–598. phie. Zeitschrift für Psychologie und Neurologie,
Mion, M., Patterson, K., Acosta-Cabronero, J. et al. 14: 115–130.
(2010). What the left and right anterior fusiform gyri Savage, S.A., Ballard, K.J., Piguet, O. and Hodges, J.R.
tell us about semantic memory. Brain: A Journal of (2013). Bringing words back to mind – Improving
Neurology, 133: 3256–3268. word production in semantic dementia. Cortex,
Mummery, C.J., Patterson, K., Price, C.J. et al. (2000). 49: 1823–1832.
A voxel-based morphometry study of semantic demen- Schwartz, M.F., Marin, O.S.M. and Saffran, E.M. (1979).
tia: Relationship between temporal lobe atrophy and Dissociations of language function in dementia: A case
semantic memory. Annals of Neurology, 47: 36–45. study. Brain and Language, 7: 277–306.
794 Dementia

Snowden, J.S. (2001). Commentary on Liepmann 1908 and Snowden, J.S., Thompson, J.C. and Neary, D. (2012).
Rosenfeld 1909. Cortex, 37: 563–571. Famous people knowledge and the right and left tem-
Snowden, J.S., Bathgate, D., Varma, A. et al. (2001). poral lobes. Behavioural Neurology, 25: 35–44.
Distinct behavioural profiles in frontotemporal demen- Snowden, J.S., Thompson, J.C., Stopford, C.L. et al. (2011).
tia and semantic dementia. Journal of Neurology, Clinical diagnosis of early-onset dementias: Diagnostic
Neurosurgery, and Psychiatry, 70: 323–332. accuracy and clinicopathological relationships. Brain:
Snowden, J.S., Goulding, P.J. and Neary, D. (1989). A Journal of Neurology, 134: 2478–2492.
Semantic dementia: A form of circumscribed atrophy. Tyrrell, P.J., Warrington, E.K., Frackowiak, R.S.J. and Rossor,
Behavioural Neurology, 2: 167–182. M.N. (1990a). Heterogeneity in progressive aphasia due
Snowden, J.S., Griffiths, H.L. and Neary, D. (1995). to focal cortical atrophy. A clinical and PET study. Brain:
Autobiographical experience and word meaning. A Journal of Neurology, 113: 1321–1336.
Memory, 3 (3–4): 225–246. Tyrrell, P.J., Warrington, E.K., Frackowiak, R.S.J. and
Snowden, J.S., Griffiths, H. and Neary, D. (1994). Semantic Rossor, M.N. (1990b). Progressive degeneration of the
dementia: Autobiographical contribution to pres- right temporal lobe studied with positron emission
ervation of meaning. Cognitive Neuropsychology, tomography. Journal of Neurology, Neurosurgery, and
11: 265–288. Psychiatry, 53: 1046–1050.
Snowden, J.S. and Neary, D. (2002). Relearning of ver- Warrington, E.K. (1975). The selective impairment
bal labels in semantic dementia. Neuropsychologia, of semantic memory. The Quarterly Journal of
40: 1715–1728. Experimental Psychology, 27 (4): 635–637.
Snowden, J.S., Neary, D. and Mann, D.M.A. (1996). Warrington, E.K. and James, M. (1991). The Visual
Frontotemporal Lobar Degneration: Frontotemporal Object and Space Perception Battery. Oxford, United
Dementia, Progressive Aphasia, Semantic Dementia. Kingdom: Pearson Assessment.
London, United Kingdom: Churchill-Livingstone. Westmacott, R., Black, S.E., Freedman, M. and
Snowden, J.S., Neary, D., Mann, D.M.A. et al. (1992). Moscovitch, M. (2003). The contribution of autobio-
Progressive language disorder due to lobar atrophy. graphical significance to semantic memory: Evidence
Annals of Neurology, 31: 174–183. from Alzheimer’s disease, semantic dementia and
Snowden, J.S., Thompson, J.C. and Neary, D. (2004). amnesia. Neuropsychologia, 42: 25–48.
Knowledge of famous faces and names in seman- Woollams, A.M., Cooper-Pye, E., Hodges, J.R. and Patterson,
tic dementia. Brain: A Journal of Neurology, K. (2008). Anomia: A doubly typical signature of semantic
127: 860–872. dementia. Neuropsychologia, 46: 2503–2514.
69
Primary progressive aphasia and posterior
cortical atrophy

JONATHAN D. ROHRER, SEBASTIAN J. CRUTCH AND JASON D. WARREN

This chapter consists of two sections covering the focal clinico-pathological entity with characteristic clinical, neu-
dementia syndromes of primary progressive aphasia (PPA) ropsychological and radiological findings (see Chapter 68).
(Section 69.1) and posterior cortical atrophy (PCA) (Section SvPPA presents usually with a ‘fluent’ aphasia (and there-
69.2). Though uncommon, these heterogeneous clinico- fore has previously been called fluent PPA: Adlam et al.,
pathological entities have a disproportionate importance 2006), however two other major PPA syndromes are associ-
due to the insights they provide into regional brain mecha- ated with ‘nonfluent’ language output: these nonfluent dis-
nisms of neurodegeneration and the particular challenges orders, progressive nonfluent aphasia (PNFA) or nonfluent
they pose for clinical diagnosis and management. variant PPA (nfvPPA) and logopenic aphasia (LPA) or logo-
penic variant PPA (lvPPA), are discussed in detail in this
chapter. It remains unclear if these are the only subtypes
of PPA and how this heterogeneous group of conditions
69.1 NONFLUENT AND LOGOPENIC maps onto the underlying genetic and pathological causes.
VARIANTS OF PRIMARY While early nfvPPA can resemble Broca’s aphasia and early
PROGRESSIVE APHASIA lvPPA may resemble conduction aphasia (Hachisuka et al.,
1999; reviewed in Rohrer et al., 2008a), the PPA subtypes do
69.1.1 INTRODUCTION AND NOSOLOGY not correspond closely with the acute aphasia syndromes
of stroke, due both to differing neuroanatomical patterns
PPA is a term that has been used to encompass all patients of involvement and the progressive nature of the disease.
with progressive language impairment as the initial fea- Initial consensus criteria for PNFA and SD (Neary et al.,
ture of a degenerative disorder (Mesulam, 1982; Mesulam, 1998) and other criteria for PPA as a unitary syndrome
2001; Mesulam, 2003). There is overlap with the frontotem- (Mesulam, 2001, 2003) have recently been updated, defin-
poral dementia (FTD) spectrum both pathologically and ing criteria for the semantic, nonfluent and logopenic vari-
genetically (Neary et al., 1998; Mackenzie and Rademakers, ants of PPA (Gorno-Tempini et al., 2011).
2007). Although some research groups have previously
classified all language disorders under this one term of PPA 69.1.2 SYNONYMS
(Mesulam 2001; Mesulam et al., 2003), it is clear that a num-
ber of subtypes can be identified (see reviews by Grossman
et al., 2004; Amici et al., 2006; Hodges and Patterson, 2007; ●● Nonfluent variant of PPA (nfvPPA), progressive non-
Mesulam et al., 2007; Rohrer et al., 2008a; Grossman, 2012; fluent aphasia (PNFA), nonfluent progressive aphasia
Mesulam et al., 2014). The fluency of speech output forms (NFPA), progressive apraxia of speech (AOS), agram-
the basis for a basic clinical descriptive subclassification matic variant of PPA.
of PPA; however, fluency is often problematic to opera- ●● Logopenic variant of PPA (lvPPA), logopenic/phonologi-
tionalize; it has been used to refer to reduced, effortful or cal aphasia (LPA), logopenic aphasia (LPA), progressive
sparse speech, however these may map onto distinct syn- logopenic aphasia (PLA).
dromes within the PPA syndrome. Semantic dementia ●● Semantic variant of PPA (svPPA), semantic dementia
(SD) or semantic variant PPA (svPPA) is a homogeneous (SD), fluent variant of PPA, ‘Gogi aphasia.’

795
796 Dementia

69.1.3 EPIDEMIOLOGY mutations fall within the FTLD-TDP spectrum pathologi-


cally. See Figure 69.1 for a schematic diagram of the clinico-
The PPA variants are collectively uncommon although there pathological associations of PPA.
are no large epidemiological studies of PPA. There are no
clearly associated non-genetic risk factors for PPA although 69.1.6 CLINICAL AND
one study has shown an increased frequency of learning dis- NEUROPSYCHOLOGICAL FEATURES
ability in patients with PPA and their first-degree relatives
(Rogalski et al., 2008a, 2013). Table 69.1 summarizes the clinical and neuropsychological
features of the PPA syndromes; svPPA is also included for
69.1.4 GENETICS the purposes of comparison.

PPA is usually sporadic but can rarely be familial (Krefft


69.1.7 NONFLUENT VARIANT OF PPA
et al., 2003; Goldman et al., 2005) with one study showing
that nfvPPA was associated with an autosomal dominant 69.1.7.1 Spontaneous speech
family history of PPA or FTLD in 6.9% of cases (Goldman
et al., 2005). Many familial PPA patients have been found to The characteristic features of nfvPPA are the presence of
have mutations in the progranulin (GRN) gene (Snowden agrammatism and hesitant, effortful speech secondary to
et al., 2006, 2007a, 2007b, 2007c; Mesulam et al., 2007; Beck a motor speech impairment that is usually characterised as
et al, 2008), and more recently a few familial PPA patients an AOS (Weintraub et al., 1990; Tyrrell et al., 1991; Turner
have been found to have mutations in the C9orf72 gene et al., 1996; Westbury and Bub, 1997; Neary et al., 1998;
(Rohrer et al., 2015). In contrast, lvPPA and svPPA are only Gorno-Tempini et al., 2004a, 2004b; Ogar et al., 2007).
very rarely familial. Beyond Mendelian genetics, there have Either of these features may dominate at presentation
been inconsistent results from studies investigating apoli- though there is some controversy as to which is the core
poprotein E (ApoE) and prion protein (PRNP) codon 129 deficit and how they are related to each other. Cases of pure
genotypes as well as microtubule-associated protein tau progressive AOS have been described; however, most cases
(MAPT) haplotype in PPA, and no clear genetic risk factors with longitudinal follow-up appear to develop agramma-
have emerged (Mesulam et al., 1997; Sobrido et al., 2003; tism/aphasia. Similarly, there are patients who have pure
Gorno-Tempini et al., 2004a, 2004b; Li et al., 2005; Acciarri agrammatism initially. Both of these symptoms are neuro-
et al., 2006; Rohrer et al., 2006; Daniele et al., 2009). anatomically related to left inferior frontal/insular atrophy
and it is therefore unsurprising that they commonly occur
69.1.5 PATHOLOGY together. Nonetheless, there are still some authorities who
exclude pure motor speech impairment in the absence of
The main pathological causes of PPA fall into three categories: other speech and language deficits from the PPA spectrum
the two major frontotemporal lobar degeneration (FTLD) (Mesulam et al., 2007). One other ongoing difficulty has
pathologies known as FTLD-tau and FTLD-TDP (where been in defining AOS (a problem of planning articulation)
TDP is the TAR DNA binding protein) and Alzheimer’s dis- so that it is uniformly applied by different centres (Ogar
ease (AD) pathology. SvPPA is typically a FTLD-TDP type C et al., 2005): the key features are difficulty initiating speech
disorder but can rarely be caused by FTLD-tau Pick’s disease and trial and error groping towards the correct word lead-
or globular glial tauopathy, or less commonly Alzheimer’s ing to dysprosodic, hesitant and effortful speech output.
pathology (Knibb et al., 2006; Snowden et al., 2007a, 2007b, Errors in patients with AOS are sound distortions (often
2007c; Pereira et al., 2009; Clark et al., 2015). In contrast, additions) and are termed apraxic or phonetic errors. In
nfvPPA can be caused by all three pathologies, although practice, these can be difficult to distinguish from phone-
patients with a prominent motor speech impairment are mic errors (i.e. errors due to the incorrectly selected sound)
associated particularly with the FTLD-tau pathologies, corti- although it is likely that both phonetic and phonemic errors
cobasal degeneration (CBD), progressive supranuclear palsy occur in nfvPPA. The importance of defining patients as
(PSP) and Pick’s disease (Josephs et al., 2006a) rather than nfvPPA with or without motor speech impairment is the
FTLD-TDP. However, a single case of progressive motor association with a particular pathological substrate: AOS
speech impairment and Alzheimer pathology is reported is likely to predict tau-positive histopathology (Josephs
(Gerstner et al., 2007). LvPPA appears to be most commonly et al., 2006a). In imaging studies, AOS has been associated
associated with AD pathology although some cases have with premotor and supplementary motor areas (Josephs
been reported with FTLD-TDP inclusions (Mesulam et al., et al, 2006a) as well as the insula and basal ganglia (Ogar
2008): In a parallel theme in the literature and prior to the et al., 2007). Most patients will eventually become mute
detailed description of lvPPA, a number of research groups although non-speech vocalizations such as laughter may be
described an atypical language variant of AD, many cases of present even when there is no speech (Rohrer et al., 2009c).
which might now be characterized as lvPPA (Greene et al., Early mutism in nfvPPA has been associated with left pars
1996; Galton et al., 2000; Alladi et al., 2007; Josephs et al., opercularis and basal ganglia atrophy (Gorno-Tempini
­
2008). Those PPA cases associated with GRN and C9orf72 et al., 2006).
Primary progressive aphasia and posterior cortical atrophy 797

Primary progressive aphasia (PPA)

Sporadic PPA Familial PPA

GRN-associated C9orf72-associated
svPPA nfvPPA IvPPA
aphasia aphasia

Pathological subtypes

FTLD-TDP FTLD-TDP FTLD-TDP FTLD-tau FTLD-tau FTLD-tau Alzheimer


Type A Type B Type C CBD/PSP Pick’s GGT pathology

Figure 69.1 (See colour insert.) Clinico-pathological and clinico-genetic associations in primary progressive aphasia (PPA).
PPA as a syndrome has heterogeneous genetic and pathological associations. However, the importance of subtyping PPA
is indicated by the third row of boxes, which show in a schematic manner the pathological associations with semantic vari-
ant PPA (svPPA), nonfluent variant PPA (nfvPPA), logopenic PPA (lvPPA) and with the familial progranulin (GRN)-associated
or C9orf72-associated form of PPA, where one pathological subtype tends to dominate. Each of the pathological subtypes
are indicated by a separate coloured box: frontotemporal lobar degeneration – TAR DNA binding protein (FTLD-TDP)
types A to C, FTLD-tau (CBD, corticobasal degeneration; PSP, progressive supranuclear palsy; GGT, globular glial tauopa-
thy; and Pick’s disease) and Alzheimer pathology.

69.1.7.2 Naming and single-word comprehension deficit. This has been studied in great-
comprehension est detail in a series of studies by Grossman et al. (Cooke
et al., 2003; Grossman and Moore, 2005; Peelle et al., 2007).
Other features include anomia that is initially mild with a sug- Patients perform poorly on complex sentences but relatively
gestion that verb naming may be affected more than nouns normally with simple sentence structures. One small func-
(broadly, the reverse pattern to that seen in svPPA: Hillis et al., tional magnetic resonance imaging (fMRI) study showed
2002, 2004). The underlying cognitive deficit causing anomia decreased activation in left ventral inferior frontal lobe
has not been fully established in nfvPPA and although there areas known to be associated with grammatical processing
is some evidence to implicate a primary word retrieval deficit, (Cooke et al., 2003).
this may not be the only or even the primary domain (Rogalski
et al., 2008b). Indeed, imaging studies have implicated a net- 69.1.7.4 Repetition
work of brain areas in association with anomia in nfvPPA,
particularly inferior frontal, lateral temporal and anterior pari- Patients with nfvPPA have early difficulty with repetition
etal lobes (Grossman et al., 2004; McMillan et al, 2004; Amici of polysyllabic words and sentences. This progresses such
et al, 2007). Single-word comprehension is essentially normal that later in the disease even monosyllabic word repetition
early in the disease but becomes affected a number of years becomes difficult. Impaired polysyllabic word repetition
into the illness (Blair et al., 2007). The cause of single-word with intact single-word comprehension has been proposed
comprehension impairment in nfvPPA as the disease develops as a simple bedside measure for distinguishing the nonflu-
is unclear though this may partly reflect a more generalized ent aphasias from svPPA, e.g. by asking the patient to repeat
auditory agnosia associated with posterior perisylvian damage ‘hippopotamus’ and then to identify a picture of a hippo-
(Uttner et al., 2006; Goll et al., 2010). potamus (Hodges et al., 2008).

69.1.7.3 Grammar and sentence 69.1.7.5 Literacy


comprehension
Patients with nfvPPA often have phonological dyslexia.
Like the expressive agrammatism seen in nfvPPA, Writing may be agrammatic with phonological errors,
patients also have receptive agrammatism and a sentence although tends to be affected later than speech.
798 Dementia

Table 69.1 Summary of language, neuropsychological and imaging features in PPA

svPPA nfvPPA lvPPA


Speech rate/pauses Normal Slow with hesitancy, Slow with word-finding
effortfulness secondary to pauses
motor speech disorder
and/or agrammatism
Speech errors Semantic Phonetic/apraxic errors Occasional phonemic errors,
Phonemic errors rare semantic errors
Apraxia of speech None Present None
Dysarthria None Can be present as the None
disease progresses
Naming Anomia Initially can be normal but Anomia
anomic as disease
progresses
Single-word comprehension Impaired secondary to Initially intact but in late Initially relatively intact but
verbal semantic disease becomes affected becomes affected as
impairment disease progresses
Sentence comprehension Normal initially but Impaired for complex Impaired for simple and
becomes impaired as sentences complex sentences
single-word
comprehension
deteriorates
Single-word repetition Normal Impaired with phonetic/ Impaired with occasional
apraxic errors phonemic errors
Sentence repetition Often normal initially but Can be impaired Very impaired
can make transposition
errors
Reading Surface dyslexia Phonological dyslexia Phonological dyslexia
Other cognitive domains Non-verbal semantic Can later develop dominant Verbal short-term
involved impairment can develop parietal impairment (phonological) memory
object agnosia/ (dyscalculia, limb apraxia) and other dominant
prosopagnosia particularly if associated parietal functions –
with CBS dyscalculia, limb apraxia
Behaviour Disinhibition, appetite Depression, apathy Apathy, anxiety
change
Additional neurological Usually none Can be associated with Usually none
abnormalities CBS, hemiparkinsonism,
PSP syndrome
Neuroanatomy Asymmetrical anteroinferior Asymmetrical L > R inferior Asymmetrical L > R posterior
temporal lobes frontal lobe/insula temporal–inferior parietal
lobes
Abbreviations: PPA, primary progressive aphasia; svPPA, semantic variant; nfvPPA, nonfluent variant PPA; lvPPA, logopenic PPA; CBS, corti-
cobasal syndrome; PSP, progressive supranuclear palsy; L, left hemisphere; R, right hemisphere.

69.1.7.6 Other cognitive domains features similar to the behavioural variant of FTD as the
disease progresses. Early on there may be a co-existing
Other domains including calculation and limb praxis may depression (Medina and Weintraub, 2007) with subsequent
also be affected (Joshi et al., 2003). Episodic memory, visuo- emergence of apathy, anxiety, and in particular, irritabil-
spatial and visuoperceptual skills generally remain rela- ity (Marczinski et al., 2004; Rosen et al., 2006; Banks and
tively intact in patients with nfvPPA. Weintraub, 2008).

69.1.7.7 Behavioural features 69.1.7.8 Additional neurological features


Although language impairment is the dominant feature, Recent work has emphasized that the presence of a motor
early on in the disease patients do develop behavioural speech disorder, in particular AOS, predicts the pathological
Primary progressive aphasia and posterior cortical atrophy 799

diagnosis of a tauopathy, either CBD or PSP (Josephs et al., 69.1.8.6 Other cognitive domains
2008, 2012, 2013). Similarly, during life a corticobasal
syndrome or PSP syndrome often accompanies nfvPPA Verbal or phonological short-term memory appears to be a
(Kertesz et al., 1999; Mochizuki et al., 2003; Gorno-Tempini key cognitive domain affected in lvPPA. This is reflected in
et al., 2004b; McMonagle et al., 2006a; Josephs et al., 2008). the poor forwards digit span exhibited by these patients and
Dysphagia can occur as a late feature in nfvPPA (Fuh et al., may underpin linguistic deficits such as impaired sentence
1994). repetition and sentence comprehension that help to define
Rarely, nfvPPA can be associated with motor neurone the syndrome (Gorno-Tempini et al., 2008). Consistent
­disease (MND)/amyotrophic lateral sclerosis (ALS) (Caselli with underlying Alzheimer’s pathology in a high pro-
et al., 1993; Tsuchiya et al., 2000; Bak et al., 2001; Catani et al., portion of cases, episodic memory appears frequently to
2004; da Rocha et al., 2007) although there are currently no become affected later in the disease (Gorno-Tempini et al.,
systematic studies of MND/ALS in PPA. Limited evidence 2004b) though less prominently than in typical amnestic
suggests that selective impairment of verb processing may be AD (tAD). However, few studies have addressed the lan-
an aphasic hallmark of PPA/MND (Bak et al., 2001). guage features of typical AD in comparison with lvPPA
(Westbury et al., 2002; Mendez et al., 2003; Taler and
Phillips, 2008).
69.1.8 LOGOPENIC VARIANT OF PPA
69.1.8.1 Spontaneous speech 69.1.8.7 Behavioural features
The spontaneous speech of patients with lvPPA is slow with As in nfvPPA, behavioural features are generally not salient
long word-finding pauses but without agrammatism or early in lvPPA but may emerge later in the course. One study
motor speech impairment. When the patient does actually identified depression, anxiety and apathy as behavioural
produce speech, output is relatively ‘fluent’ (non-effortful): features of lvPPA (Rosen et al., 2006).
an important bedside distinction from nfvPPA.
69.1.8.8 Additional neurological features
69.1.8.2 Naming and single-word
There are usually no additional neurological features in
comprehension lvPPA, and in particular (in contrast to nfvPPA) associated
Naming is affected to a greater extent than in nfvPPA (but parkinsonism or MND appear to be rare.
less prominently than with svPPa). Similarly, single-word
comprehension is relatively intact initially but may become 69.1.9 GENETIC PPA
affected as the disease progresses.
There have been few detailed studies of the aphasia associ-
ated with mutations in the GRN or C9orf72 genes. Initial
69.1.8.3 Grammar and sentence reports of GRN-associated aphasia suggested a nonfluent
comprehension aphasia, but more detailed studies have reported cases
with a prominent early anomia, no motor speech impair-
Patients with lvPPA have difficulty with both simple and ment and a relatively early single-word comprehension
complex sentence comprehension. This is thought to be deficit (Snowden et al., 2006, 2007a, 2007b, 2007c; Rohrer
secondary to a verbal short-term (phonological) memory et al., 2008b). Further detailed studies of patients with
deficit rather than a primary grammatical deficit (Gorno- GRN-and C9orf72-associated PPA will be required to
Tempini et al., 2008). understand where such patients fall within the nonfluent
PPA spectrum and their relationship to sporadic nfvPPA
69.1.8.4 Repetition and lvPPA.

Sentence repetition is particularly affected in patients with 69.1.10 OTHER FOCAL SPEECH AND
lvPPA with single-word repetition relatively intact at least
LANGUAGE SYNDROMES
early in the disease (Gorno-Tempini et al., 2008; Gorno-
Tempini et al., 2011). A number of other speech and language syndromes have
been described that do not clearly fit into the current
69.1.8.5 Literacy scheme of the progressive aphasias. As these presently lack
defined pathological or genetic associations, it remains
A study of reading in lvPPA suggested that patients have unclear exactly how they are related to svPPA, nfvPPA and
greater difficulty reading nonsense or pseudowords that lvPPA. Progressive articulatory disorders associated with
they characterized as phonological dyslexia, associated cortical disease have been described as ‘progressive dysar-
neuroanatomically with left temporo-parietal atrophy thria’ (Soliveri et al., 2003), ‘slowly progressive anarthria’
(Brambati et al., 2009a, 2009b). (Broussolle et al., 1996; Lucchelli and Papagno, 2005), or
800 Dementia

‘pure progressive aphemia’ (Cohen et al., 1993). Although it Knopman et al., 2009): with disease progression, there is
is clear that dysarthria commonly occurs as an accompani- spread from the left inferior frontal and insular cortex to
ment to AOS and aphasia in nfvPPA (Gorno-Tempini et al., involve superior temporal, middle and superior frontal
2004a; Ogar et al., 2007), it remains unclear whether iso- and anterior parietal lobes (Gorno-Tempini et al., 2004b;
lated ‘cortical dysarthrias’ eventually evolve into the same Rohrer et al., 2009b), and whole brain atrophy rates are
syndrome or whether they may remain isolated. Similarly, similar to other neurodegenerative diseases (1.6% per
progressive impairment of prosody has been described as a year) (Knopman et al., 2009).
selective deficit (Ghacibeh and Heilman, 2003; Luzzi et al., LvPPA has been less widely studied than nfvPPA (Gorno-
2008), but probably occurs more frequently as part of a PPA Tempini et al., 2004a, 2008; Wilson et al., 2009); however,
syndrome (Tsao et al., 2004). So-called ‘dynamic aphasia’, most studies have shown most significant involvement of
a selective disorder of verbal planning, may manifest as a the left posterior superior temporal and inferior parietal
progressive disorder independent of a widespread apathy or lobes with spread to the posterior cingulate and middle/
abulia (Warren et al., 2003; Perez et al., 2013). inferior temporal lobe as the disease unfolds. Consistent
with the hypothesis that most patients with lvPPA have
69.1.11 NEUROIMAGING Alzheimer pathology, multiple studies have now shown that
the majority of lvPPA cases have positive amyloid PET scans
There have been a number of imaging studies investigat- (Rabinovici et al., 2008; Leyton et al., 2011).
ing the neuroanatomical patterns of atrophy or hypo- There are few neuroimaging studies of GRN-associated
metabolism in nfvPPA and lvPPA (Nestor et al., 2003; PPA (Borroni et al., 2008; Cruchaga et al., 2008; Rohrer
Gorno-Tempini et al., 2004a; Grossman et al., 2004; Clark et al., 2008b). There tends to be asymmetrical left greater
et al., 2005; Zahn et al., 2005; Josephs et al., 2006b; Nestor than right hemisphere atrophy affecting frontal, temporal
et al., 2007; Ogar et al., 2007; Schroeter et al., 2007; Gorno- and (to a lesser extent) parietal lobes with somewhat more
Tempini et al., 2008; Lindberg et al., 2009; Rohrer et al., prominent posterior atrophy than usually occurs in nfvPPA
2009a; Galantucci et al., 2011; Mahoney et al., 2013; Caso and more anterior temporal lobe atrophy than occurs
et al., 2014; Rogalski et al., 2014). As with the clinical and in lvPPA.
neuropsychological findings discussed above, neuroim- Little information is available for nfvPPA associated with
aging findings in PPA have been relatively heterogeneous MND/ALS, but in single cases, bilateral (often more severe
indicating involvement mainly of the left hemisphere on the left) frontal or frontotemporal atrophy/hypometabo-
fronto-temporo-parietal language network (Sonty et al., lism has been reported (Bak et al, 2001; Catani et al., 2004;
2003; Vandenbulcke et al., 2005; Sonty et al., 2007). This is da Rocha et al., 2007).
largely attributable to a scarcity of well-defined, uniform
clinico-pathological cohorts: most studies have been per- 69.1.12 MANAGEMENT
formed in clinically defined cohorts that (at least in older
studies) are likely to contain patients with heterogeneous There are currently no symptomatic or curative phar-
tissue pathologies. Nonetheless, in most studies of nfvPPA, macological therapies for PPA. Small trials have yielded
the most significantly affected areas are in the left infe- inconclusive results (bromocriptine: Reed et al., 2004;
rior frontal lobe and anterior insula (Nestor et al., 2003; galantamine: Kertesz et al., 2008) and there are a number
Gorno-Tempini et al., 2004a; Ogar et al., 2007; Rohrer of unsubstantiated single case reports of the use of a vari-
et al., 2009a), with the left middle and superior frontal ety of drugs (Decker and Heilman, 2008; Tobinick, 2008).
and superior temporal lobes also commonly affected as Although there is no clear evidence base, many patients find
the disease evolves (Gorno-Tempini et al., 2004b; Ogar speech and language therapy helpful to provide communi-
et al., 2007; Rohrer et al., 2009b). Anterior parietal lobe cation strategies: the use of Augmentative and Alternative
involvement is less commonly reported but may occur Communication (AAC) methods are little studied but low-
particularly with disease progression and when there is technology input such as communication notebooks are
an accompanying corticobasal syndrome (Gorno-Tempini generally favoured over more high-technology devices such
et al., 2004b; Rohrer et al., 2009b). Pathologically con- as handheld computers (Rogers et al., 2000; Beukelman
firmed studies of nfvPPA have usually been based on et al., 2007). Genetic counselling is important in those with
mixed pathological groups (e.g. tau-positive but a mix- a family history and/or a known GRN or C9orf72 mutation.
ture of PSP, CBD and/or Pick’s disease) but have also
shown broadly similar involvement of anterior insula and 69.1.13 CONCLUSIONS
inferior frontal lobe (Josephs et al., 2006b; Rohrer et al.,
2009b). Metabolic (positron emission tomography/single- The progressive aphasias have provided uniquely valuable
photon emission computed tomography [PET/SPECT]) neurobiological perspectives on the processes of focal neuro-
brain imaging may be helpful in distinguishing nonflu- degeneration, yet there is continuing controversy within the
ent patients with Alzheimer pathology from others with field. How many subtypes of nonfluent PPA are there?; how
FTLD pathology (Nestor et al., 2007). There are few lon- can motor speech disorders/AOS best be operationalized?;
gitudinal studies of nfvPPA (Gorno-Tempini et al., 2004b; is the first symptom of agrammatism compared to a motor
Primary progressive aphasia and posterior cortical atrophy 801

speech disorder significant?; what are the clinico-patholog- Statistical Manual of Mental Disorders, Fourth Edition
ical correlates of the different subtypes?; how do patterns of [DSM-IV]).
brain atrophy change over time? Only studies with larger
cohorts of patients followed over a longer period of time will 69.2.2 SYNONYMS
resolve these key issues. None of the PPA syndromes has so
far achieved consensus criteria for diagnosis, and although Posterior cortical atrophy, Benson’s syndrome, progressive
new criteria are currently in development, it seems likely that posterior cortical dysfunction, biparietal AD, visual variant
only future research studies with detailed anatomical, patho- of AD.
logical and genetic correlation will yield sufficiently robust
criteria. The arrival of disease-modifying therapy is likely to 69.2.3 EPIDEMIOLOGY
accelerate this process: together with molecular biomark-
ers, criteria that allow pathological diagnosis to be predicted The exact prevalence and incidence of PCA are not known
with accuracy in life will ultimately be required for the ratio- and any figure is likely to be an underestimate because
nal application of effective treatments. of poor general knowledge of the syndrome’s existence.
However, in a study of 523 patients with AD at a single
specialist centre, a visual presentation (also labelled PCA)
was reported in 5% of the cohort (Snowden et al., 2007a,
69.2 POSTERIOR CORTICAL ATROPHY 2007b, 2007c). Studies comparing PCA and amnestic AD
suggest few epidemiological differences apart from age of
69.2.1 INTRODUCTION AND NOSOLOGY disease onset, which tends to be earlier in PCA around the
mid-50s and early 60s (e.g. Mendez et al., 2002; McMonagle
PCA is a clinico-radiologic syndrome characterized by et al., 2006b) although some studies report a wide distri-
­progressive decline in visual processing skills, relatively bution (40–86 years) (Tang-Wai et al., 2004). Group studies
intact memory and language in the early stages and atro- and reviews have indicated either no difference in preva-
phy of posterior brain regions (Benson et al., 1988; Mendez lence among the genders (e.g. Mendez et al., 2002, Renner
et al., 2002; Tang-Wai et al., 2004; see Crutch et al., 2012 et al., 2004; McMonagle et al., 2006b) or over-representation
for a review). Histopathological studies have identified amy- among women (e.g. Tang-Wai et al., 2004; Snowden et al.,
loid plaques and neurofibrillary tangles as the most com- 2007a, 2007b, 2007c).
mon underlying pathology, and PCA is now recognized
as a common form of atypical AD (McKhann et al., 2011;
69.2.4 GENETICS
Dubois et al., 2014). However, the occurrence of PCA associ-
ated with alternative aetiologies has led to renewed calls for The proportion of individuals with PCA with a positive
PCA to be considered as a distinct nosological entity with family history of dementia is not significantly different to
its own diagnostic criteria (Tang-Wai and Mapstone, 2006; individuals with typical AD (Mendez et al., 2002; Tang-Wai
Crutch et al., 2013a). et al., 2004). It is of note that there have been no convinc-
Many questions remain over whether PCA should be ing reports of an autosomal-dominant inheritance pattern
considered a unitary clinico-anatomical syndrome or as in PCA, and indeed of the 11 PCA patients (27.5%) in the
a collection of related but distinct syndromic subtypes. Tang-Wai et al. (2004) study who had a family history of
Extrapolating from basic neuroscience evidence of distinct a dementing illness, none of those family members had
cortical streams which process different kinds of visual a posterior cortical syndrome. These studies also report
information (Ungerleider and Mishkin, 1982; Goodale and no difference in the ApoE status of PCA relative to typi-
Milner, 1992), it has been suggested that separate parietal cal AD. However, a difference between the ApoE status of
(dorsal), occipitotemporal (ventral) and primary visual individuals with posterior cortical presentations of AD and
(posterior) forms of PCA exist (Galton et al., 2000). One amnestic AD has been suggested (Schott et al., 2006; van
difficulty is that such claims are based upon the obser- der Flier, 2006; Snowden et al., 2007a, 2007b, 2007c). Schott
vation of patterns of impairment in single cases, and the et al. (2006) reported that fewer patients with biparietal AD
existence of an occipitotemporal variant has not been sup- than typical AD have one or more ε4 alleles (20% and 86%,
ported by larger group studies (McMonagle et al., 2006b). respectively). In a larger study examining the relationship
These anterior-posterior and superior-inferior distinctions between cognitive profile and ApoE status in 302 patients
also fail to capture the pronounced asymmetry apparent with typical or atypical AD (Snowden et al., 2007a, 2007b,
in the neuropsychological and neuroimaging profiles of 2007c), the percentage of patients with a visual presenta-
many individuals with PCA (e.g. Freedman et al., 1991; tion possessing at least one ε4 allele was significantly lower
Snowden et al., 2007a, 2007b, 2007c). At a more general than patients with an amnestic presentation (30% and 82%,
level, it is noteworthy that with a relative preservation respectively; typical AD: 55%) and not different to a popula-
of episodic memory, individuals with PCA do not meet tion of 756 healthy individuals from the same region (27%;
all established criteria for dementia (e.g. Diagnostic and Payton et al., 2003). This evidence suggests that risk factors
802 Dementia

other than ApoE underpin posterior cortical syndromes in (24%) and grasp reflex (26%) was found to be compara-
AD. A recent collaborative study with 302 patients has con- ble to individuals with typical AD (Snowden et al., 2007a,
firmed that ApoE alters PCA risk, but with a smaller effect 2007b, 2007c). Approximately 30% of PCA patients exhibit
than for typical AD, and has identified three candidate loci asymmetrical left upper limb rigidity in addition to their
(near CNTNAP5, FAM46A and SEMA3C) that, if repli- visual dysfunction (Ryan et al., 2014). A recent systematic
cated, may provide insights into selective vulnerability and evaluation of oculomotor function using tests of fixation,
phenotypic diversity in AD (Schott et al., 2016). saccade and smooth pursuit has also shown that eye move-
ment abnormalities are near-ubiquitous in PCA (80% PCA,
69.2.5 PATHOLOGY 17% tAD, 5% controls) (Shakespeare et al., 2015a). Up to
25% of PCA patients may also experience visual halluci-
Histopathological studies have revealed that AD is the nations (Tang-Wai et al., 2004; McMonagle et al., 2006b).
most common underlying cause of PCA (Hof et al., 1989, In a series of 59 patients with PCA, visual hallucinations
1990; Ross et al., 1996; Galton et al., 2000; Renner et al., were observed in 13 individuals (22%) and were associated
2004; Tang-Wai et al., 2004; Alladi et al., 2007; Snowden with parkinsonism, rapid eye movement sleep behaviour
et al., 2007a, 2007b, 2007c). The distinction between PCA disorder, myoclonic jerks and not only atrophy of occipito-­
and typical AD lies in the distribution of this pathology. parietal regions but also disruption of thalamocortical cir-
Compared to individuals with typical AD, patients with cuits (Josephs et al., 2006b).
PCA show a much greater density of senile plaques and
neurofibrillary tangles in occipital cortex and regions of
69.2.6.3 Neuropsychological profiles
posterior parietal cortex and temporo-occipital junction,
while showing fewer pathological changes in more anterior The most frequently cited neuropsychological deficits
areas such as prefrontal c­ ortex (e.g. Levine et al., 1993; Ross in PCA are visuospatial and visuoperceptual deficits,
et al., 1996; Hof et al., 1997). However, AD is not the only including some or all of the features of Balint’s syndrome
aetiology responsible for the syndrome, with a small num- (simultanagnosia, oculomotor apraxia, optic ataxia, envi-
ber of cases attributable to CBD (Tang-Wai et al., 2003a; ronmental agnosia), Gerstmann’s syndrome (acalculia,
Renner et al., 2004), ­dementia with Lewy bodies (Tang-Wai agraphia, finger agnosia, left/right disorientation), alexia,
et al., 2003b; Renner et al., 2004), prion disease (Renner agraphia, acalculia and apraxia (Mendez et al., 2002;
et al., 2004; Victoroff et al., 1994) and so-called subcortical Renner et al., 2004; Tang-Wai et al., 2004; Charles and
gliosis (Victoroff et al., 1994). Hillis, 2005; McMonagle et al., 2006a; Whitwell et al.,
2007; Yong et al., 2014a). Neuropsychological studies of
69.2.6 CLINICAL AND PCA suggest that of these symptoms, alexia, agraphia,
NEUROPSYCHOLOGICAL simultanagnosia and optic ataxia are the most consistently
FEATURES identified features (e.g. McMonagle et al., 2006b; Kas et al.,
2011). Additional features reported in a proportion of
69.2.6.1 Proposed diagnostic features patients include agnosia for objects, faces and colours. In
addition to higher order visual processing deficits of object
Two broadly comparable sets of diagnostic criteria have and space perception, impairments of more basic visual
been proposed (Mendez et al., 2002; Tang-Wai et al., functions (e.g. edge detection, form and motion coher-
2004). Suggested core features include: (1) insidious onset ence) also affect the majority of PCA patients (Lehmann
and gradual progression; (2) presentation with visual et al., 2011b). Excessive crowding has recently been shown
complaints in the absence of ocular disease; (3) relatively to particularly limit object perception in the central vision
preserved episodic memory, verbal fluency and personal of PCA patients (Yong et al., 2014b). Overall, the plethora
insight; (4) presence of symptoms including visual agno- of associated posterior cognitive deficits have predict-
sia, simultanagnosia, optic ataxia, ocular apraxia, dys- able consequences for the performance of PCA patients
praxia and environmental disorientation; (5) absence on more general neuropsychological tests such as perfor-
of stroke or tumour. Supportive features include alexia, mance intelligence quotient (IQ) (often up to 30–40 points
ideomotor apraxia, agraphia, acalculia, onset before the lower than verbal IQ scores) and constructional tasks (e.g.
age of 65 years and neuroimaging evidence of PCA or Rey figure copy, clock drawing). Longitudinal studies have
hypoperfusion. demonstrated that anterograde memory functions, linguis-
tic skills and frontal lobe functions, which are sometimes
69.2.6.2 Additional neurological features strikingly preserved in the earlier stages of the condition,
do gradually deteriorate as individuals progress to a more
Neurological signs have been inconsistently reported in global dementia state (e.g. Levine et al., 1993; McMonagle
group studies of PCA, and estimates of symptom frequency et al., 2006a). The aphasic difficulties are characterized
are dependent upon the composition of the study popula- by progressive anomia and phonological impairment and
tion. However, among 24 PCA patients with probable AD, increasingly resemble the lvPPA syndrome described above
the frequency of extrapyramidal signs (41%), myoclonus (Crutch et al., 2012).
Primary progressive aphasia and posterior cortical atrophy 803

69.2.6.4 Unusual symptoms less thinning in the left entorhinal cortex among PCA patients
(Lehmann et al., 2011a) (see Figure 69.2).
Although individuals with PCA experience the loss of many Consistent with these structural findings of atrophic
visual functions, many also describe unusual new experi- changes, functional imaging techniques such as SPECT
ences or ‘positive perceptual phenomena’. These phenomena and PET imaging often reveal hypoperfusion and indicate
have not been widely recognized, but include abnormally hypometabolism in dorsal occipito-parietal more than
prolonged colour afterimages (Chan et al., 2001), reverse size ventral occipito-temporal regions (e.g. Pietrini et al., 1996;
phenomena (e.g. Stark et al., 1997; Yong et al., 2014a), the Aharon-Peretz et al., 1999; Goethals and Santens, 2001; Kas
perception of movement among static stimuli, and in one et al., 2011; Lehmann et al., 2013; Ossenkoppele et al., 2015).
case even the 180° upside-down reversal of vision (Crutch In addition to posterior regions, fludeoxyglucose-positron
et al., 2010). Anecdotally, individuals with PCA also report a emission tomography (FDG-PET) has revealed specific areas
range of localized sensory and pain phenomena, and distur- of hypometabolism in the frontal eye fields bilaterally, which
bances of balance and bodily orientation potentially linked may occur secondary to loss of input from occipito-parietal
to deranged visuo–vestibular interactions. Detailed inves- regions and underpin ocular apraxia in PCA (Nestor et al.,
tigation of these positive symptoms could potentially yield 2003). Pathophysiological studies using fMRI are lacking in
important new insights about the pathophysiology as well as PCA and may be especially pertinent in PCA as the positive
the phenomenology of PCA. perceptual phenomena described by these patients suggest
not merely loss of activity but aberrant activity in affected
69.2.7 OTHER FOCAL POSTERIOR cortical areas. Diffusion tensor imaging (DTI) has revealed
CORTICAL SYNDROMES extensive degeneration of the major anterior–posterior con-
necting fibre bundles and of commissural frontal lobe tracts
The proposed diagnostic criteria for PCA (Mendez et al., (Cerami et al., 2015). Molecular imaging indicates that in
2002; Tang-Wai et al., 2004) both consider ‘presentation individuals with PCA attributable to AD, the focal poste-
with visual complaints’ as a core feature of the syndrome. rior pattern of cortical dysfunction involvement is evident
However, some patients with AD present initially with focal with tau imaging but less clear-cut with amyloid imaging
deterioration in other cognitive domains such as praxis or where results indicate with focal tangle distribution rather
spelling (e.g. De Renzi, 1986; Green et al., 1995; Aharon- than a more global pattern of amyloid deposition, which
Peretz et al., 1999; Snowden et al., 2007a, 2007b, 2007c) is global (Lehmann et al., 2013; Ossenkoppele et al., 2015).
with relative early sparing of visual function. Often these These findings suggest that tau is more closely linked to
individuals then progress to a more general posterior corti- PCA symptomatology than amyloid.
cal syndrome with memory relatively spared until later in
the disease course and consequently could be considered 69.2.9 MANAGEMENT
to fall within the spectrum of PCA phenotypes. Given the
increasing awareness of non-AD pathologies underpinning There are currently no published studies examining the
PCA and the move towards considering PCA as a distinct ­effectiveness of acetyl cholinesterase therapy specifically in
nosologic entity in its own right, it may be that the available PCA. However, anecdotal and limited single case reports
diagnostic criteria for PCA will require some expansion to suggest that donepezil, rivastigmine and galantamine
include posterior cortical presentations, which are not pri- are associated in symptomatic benefit in a proportion of
marily visual in nature (Crutch et al., 2012, 2013b). the population (e.g. Kim et al., 2005), most likely those
­individuals with an underlying pathological diagnosis of
69.2.8 NEUROIMAGING AD or dementia with Lewy bodies. There is also insuffi-
cient understanding of the psychological impact of PCA;
As the term PCA suggests, the syndrome is associated with ­preliminary studies suggest comparable levels of anxiety
tissue loss primarily of the occipital, parietal and temporo- and low mood but lower levels of apathy in PCA compared
occipital cortices. This pattern of atrophy is often evident from with typical AD ( Shakespeare et al., 2015b) but a formal
visual inspection of structural magnetic resonance (MR) or study of the neuropsychiatric profile of PCA is yet to be
computed tomography (CT) images, but less specific radiolog- conducted.
ical findings such as generalized atrophy or normal volume for A lack of understanding of PCA in the wider community
age are also common (e.g. Sala et al., 1996; Galton et al., 2000; limits patients’ access to relevant services, and the care and
Mendez et al., 2002). Systematic evaluations of brain struc- advice which is provided is often inappropriate, catering
ture in PCA using automated methods have been conducted, to problems that are not significant (e.g. memory deficits)
with voxel-based morphometry revealing greater right pari- while failing to cater to functionally critical perceptual defi-
etal and less left medial temporal and hippocampal atrophy in cits (e.g. many activities in day centres and nursing homes
PCA patients compared with those with typical AD (Whitwell are visually mediated). Owing to the relative preservation of
et al., 2007; Lehmann et al., 2011a). In addition, direct cortical memory, language and personal insight particularly in the
thickness comparisons between PCA and typical AD revealed mild and moderate stages of the condition, individuals with
greater cortical thinning in the right superior parietal lobe and PCA are well-disposed to take advantage of peer support
804 Dementia

Figure 69.2 (See colour insert.) Regional percentage differences in cortical thickness in posterior cortical atrophy (PCA)
(N = 48) compared with typical amnestic Alzheimer’s disease (tAD; N = 30) for the left and right hemispheres. The colour
scale represents magnitude of cortical thickness difference. Red and yellow (positive values) represent lower cortical
­thickness in PCA subjects compared with tAD, whereas dark to light blue (negative values) represents greater cortical
thickness. (From Lehmann, M., et al., Cerebral Cortex, 21, 2122–2132, 2011a.)

meetings and group, couple and individual psychological studies regarding the classification of PCA at the disease
therapies where the need exists. Peer support meetings, in or syndrome level is likely to continue until more detailed
particular, provide an important opportunity for reducing diagnostic criteria and terminology are available. Better
social isolation, for sharing the experience of what is often understanding and awareness of the syndrome among the
a particularly long and difficult route to diagnosis and for medical and lay communities is necessary to improve the
exchanging practical tips and advice for managing problems support services and information provided to individu-
associated with the condition. Individuals with PCA often als with PCA and their families. Dedicated clinical trials
benefit from resources designed primarily for the blind and assessing the effectiveness of pharmacological and non-
partially sighted, such as talking watches, mobile phones pharmacological interventions are also required in this
with simplified displays, voice recognition software, talking small but significant population of patients with degenera-
books, culinary aids and lamps to increase ambient light tive conditions.
levels in the home. Referral to an ophthalmologist may also
be required for an individual to register as partially sighted
under statutory invalidity schemes, which can enable access REFERENCES
to financial and social benefits and services.
Acciarri, A., Masullo, C., Bizzarro, A. et al. (2006). ApoE
69.2.10 CONCLUSIONS epsilon2-epsilon4 genotype is a possible risk factor for
primary progressive aphasia. Annals of Neurology, 59
PCA is a debilitating and under-recognized focal degener- (2): 436–437.
ative condition, which is associated with a range of differ- Adlam, A.L., Patterson, K., Rogers, T.T. et al. (2006).
ent disease pathologies. Analogies can be drawn between Semantic dementia and fluent primary progressive
the key focal dementia syndromes of PCA and PPA: like aphasia: Two sides of the same coin? Brain, 129:
PPA, PCA at once provides a crucial window on the neu- 3066–3080.
robiology of neurodegeneration and raises substantial Aharon-Peretz, J., Israel, O., Goldsher, D. and Peretz,
nosological difficulties. The PCA syndrome justifies an A. (1999). Posterior cortical atrophy variants of
independent nosological status, but with AD as the most Alzheimer’s disease. Dementia and Geriatric Cognitive
common underlying cause, a lack of consistency between Disorders, 10 (6): 483–487.
Primary progressive aphasia and posterior cortical atrophy 805

Alladi, S., Xuereb, J., Bak, T. et al. (2007). Focal corti- Caso, F., Mandelli, M.L., Henry, M. et al. (2014). In vivo
cal presentations of Alzheimer’s disease. Brain, 130 signatures of nonfluent/agrammatic primary progres-
(Pt 10): 2636–2645. sive aphasia caused by FTLD pathology. Neurology,
Amici, S., Gorno-Tempini, M.L., Ogar, J.M. et al. (2006). An 82 (3): 239–247.
overview on primary progressive aphasia and its vari- Catani, M., Piccirilli, M., Geloso, M.C. et al. (2004). Rapidly
ants. Behavioural Neurology, 17 (2): 77–87. progressive aphasic dementia with motor neuron
Amici, S., Ogar, J., Brambati, S.M. et al. (2007). disease: A distinctive clinical entity. Dementia and
Performance in specific language tasks ­correlates Geriatric Cognitive Disorders, 17 (1–2): 21–28.
with regional volume changes in progressive Cerami, C., Crespi, C., Della Rosa, P.A. et al. (2015).
­aphasia. Cognitive and Behavioral Neurology, Brain changes within the visuo-spatial attentional
20 (4): 203–211. network in posterior cortical atrophy. Journal of
Bak, T.H., O’Donovan, D.G., Xuereb, J.H. et al. (2001). Alzheimer’s Disease, 43 (2): 385–395. doi: 10.3233/
Selective impairment of verb processing associated JAD-141275
with pathological changes in Brodmann areas 44 and Chan, D., Crutch, S.J. and Warrington, E.K. (2001). A
45 in the motor neurone disease-dementia-aphasia disorder of colour perception associated with abnor-
syndrome. Brain, 124 (Pt 1): 103–120. mal colour after-images: A defect of the primary
Banks, S.J. and Weintraub, S. (2008). Neuropsychiatric visual cortex. Journal of Neurology Neurosurgery and
symptoms in behavioral variant frontotemporal Psychiatry, 71 (4): 515–517.
dementia and primary progressive aphasia. Journal of Charles, R.F. and Hillis, AE. (2005). Posterior cortical atro-
Geriatric Psychiatry and Neurology, 21 (2): 133–141. phy: Clinical presentation and cognitive deficits com-
Beck, J., Rohrer, J.D., Campbell, T. et al. (2008). A distinct pared to Alzheimer’s disease. Behavioural Neurology,
clinical, neuropsychological and radiological pheno- 16 (1): 15–23.
type is associated with progranulin gene mutations in Clark, D.G., Charuvastra, A., Miller, B.L. et al. (2005).
a large UK series. Brain, 131 (Pt 3): 706–720. Fluent versus nonfluent primary progressive aphasia:
Benson, F., Davis, J. and Snyder, B.D. (1988). Posterior A comparison of clinical and functional neuroimaging
cortical atrophy. Archives of Neurology, features. Brain and Language, 94 (1): 54–60.
45: 789–793. Clark, C.N., Lashley, T., Mahoney, C.J. et al. (2015).
Beukelman, D.R., Fager, S., Ball, L. and Dietz, A. (2007). Temporal variant frontotemporal dementia is associ-
AAC for adults with acquired neurological condi- ated with globular glial tauopathy. Cognitive and
tions: A review. Augmentative and Alternative Behavioral Neurology, 28 (2): 92–97.
Communication, 23 (3): 230–242. Cohen, L., Benoit, N., Van Eeckhout, P. et al. (1993).
Blair, M., Marczinski, C.A., Davis-Faroque, N. and Pure progressive aphemia. Journal of Neurology,
Kertesz, A. (2007). A longitudinal study of lan- Neurosurgery and Psychiatry, 56 (8): 923–924.
guage decline in Alzheimer’s disease and fronto- Cooke, A., DeVita, C., Gee, J. et al. (2003). Neural basis
temporal dementia. Journal of the International for sentence comprehension deficits in frontotemporal
Neuropsychological Society, 13 (2): 237–245. dementia. Brain and Language, 85 (2): 211–221.
Borroni, B., Alberici, A., Premi, E. et al. (2008). Brain Cruchaga, C., Fernández-Seara, M.A., Seijo-Martínez,
magnetic resonance imaging structural changes in a M. et al. (2008). Cortical atrophy and language
pedigree of asymptomatic progranulin mutation carri- network reorganization associated with a novel
ers. Rejuvenation Research, 11 (3): 585–595. ­progranulin mutation. Cerebral Cortex, 19 (8):
Brambati, S.M., Ogar, J., Neuhaus, J. et al. (2009a). 1751–1760.
Reading disorders in primary progressive apha- Crutch, S.J., Lehmann, M., Schott, J.M. et al. (2012).
sia: A behavioral and neuroimaging study. Posterior cortical atrophy. The Lancet Neurology, 11:
Neuropsychologia, 47 (8–9): 1893–1900. 170–178.
Brambati, S.M., Rankin, K.P., Narvid, J. et al. (2009b). Crutch, S.J., Lehmann, M., Warren, J.D. and Rohrer, J.D.
Atrophy progression in semantic dementia with (2013a). The language profile of posterior cortical
asymmetric temporal involvement: A tensor-based atrophy. Journal of Neurology, Neurosurgery and
morphometry study. Neurobiology of Aging, Psychiatry, 84 (4): 460–466.
30 (1): 103–111. Crutch, S.J., Lehmann, M. and Warrington, E.K.
Broussolle, E., Bakchine, S., Tommasi, M. et al. (1996). (2010). Abnormal visual phenomena in posterior
Slowly progressive anarthria with late anterior opercu- ­cortical atrophy: Upside down reversal of vision,
lar syndrome: A variant form of frontal cortical atrophy static motion and after-image effects. Neurocase,
syndromes. Journal of the Neurological Sciences, 17 (2): 160–177.
144 (1–2): 44–58. Crutch, S.J., Schott, J.M., Rabinovici, G.D. et al. (2013b).
Caselli, R.J., Windebank, A.J., Petersen, R.C. et al. (1993). Shining a light on posterior cortical atrophy.
Rapidly progressive aphasic dementia and motor neu- Alzheimer’s & Dementia: Journal of the Alzheimer’s
ron disease. Annals of Neurology, 33 (2): 200–207. Association, 9 (4): 463–465.
806 Dementia

da Rocha, A.J., Valério, B.C., Buainain, R.P. et al. (2007). Goodale, M.A. and Milner, A.D. (1992). Separate visual
Motor neuron disease associated with non-fluent pathways for perception and action. Trends in
rapidly progressive aphasia: Case report and review Neurosciences, 15 (1): 20–25.
of the literature. European Journal of Neurology, Gorno-Tempini, M.L., Brambati, S.M., Ginex, V. et al.
14 (9): 971–975. (2008). The logopenic/phonological v­ ariant of
Daniele, A., Matera, M.G., Seripa, D. et al. (2009). APOE primary progressive aphasia. Neurology, 71
{varepsilon}2/{varepsilon}4 genotype a risk factor for (16): 1227–1234.
primary progressive aphasia in women. Archives of Gorno-Tempini, M.L., Dronkers, N.F., Rankin, K.P. et al.
Neurology, 66 (7): 910–912. (2004a). Cognition and anatomy in three variants of
De Renzi, E. (1986). Slowly progressive visual primary progressive aphasia. Annals of Neurology,
­agnosia or apraxia without dementia. Cortex, 22 (1): 55 (3): 335–346.
171–180. Gorno-Tempini, M.L., Hillis, A.E., Weintraub, S. et al.
Decker, D.A. and Heilman, K.M. (2008). Steroid treatment (2011). Classification of primary ­progressive
of primary progressive aphasia. Archives of Neurology, ­aphasia and its variants. Neurology, 76 (11):
65 (11): 1533–1535. 1006–1014.
Dubois, B., Feldman, H.H., Jacova, C. et al. (2014). Gorno-Tempini, M.L., Murray, R.C., Rankin, K.P.
Advancing research diagnostic criteria for Alzheimer’s et al. (2004b). Clinical, cognitive and anatomical
disease: The IWG-2 criteria. The Lancet Neurology, ­evolution from nonfluent progressive aphasia to
13: 614–629. ­corticobasal syndrome: A case report. Neurocase,
Freedman, L., Selchen, D.H., Black, S.E. et al. 10 (6): 426–436.
(1991). Posterior cortical dementia with alexia: Gorno-Tempini, M.L., Ogar, J.M., Brambati, S.M. et al.
Neurobehavioural, MRI, and PET findings. Journal (2006). Anatomical correlates of early mutism in
of Neurology, Neurosurgery and Psychiatry, progressive nonfluent aphasia. Neurology, 67 (10):
54 (5): 443–448. 1849–1851.
Fuh, J.L., Liao, K.K., Wang, S.J. and Lin, K.N. (1994). Green, R.C., Goldstein, F.C., Mirra, S.S. et al. (1995). Slowly
Swallowing difficulty in primary progressive aphasia: progressive apraxia in Alzheimer’s disease. Journal
A case report. Cortex, 30 (4): 701–705. of Neurology, Neurosurgery and Psychiatry, 59 (3):
Galantucci, S., Tartaglia, M.C., Wilson, S.M. et al. 312–315.
(2011). White matter damage in primary progressive Greene, J.D., Patterson, K., Xuereb, J. and Hodges,
aphasias: A diffusion tensor tractography study. Brain, J.R. (1996). Alzheimer disease and nonfluent
134 (Pt 10): 3011–3029. ­progressive aphasia. Archives of Neurology, 53 (10):
Galton, C.J., Patterson, K., Xuereb, J.H. and Hodges, J.R. 1072–1078.
(2000). Atypical and typical presentations of Alzheimer’s Grossman, M. (2012). The non-fluent/agrammatic variant
disease: A clinical, neuropsychological, neuroimaging of primary progressive aphasia. The Lancet Neurology,
and pathological study of 13 cases. Brain, 123 (Pt 3): 11 (6): 545–555.
484–498. Grossman, M., McMillan, C., Moore, P., Ding, L. et al.
Gerstner, E., Lazar, R.M., Keller, C. et al. (2007). (2004). What’s in a name: Voxel-based morphometric
A case of progressive apraxia of speech in analyses of MRI and naming difficulty in Alzheimer’s
­pathologically verified Alzheimer disease. Cognitive disease, frontotemporal dementia and corticobasal
and Behavioral Neurology: Official Journal of the degeneration. Brain, 127 (Pt 3): 628–649.
Society for Behavioral and Cognitive Neurology, Grossman, M. and Moore, P. (2005). A longitudinal study
20 (1): 15–20. of sentence comprehension difficulty in primary pro-
Ghacibeh, G.A. and Heilman, K.M. (2003). Progressive gressive aphasia. Journal of Neurology, Neurosurgery
affective aprosodia and prosoplegia. Neurology, and Psychiatry, 76 (5): 644–649.
60 (7): 1192–1194. Hachisuka, K., Uchida, M., Nozaki, Y. et al. (1999).
Goethals, M. and Santens, P. (2001). Posterior corti- Primary progressive aphasia presenting as
cal atrophy. Two case reports and a review of the ­conduction aphasia. Journal of the Neurological
­literature. Clinical Neurology and Neurosurgery, Sciences, 167 (2): 137–141.
103 (2): 115–119. Hillis, A.E., Oh, S. and Ken, L. (2004). Deterioration
Goldman, J.S., Farmer, J.M., Wood, E.M. et al. of naming nouns versus verbs in primary
(2005). Comparison of family histories in FTLD ­progressive aphasia. Annals of Neurology, 55
­subtypes and related tauopathies. Neurology, 65 (2): 268–275.
(11): 1817–1819. Hillis, A.E., Tuffiash, E. and Caramazza, A. (2002).
Goll, J.C., Crutch, S.J., Loo, J. et al. (2010). Non-verbal Modality-specific deterioration in naming verbs in
sound processing in the primary progressive aphasias. nonfluent primary progressive aphasia. Journal of
Brain, 133 (Pt 1): 272–285. Cognitive Neuroscience, 14 (7): 1099–1108.
Primary progressive aphasia and posterior cortical atrophy 807

Hodges, J.R., Martinos, M., Woollams, A.M. et al. (2008). Kertesz, A., Davidson, W. and Munoz, D.G. (1999).
Repeat and point: Differentiating semantic demen- Clinical and pathological overlap between fronto-
tia from progressive non-fluent aphasia. Cortex, temporal dementia, primary progressive aphasia
44 (9): 1265–1270. and ­corticobasal degeneration: The Pick com-
Hodges, J.R. and Patterson, K. (2007). Semantic dementia: plex. Dementia and Geriatric Cognitive Disorders,
A unique clinicopathological syndrome. The Lancet 10 (Suppl. 1): 46–49.
Neurology, 6 (11): 1004–1014. Kertesz, A., Morlog, D., Light, M. et al. (2008).
Hof, P.R., Bouras, C., Constantinidis, J. and Galantamine in frontotemporal dementia and primary
Morrison, J.H. (1989). Balint’s syndrome in progressive aphasia. Dementia and Geriatric Cognitive
Alzheimer’s disease: Specific disruption of the Disorders, 25 (2): 178–185.
occipito-parietal visual pathway. Brain Research, Kim, E., Lee, Y., Lee, J. and Han, S.H. (2005). A case
493 (2): 368–375. with cholinesterase inhibitor responsive asymmetric
Hof, P.R., Bouras, C., Constantinidis, J. and Morrison, posterior cortical atrophy. Clinical Neurology and
J.H. (1990). Selective disconnection of specific visual Neurosurgery, 108 (1): 97–101.
association pathways in cases of Alzheimer’s dis- Knibb, J.A., Xuereb, J.H., Patterson, K. and Hodges,
ease presenting with Balint’s syndrome. Journal of J.R. (2006). Clinical and pathological characteriza-
Neuropathology and Experimental Neurology, 49 (2): tion of progressive aphasia. Annals of Neurology,
168–184. 59 (1): 156–165.
Hof, P.R., Vogt, B.A., Bouras, C. and Morrison, J.H. Knopman, D.S., Jack, C.R. Jr., Kramer, J.H. et al. (2009).
(1997). Atypical form of Alzheimer’s disease with Brain and ventricular volumetric changes in fronto-
prominent posterior cortical atrophy: A review temporal lobar degeneration over 1 year. Neurology,
of lesion distribution and circuit d­ isconnection 72 (21): 1843–1849.
in cortical visual pathways. Vision Research, Krefft, T.A., Graff-Radford, N.R., Dickson, D.W.
37 (24): 3609–3625. et al. (2003). Familial primary progressive aphasia.
Josephs, K.A. and Duffy, J.R. (2008). Apraxia of speech Alzheimer Disease and Associated Disorders, 17 (2):
and nonfluent aphasia: A new clinical marker 106–112.
for ­corticobasal degeneration and progressive Lehmann, M., Barnes, J., Ridgway, G.R. et al. (2011a).
­supranuclear palsy. Current Opinion in Neurology, Basic visual function and cortical thickness pat-
21 (6): 688–692. terns in posterior cortical atrophy. Cerebral Cortex,
Josephs, K.A., Duffy, J.R., Strand, E.A. et al. (2006a). 21: 2122–2132.
Clinicopathological and imaging correlates of Lehmann, M., Crutch, S.J., Ridgway, G.R. et al. (2011b).
­progressive aphasia and apraxia of speech. Brain, Cortical thickness and voxel-based morphometry in
129 (Pt 6): 1385–1398. posterior cortical atrophy and typical Alzheimer’s dis-
Josephs, K.A., Duffy, J.R., Strand, E.A. et al. (2012). ease. Neurobiology of Aging, 32: 1466–1476.
Characterizing a neurodegenerative syndrome: Primary Lehmann, M., Ghosh, P.M., Madison, C. et al. (2013).
progressive apraxia of speech. Brain, 135 (Pt 5): Diverging patterns of amyloid deposition and hypo-
1522–1536. metabolism in clinical variants of probable Alzheimer’s
Josephs, K.A., Duffy, J.R., Strand, E.A. et al. (2013). disease. Brain, 136 (Pt 3): 844–858.
Syndromes dominated by apraxia of speech show dis- Levine, D.N., Lee, J.M. and Fisher, C.M. (1993). The visual
tinct characteristics from agrammatic PPA. Neurology, variant of Alzheimer’s disease: A clinicopathologic
81 (4): 337–345. case study. Neurology, 43 (2): 305–313.
Josephs, K.A., Whitwell, J.L., Boeve, B.F. et al. (2006b). Leyton, C.E., Villemagne, V.L., Savage, S. et al. (2011).
Visual hallucinations in posterior cortical atrophy. Subtypes of progressive aphasia: Application of the
Archives of Neurology, 63 (10): 1427–1432. International Consensus Criteria and validation using
Josephs, K.A., Whitwell, J.L., Duffy, J.R. et al. (2008). β-amyloid imaging. Brain, 134 (Pt 10): 3030–3043.
Progressive aphasia secondary to Alzheimer disease vs Li, X., Rowland, L.P., Mitsumoto, H. et al. (2005). Prion
FTLD pathology. Neurology, 70 (1): 25–34. protein codon 129 genotype prevalence is altered in
Joshi, A., Roy, E.A., Black, S.E. and Barbour K. (2003). primary progressive aphasia. Annals of Neurology,
Patterns of limb apraxia in primary progressive apha- 58 (6): 858–864.
sia. Brain and Cognition, 53 (2): 403–407. Lindberg, O., Ostberg, P., Zandbelt, B.B. et al. (2009).
Kas, A., de Souza, L.C., Samri, D. et al. (2011). Neural cor- Cortical morphometric subclassification of fronto-
relates of cognitive impairment in posterior cortical temporal lobar degeneration. American Journal of
atrophy. Brain, 134: 1464–1478. Neuroradiology, 30 (6): 1233–1239.
Kertesz, A., Davidson, W., McCabe, P. et al. (2003). Lucchelli, F. and Papagno, C. (2005). Is slowly ­progressive
Primary progressive aphasia: Diagnosis, varieties, evo- anarthria a "pure" motor-speech disorder?
lution. Journal of the International Neuropsychological Evidence from writing performance. Neurocase,
Society, 9 (5): 710–719. 11 (3): 234–241.
808 Dementia

Luzzi, S., Viticchi, G., Piccirilli, M. et al. (2008). Foreign Mesulam, M.M. (2003). Primary progressive aphasia:
accent syndrome as the initial sign of primary progres- A language-based dementia. New England Journal of
sive aphasia. Journal of Neurology, Neurosurgery and Medicine, 349 (16): 1535–1542.
Psychiatry, 79 (1): 79–81. Mesulam, M.M., Grossman, M., Hillis, A. et al. (2003).
Mackenzie, I.R. and Rademakers, R. (2007). The molecular The core and halo of primary progressive apha-
genetics and neuropathology of frontotemporal lobar sia and semantic dementia. Annals of Neurology,
degeneration: Recent developments. Neurogenetics, 54 (Suppl. 5): S11–S14.
8 (4): 237–248. Mesulam, M.M., Johnson, N., Grujic, Z. and Weintraub, S.
Mahoney, C.J., Malone, I.B., Ridgway, G.R. et al. (2013). (1997). Apolipoprotein E genotypes in primary pro-
White matter tract signatures of the progressive apha- gressive aphasia. Neurology, 49 (1): 51–55.
sias. Neurobiology of Aging, 34 (6): 1687–1699. Mesulam, M.M., Rogalski, E.J., Wieneke, C. et al. (2014).
Marczinski, C.A., Davidson, W. and Kertesz, A. (2004). Primary progressive aphasia and the evolving neu-
A longitudinal study of behavior in frontotemporal rology of the language network. Nature Reviews
dementia and primary progressive aphasia. Cognitive Neurology, 10 (10): 554–69.
and Behavioral Neurology, 17 (4): 185–190. Mochizuki, A., Ueda, Y., Komatsuzaki, Y. et al. (2003).
McKhann, G.M., Knopman, D.S., Chertkow, H. et al. (2011). Progressive supranuclear palsy presenting with
The diagnosis of dementia due to Alzheimer’s disease: ­primary progressive aphasia: Clinicopathological
Recommendations from the National Institute on report of an autopsy case. Acta Neuropathologica,
Aging-Alzheimer’s Association workgroups on diagnos- 105 (6): 610–614.
tic guidelines for Alzheimer’s disease. Alzheimer’s & Neary, D., Snowden, J.S., Gustafson, L. et al. (1998).
Dementia: Journal of the Alzheimer’s Association, Frontotemporal lobar degeneration: A consen-
7: 263–269. sus on clinical diagnostic criteria. Neurology,
McMillan, C., Gee, J., Moore, P. et al. (2004). 51: 1546–1554.
Confrontation naming and morphometric analyses of Nestor, P.J., Balan, K., Cheow, H.K. et al. (2007).
structural MRI in frontotemporal dementia. Dementia Nuclear imaging can predict pathologic diagnosis
and Geriatric Cognitive Disorders, 17 (4): 320–323. in ­progressive nonfluent aphasia. Neurology, 68 (3):
McMonagle, P., Blair, M. and Kertesz, A. (2006a). 238–239.
Corticobasal degeneration and progressive aphasia. Nestor, P.J., Caine, D., Fryer, T.D. et al. (2003). The
Neurology, 67 (8): 1444–1451. topography of metabolic deficits in posterior cortical
McMonagle, P., Deering, F., Berliner, Y. and Kertesz, atrophy (the visual variant of Alzheimer’s disease) with
A.(2006b). The cognitive profile of posterior cortical FDG-PET. Journal of Neurology, Neurosurgery and
atrophy. Neurology, 66 (3): 331–338. Psychiatry, 74 (11): 1521–1529.
Medina, J. and Weintraub, S. (2007). Depression in Nestor, P.J., Graham, N.L., Fryer, T.D. et al. (2003).
primary progressive aphasia. Journal of Geriatric Progressive non-fluent aphasia is associated with
Psychiatry and Neurology, 20 (3): 153–160. hypometabolism centred on the left anterior insula.
Mendez, M.F., Clark, D.G., Shapira, J.S. and Cummings, J.L. Brain, 126 (Pt 11): 2406–2418.
(2003). Speech and language in progressive nonflu- Ogar, J.M., Dronkers, N.F., Brambati, S.M. et al. (2007).
ent aphasia compared with early Alzheimer’s disease. Progressive nonfluent aphasia and its characteris-
Neurology, 61 (8): 1108–1113. tic motor speech deficits. Alzheimer Disease and
Mendez, M.F., Ghajarania, M. and Perryman, K.M. (2002). Associated Disorders, 21 (4): S23–S30.
Posterior cortical atrophy: Clinical characteristics and Ogar, J., Slama, H., Dronkers, N. et al. (2005). Apraxia
differences compared to Alzheimer’s disease. Dementia of speech: An overview. Neurocase, 11 (6):
and Geriatric Cognitive Disorders, 14 (1): 33–40. 427–432.
Mesulam, M., Johnson, N., Krefft, T.A. et al. (2007). Ossenkoppele, R., Schonhaut, D.R., Baker, S.L. et al.
Progranulin mutations in primary progressive aphasia: (2015). Tau, amyloid, and hypometabolism in a patient
The PPA1 and PPA3 families. Archives of Neurology, with posterior cortical atrophy. Annals of Neurology,
64 (1): 43–47. 77 (2): 338–342.
Mesulam, M., Wicklund, A., Johnson, N. et al. (2008). Payton, A., Holland, F., Diggle, P. et al. (2003). Cathepsin
Alzheimer and frontotemporal pathology in subsets D exon 2 polymorphism associated with general
of primary progressive aphasia. Annals of Neurology, intelligence in a healthy older population. Molecular
63 (6): 709–719. Psychiatry, 8 (1): 14–18.
Mesulam, M.M. (1982). Slowly progressive aphasia Peelle, J.E., Cooke, A., Moore, P. et al. (2007). Syntactic
without generalized dementia. Annals of Neurology, and thematic components of sentence processing
11 (6): 592–598. in progressive nonfluent aphasia and nonaphasic
Mesulam, M.M. (2001). Primary progressive aphasia. ­frontotemporal dementia. Journal of Neurolinguistics,
Annals of Neurology, 49 (4): 425–432. 20 (6): 482–494.
Primary progressive aphasia and posterior cortical atrophy 809

Pereira, J.M., Williams, G.B., Acosta-Cabronero, J. et al. Rohrer, J.D., Rossor, M.N. and Warren, J.D. (2009a).
(2009). Atrophy patterns in histologic vs clinical group- Neologistic jargon aphasia and agraphia in primary
ings of frontotemporal lobar degeneration. Neurology, progressive aphasia. Journal of the Neurological
72 (19): 1653–1660. Sciences, 277 (1–2): 155–159.
Perez, D.L., Dickerson, B.C., McGinnis, S.M. et al. (2013). Rohrer, J.D., Warren, J.D., Barnes, J. et al. (2008b).
You don’t say: Dynamic aphasia, another variant of Mapping the progression of progranulin-associated
primary progressive aphasia? Journal of Alzheimer’s frontotemporal lobar degeneration. Nature Clinical
Disease, 34 (1): 139–144. Practice Neurology, 4 (8): 455–460.
Pietrini, P., Furey, M.L., GraffRadford, N. et al. (1996). Rohrer, J.D., Warren, J.D., Modat, M. et al. (2009b).
Preferential metabolic involvement of visual cor- Patterns of cortical thinning in the language variants
tical areas in a subtype of Alzheimer’s disease: of frontotemporal lobar degeneration. Neurology,
Clinical implications. American Journal of Psychiatry, 72 (18): 1562–1569.
153 (10): 1261–1268. Rohrer, J.D., Warren, J.D. and Rossor, M.N. (2009c).
Rabinovici, G.D., Jagust, W.J., Furst, A.J. et al. (2008). Abnormal laughter-like vocalisations replacing
Abeta amyloid and glucose metabolism in three speech in primary progressive aphasia. Journal of the
variants of primary progressive aphasia. Annals of Neurological Sciences, 284 (1): 120–123.
Neurology, 64 (4): 388–401. Rosen, H.J., Allison, S.C., Ogar, J.M. et al. (2006).
Reed, D.A., Johnson, N.A., Thompson, C. et al. (2004). Behavioral features in semantic dementia vs
A clinical trial of bromocriptine for treatment of other forms of progressive aphasias. Neurology,
­primary progressive aphasia. Annals of Neurology, 67 (10): 1752–1756.
56 (5): 750. Ross, S.J., Graham, N., Stuart Green, L. et al. (1996).
Renner, J.A., Burns, J.M., Hou, C.E. et al. (2004). Progressive biparietal atrophy: An atypical p ­ resentation
Progressive posterior cortical dysfunction: A clinico- of Alzheimer’s disease. Journal of Neurology,
pathologic series. Neurology, 63 (7): 1175–1180. Neurosurgery and Psychiatry, 61 (4): 388–395.
Rogalski, E., Cobia, D., Martersteck, A. et al. (2014). Ryan, N.S., Shakespeare, T.J., Lehmann, M. et al. (2014).
Asymmetry of cortical decline in subtypes of primary Motor features in posterior cortical atrophy and
progressive aphasia. Neurology, 83 (13): 1184–1191. their imaging correlates. Neurobiology of Aging,
Rogalski, E., Johnson, N., Weintraub, S. and Mesulam, M. 35 (12): 2845–2857.
(2008a). Increased frequency of learning disability Sala, S.D., Spinnler, H. and Trivelli, C. (1996). Slowly pro-
in patients with primary progressive aphasia and gressive impairment of spatial exploration and visual
their first-degree relatives. Archives of Neurology, perception. Neurocase, 2 (4): 299–323.
65 (2): 244–248. Schott, J.M., Crutch, S.J., Carrasquillo, M.M. et al. (2016)
Rogalski, E., Rademaker, A., Mesulam, M. and Weintraub, S. Genetic risk factors for the posterior cortical atro-
(2008b). Covert processing of words and pictures phy variant of Alzheimer’s disease. Alzheimer’s &
in nonsemantic variants of primary progressive aphasia. Dementia: Journal of the Alzheimer’s Association,
Alzheimer Disease and Associated Disorders, 22 (4): pii: S1552-5260(16)00077-7.
343–351. Schott, J.M., Ridha, B.H., Crutch, S.J. et al. (2006).
Rogalski, E., Weintraub, S. and Mesulam, MM. (2013). Are Apolipoprotein E genotype modifies the pheno-
there susceptibility factors for primary progressive type of Alzheimer disease. Archives of Neurology,
aphasia? Brain and Language, 127 (2): 135–138. 63 (1): 155–156.
Rogers, M.A., King, J.M. and Alarcon, N.B. (2000). Schroeter, M.L., Raczka, K., Neumann, J. and Yves
Proactive management of primary progressive apha- von Cramon, D. (2007). Towards a nosology for
sia. In Beukelman, D.R. Yorkston, K.M. and Reichle, J. ­frontotemporal lobar degenerations-a ­meta-­analysis
(eds.), Augmentative and Alternative Communication involving 267 subjects. Neuroimage, 36 (3): 497–510.
for Adults with Acquired Neurologic Disorders. Shakespeare, T.J., Kaski, D., Yong, K.X. et al.
Baltimore, MD: Brookes. (2015a). Abnormalities of fixation, saccade and
Rohrer, J.D., Isaacs, A.M., Mizielinska, S. et al. (2015). ­pursuit in ­posterior cortical atrophy. Brain, 138 (Pt 7):
C9orf72 expansions in frontotemporal dementia and 1976–1991.
amyotrophic lateral sclerosis. The Lancet Neurology, Shakespeare, T.J., Yong, K.X., Foxe, D. et al. (2015b).
14 (3): 291–301. Pronounced impairment of everyday skills and
Rohrer, J.D., Knight, W.D., Warren, J.E. et al. (2008a). self-care in posterior cortical atrophy. Journal of
Word-finding difficulty: A clinical analysis of the pro- Alzheimer’s Disease, 43 (2): 381–384.
gressive aphasias. Brain, 131 (Pt 1): 8–38. Snowden, J., Neary, D. and Mann, D. (2007a).
Rohrer, J.D., Mead, S., Omar, R. et al. (2006). Prion protein Frontotemporal lobar degeneration: Clinical and
(PRNP) genotypes in frontotemporal lobar degenera- pathological relationships. Acta Neuropathologica,
tion syndromes. Annals of Neurology, 60 (5): 616. 114 (1): 31–38.
810 Dementia

Snowden, J.S., Pickering-Brown, S.M., Du Plessis, D. et al. Tsao, J.W., Dickey, D.H. and Heilman, K.M. (2004).
(2007b). Progressive anomia revisited: Focal degen- Emotional prosody in primary progressive aphasia.
eration associated with progranulin gene mutation. Neurology, 63 (1): 192–193.
Neurocase, 13 (5): 366–377. Tsuchiya, K., Ozawa, E., Fukushima, J. et al. (2000). Rapidly
Snowden, J.S., Pickering-Brown, S.M., Mackenzie, progressive aphasia and motor neuron disease:
I.R. et al. (2006). Progranulin gene mutations A clinical, radiological, and pathological study of an
­associated with frontotemporal dementia and autopsy case with circumscribed lobar atrophy. Acta
­progressive ­non-fluent aphasia. Brain, 129 (Pt 11): Neuropathologica, 99 (1): 81–87.
3091–3102. Turner, R.S., Kenyon, L.C., Trojanowski, J.Q. et al. (1996).
Snowden, J.S., Stopford, C.L., Julien, C.L. et al. (2007c). Clinical, neuroimaging, and pathologic features of
Cognitive phenotypes in Alzheimer’s disease and progressive nonfluent aphasia. Annals of Neurology,
genetic risk. Cortex, 43 (7): 835–845. 39 (2): 166–173.
Sobrido, M.J., Abu-Khalil, A., Weintraub, S. et al. Tyrrell, P.J., Kartsounis, L.D., Frackowiak, R.S. et al. (1991).
(2003). Possible association of the tau H1/H1 geno- Progressive loss of speech output and orofacial dys-
type with primary progressive aphasia. Neurology, praxia associated with frontal lobe hypometabolism.
60 (5): 862–864. Journal of Neurology, Neurosurgery and Psychiatry,
Soliveri, P., Piacentini, S., Carella, F. et al. (2003). 54 (4): 351–357.
Progressive dysarthria: Definition and clinical follow- Ungerleider, L.G. and Mishkin, M. (1982). Two corti-
up. Neurological Sciences, 24 (3): 211–212. cal visual systems. In Ingle, D.J., Mansfield, R.J.W.,
Sonty, S.P., Mesulam, M.M., Thompson, C.K. et al. Goodale, M.D. (eds.), The Analysis of Visual Behaviour.
(2003). Primary progressive aphasia: PPA and Cambridge, MA: MIT Press, pp. 549–586.
the ­language network. Annals of Neurology, Uttner, I., Mottaghy, F.M., Schreiber, H. et al. (2006).
53 (1): 35–49. Primary progressive aphasia accompanied by envi-
Sonty, S.P., Mesulam, M.M., Weintraub, S. et al. (2007). ronmental sound agnosia: A neuropsychological, MRI
Altered effective connectivity within the language and PET study. Psychiatry Research Neuroimaging,
network in primary progressive aphasia. The Journal of 146 (2): 191–197.
Neuroscience, 27 (6): 1334–1345. van der Flier, W.M., Schoonenboom, S.N.M.,
Stark, M.E., Grafman, J., Fertig, E. (1997). A restricted Pijnenburg, Y.A.L. et al. (2006). The effect of APOE
‘spotlight’ of attention in visual object recognition. genotype on clinical phenotype in Alzheimer disease.
Neuropsychologia, 35 (9): 1233–1249. Neurology, 67 (3): 526–527.
Taler, V. and Phillips, N.A. (2008). Language perfor- Vandenbulcke, M., Peeters, R., Van Hecke, P. and
mance in Alzheimer’s disease and mild cogni- Vandenberghe, R. (2005). Anterior temporal lateral-
tive impairment: A comparative review. Journal ity in primary progressive aphasia shifts to the right.
of Clinical and Experimental Neuropsychology, Annals of Neurology, 58 (3): 362–370.
30 (5): 501–556. Victoroff, J., Ross, G.W., Benson, D.F. et al. (1994).
Tang-Wai, D. and Mapstone, M. (2006). What are we Posterior cortical atrophy – Neuropathologic correla-
seeing? Is posterior cortical atrophy just Alzheimer tions. Archives of Neurology, 51 (3): 269–274.
disease? Neurology, 66 (3): 300–301. Warren, J.D., Warren, J.E., Fox, N.C. and Warrington,
Tang-Wai, D.F., Graff-Radford, N.R., Boeve, B.F. et al. E.K. (2003). Nothing to say, something to sing:
(2004). Clinical, genetic, and neuropathologic char- Primary progressive dynamic aphasia. Neurocase,
acteristics of posterior cortical atrophy. Neurology, 9 (2): 140–155.
63 (7): 1168–1174. Weintraub, S., Rubin, N.P. and Mesulam, M.M. (1990).
Tang-Wai, D.F., Josephs, K.A., Boeve, B.F. et al. (2003a). Primary progressive aphasia. Longitudinal course,
Coexistent Lewy body disease in a case of visual neuropsychological profile, and language features.
variant of Alzheimer’s disease. Journal of Neurology, Archives of Neurology, 47 (12): 1329–1335.
Neurosurgery and Psychiatry, 74 (3): 389. Westbury, C. and Bub, D. (1997). Primary progressive
Tang-Wai, D.F., Josephs, K.A., Boeve, B.F. et al. (2003b). aphasia: A review of 112 cases. Brain and Language,
Pathologically confirmed corticobasal degeneration 60 (3): 381–406.
presenting with visuospatial dysfunction. Neurology, Westbury, C., Bub, D. and Chertkow, H. (2002). Distinct
61 (8): 1134–1135. neurolinguistic symptom clusters in Alzheimer’s-type
Tobinick, E. (2008). Perispinal etanercept produces dementia and primary progressive aphasia. Brain and
rapid improvement in primary progressive apha- Cognition, 48 (2–3): 611–617.
sia: Identification of a novel, rapidly reversible Whitwell, J.L., Jack, C.R., Kantarci, K. et al. (2007).
TNF-mediated pathophysiologic mechanism. The Imaging correlates of posterior cortical atrophy.
Medscape Journal of Medicine, 10 (6): 135. Neurobiology of Aging, 28 (7): 1051–1061.
Primary progressive aphasia and posterior cortical atrophy 811

Wilson, S.M., Ogar, J.M., Laluz, V. et al. (2009). Automated crowding in a neurodegenerative disease population.
MRI-based classification of primary progressive Brain, 137 (Pt 12): 3284–3299.
­aphasia variants. Neuroimage, 47 (4): 1558–1567. Zahn, R., Buechert, M., Overmans, J. et al. (2005).
Yong, K.X., Shakespeare, T.J., Cash, D. et al. (2014a). (Con) Mapping of temporal and parietal cortex in progres-
text-specific effects of visual dysfunction on reading in sive nonfluent aphasia and Alzheimer’s disease using
posterior cortical atrophy. Cortex, 57: 92–106. chemical shift imaging, voxel-based morphometry and
Yong, K.X., Shakespeare, T.J., Cash, D. et al. (2014b). positron emission tomography. Psychiatry Research:
Prominent effects and neural correlates of visual Neuroimaging, 140 (2): 115–131.
70
The cerebellum and cognitive impairment

ELSDON STOREY AND EVELYN A. LINDSAY

synapses (Ito, 1993). LTD itself develops in response to


70.1 INTRODUCTION near-simultaneous Purkinje cell activation by parallel fibres
and by climbing fibres from the inferior olive. It may thus
The motor features of the cerebellar syndrome were crystal- be considered to be analogous to associative long-term
lized by the great English neurologist Sir Gordon Holmes potentiation in the hippocampus. In the influential Marr–
soon after the World War I. He did not ascribe any cognitive Albus–Ito model of cerebellar function, the climbing fibres
function to the cerebellum and, until the last decade or two, are regarded as carrying the ‘error signals’ responsible for
this view held a general sway. However, a confluence of evi- adaptive motor learning.
dence from new anatomical tracing techniques, functional While there is no universally accepted model whereby
neuroimaging experiments and careful clinical studies now LTD in the corticonuclear microcomplex might result in
points strongly to a significant role that the cerebellum plays motor learning at the animal level (Massaquoi and Topka,
in cognition, although the exact process(es) underlying this 2002), it has been assumed from its stereotyped modu-
contribution have not been fully elucidated. This chapter lar structure that the cerebellum performs the same type
aims to distil some of the important studies supporting a of basic computation on whatever inputs it receives and
non-motor role for the cerebellum. The reader interested informs all its various efferent targets of the results of such
in a detailed exposition is referred to The Cerebellum and computation, similarly. If different areas of the cerebellum
Cognition, edited by Jeremy Schmahmann (1997), while differ in function, they do so by virtue of their differing
several shorter reviews have also been published (Timmann inputs and outputs.
and Daum, 2007; Baillieux et al., 2008; Strick et al., 2009; With this in mind, the evidence for cerebellar input from
O’Halloran et al., 2012; Koziol et al., 2014) on this topic for and output to non-motor areas can be considered. Such evi-
scholarly reference. dence is both indirect and direct. The former consists of the
observation that, in primate phylogeny and especially in
hominids, the prefrontal portion of the frontal lobes and the
70.2 THE NEUROANATOMICAL BASIS lateral neocerebellum with its associated ventral macrogyric
region of the dentate nucleus have undergone massive parallel
OF CEREBELLAR COGNITIVE
selective enlargement out of proportion to other brain areas
FUNCTION (Matano, 2001; Macleod et al., 2003). More direct evidence
has awaited the development of anterograde and retrograde
The cerebellum, which contains 80% of the neurones in the trans-synaptic (viral) tracers (Middleton and Strick, 1997).
whole brain in 10% of its total mass (Azevedo et al., 2009), Such tracers are necessary because cerebral cortical input to
has a regular and relatively simple modular structure. The the cerebellum proceeds via cortico-pontine fibres synapsing
‘corticonuclear microcomplex’ is essentially a positive loop in the pontine nuclei, which in turn project to the deep cer-
through the deep cerebellar nuclei, modulated by a vari- ebellar nuclei and cerebellar cortex, while cerebellar output
able negative side loop through the cerebellar cortex (Ito, to the cortex proceeds via synaptic relays in the thalamus
1993). The degree of inhibitory influence of the cerebel- (Schmahmann and Pandya, 1997). Studies utilizing such trac-
lar cortical Purkinje cells on the deep cerebellar nuclei is ers in primates have shown that a series of anatomically dis-
governed by the development of long-term depression crete parallel cortico-ponto-cerebello-thalamo-cortical loops
(LTD) at the excitatory parallel fibre-Purkinje cell dendrite exist, with pre-frontal input predominantly to the ventral,

812
The cerebellum and cognitive impairment 813

macrogyric portion of the dentate nucleus and associated lat- nuclei bilaterally, whereas a more lateral region of the cer-
eral neocerebellar cortex and output via the ventrolateral and ebellar right hemisphere, lobule VI bordering on Crus I and
medial dorsal thalamic nuclei (Middleton and Strick, 2000, ventrocaudal parts of the dentate were activated by silent
2001). Motor and premotor cortices, in contrast, are recipro- verb generation. Their results confirmed that, as identified
cally connected to the phylogenetically older dorsolateral, in anatomical studies of monkeys, segregated regions of the
microgyric portion of the dentate nucleus (Middleton and human cerebellar cortex and corresponding areas of the
Strick, 2001). The analogy with separate, parallel corticobasal dentate nuclei are allocated separately to motor and non-
ganglionic circuits subservient to motor and non-motor func- motor functional domains. Operationalizing a contextual
tions, as conceptualized by Alexander et al. (1986) is readily semantic priming task, a study by Moberget et al. (2014)
drawn (Middleton and Strick, 2000). This circuitry has now showed an increased fMRI response in the right cerebel-
been imaged in humans, using diffusion tensor imaging lum during target word predictability conditions, suggest-
(DTI) to identify prefrontal-pontine white matter projections ing that the cerebellum is involved in predictive language
in the cortico-ponto-cerebellar system (Ramnani et al., 2006) processing, analogous to its hypothesized role in the motor
and functional connectivity magnetic resonance imaging domain to predict and control actions.
(MRI) to map cortico-cerebellar loops associated with higher The cerebellum also appears to be involved in executive
cognitive functions (Allen et al., 2005; Habas et al., 2009; functioning and working memory. Cognitive processing dur-
O’Reilly et al., 2009; Buckner et al., 2011; see Buckner, 2013 ing attempted solution of a pegboard puzzle task compared
for a review). with simple peg movement was studied using fMRI estima-
tion of dentate activation by Kim et al. (1994). The cogni-
tively demanding task produced increased dentate activation
­bilaterally, compared with the activation seen with simple peg
70.3 FUNCTIONAL NEUROIMAGING movement. In as much as this task tapped visuospatial work-
STUDIES ing memory, it is conceptually similar to the verbal working
memory fMRI study of Desmond et al. (1997), who found
A large number of functional neuroimaging studies over that delayed matching of letter strings, but not the control
the last 25 years have reported cerebellar activation during task, activated the right inferolateral cerebellar hemisphere.
tests of various cognitive domains, utilizing positron emis- Meta-analyses have confirmed different activation patterns
sion tomography (PET) and functional magnetic resonance depending upon the type of working memory task, with ver-
imaging (fMRI) (Cabeza and Nyberg, 2000; Stoodley, 2012). bal working memory located more prominently in the right
The typical design compares patterns of cerebellar activa- hemisphere and spatial working memory in the left hemi-
tion on a cognitive task with that on what is thought to be sphere (Stoodley and Schmahmann, 2009; Keren-Happuch
a relevant control ‘motor’ and/or perceptual task. Recent et al., 2014). For instance, a study by Marvel and Desmond
meta-analyses identified consistent cerebellar involvement (2010) showed separate clusters of cerebellar activity in dif-
in the domains of language, executive function and work- ferent regions of the dentate nucleus during encoding and
ing memory (Stoodley and Schmahmann, 2009; Keren- retrieval and an increase in cerebro-cerebellar activity as a
Happuch et al., 2014). This section includes a broadly result of cognitive load. The Wisconsin Card Sorting Test
representative sample of such studies across these cognitive (WCST), a complex task often used to assess aspects of execu-
domains, as well as memory and attention. tive functioning, has also been found to activate the lateral
The phylogenetic development of the neocerebellum cerebellum (Nagahama et al., 1996). In a study examining
in humans and evidence of reciprocal pathways between whether the cerebellum is engaged only by prefrontal infor-
the cerebellum and language-dominant frontal regions mation that requires action, Balsters et al. (2013) demon-
prompted interest in a cerebellar contribution to language strated that lateral hemispheric regions of the cerebellum
(see Murdoch, 2010; Mariën et al., 2014, for reviews). For receiving prefrontal projections were activated while pro-
example, Fullbright et al. (1999) asked subjects to read cessing both rules specifying action and higher order rules
silently. Their fMRI study showed that semantic compari- independent of action.
son and judgement of non-word rhyming activated pos- The role of the cerebellum in learning and memory has
terolateral (and especially right posterolateral) cerebellar been evidenced mainly in studies of procedural memory
cortex, compared with the control conditions of line ori- and associative learning (O’Halloran et al., 2012). An aspect
entation judgement, upper versus lower case print judge- of episodic memory was addressed by Andreasen et al.
ment or judgement of real word rhyming. Xiang et al. (2003) (1999) in a ‘pure thought’ PET experiment. Subjects were
confirmed the role of the right posterolateral cerebellum in asked to recall a personal memory silently. The right lateral
semantic discrimination divorced from articulation. Such cerebellum was activated in addition to several relevant
studies have led to the concept of the ‘lateralized linguistic supratentorial areas. Fliessbach et al. (2007) found that
cerebellum’, reviewed by Mariën et al. (2001). Thürling et al. words remembered in a subsequent memory test following
(2011) used 7T MRI with a matrix block design to demon- incidental encoding evoked stronger fMRI responses in the
strate that motor speech (reading aloud) activated paraver- superior posterior region of the right cerebellum than words
mal areas of lobule VI and dorsorostral parts of the dentate that had been forgotten.
814 Dementia

A further area of recent active study has been the role(s) results. Some tests are untimed and do not require motoric
of the cerebellum in attention. Allen et al. (1997) showed an or visual speed or accuracy. In the executive domain, these
anatomical double dissociation in cerebellar regional acti- include the WCST, Raven’s Progressive Matrices (RPM) and
vation; a visual attention task activated the lateral cerebel- the Zoo Map subtest (of planning) from the Behavioural
lum, while the motor task activated more medial regions. Assessment of the Dysexecutive Syndrome (BADS) battery.
Attention is, however, a multifaceted concept, placing a Most measures of verbal anterograde episodic memory also
variety of different demands on the subject. Le et al. (1998) fall into this category, provided that dysarthria is not so
showed that shifting attention between stimulus dimen- severe as to compromise intelligibility. Other tests include
sions (colour/shape) activated the right lateral cerebellum, a timed internal control condition involving similar scan-
while sustained attention did not. However, Bischoff-Grethe ning and output to the test condition. The effects of visual,
et al. (2002) have demonstrated that the lateral cerebellum’s verbal and motor dysfunction can therefore be subtracted
role in this paradigm may actually be a reassignment of a out. Such tests within the executive function cognitive
given motor response to the new stimulus dimension rather domain would include Trails B (versus A) and the Stroop
than in shifting attention per se, a view supported more test. Some other tests requiring verbal output are timed, but
recently by Haarmeier and Thier (2007). empirical observation suggests that the speed of articula-
Theory-of-mind (TOM) refers to inferential thinking tion is not the time-limiting factor. The Controlled Oral
about the mental and emotional states of others (Premack Word Association test, first called the Verbal Associative
and Woodruff, 1978) and involves the ability to recognize Fluency Test (COWAT, FAS) appears to fall into this cat-
and identify emotions, understand implicit language and egory (Storey et al., 1999).
interpret social situations. Functional imaging has impli- An indirect way of exploring the possibility that motor
cated the cerebellum in prefrontal and limbic cortical net- dysfunction might secondarily affect performance on
works involved in TOM (or the broader concept of social cognitive tests is to study patients with Friedreich’s ataxia
cognition) (Calarge et al., 2003; Van Overwalle et al., 2014). (FA), whose motoric deficit is partly consequent upon
It is clear that the weight of evidence from functional disruption of afferent cerebellar input rather than direct
neuroimaging studies in favour of lateral cerebellar contri- disruption of the cerebellar microcorticonuclear units,
butions to cognition is strong, as illustrated by the selected although the deep cerebellar nuclei are also affected
studies cited above. However, it is also clear that the exact (Koeppen, 2002). While FA patients display impairments
role(s) of the lateral cerebellum in complex aspects of cogni- in a number of domains (Wollmann et al., 2002), they have
tion, such as language and attentional shifting, still requires been shown to be less impaired on verbal working mem-
considerable clarification. ory and visuospatial reasoning tasks than patients with
assorted other cerebellar degenerations (Botez-Marquard
and Botez, 1997). Indeed, White et al. (2000) demon-
strated slowed processing speed and decreased cognitive
70.4 DIFFICULTIES IN STUDYING inhibition on the Stroop test but preserved planning, ver-
COGNITIVE FUNCTION IN bal fluency and WCST performance.
The potential confounding factor of motor deficit has
SUBJECTS WITH CEREBELLAR
recently been explicitly addressed by Timmann et al.
DYSFUNCTION (2002), who correctly pointed out that even ataxic patients
who cope well with the motor demands of a test may do so
The results of functional neuroimaging studies in normal at the cost of attentional resources not required by control
adults notwithstanding, the practising clinician is primarily patients. These authors reported that subjects with pure
concerned with the potential impact of cerebellar disease or cerebellar cortical atrophies (including spinocerebellar
dysfunction on cognition. Unfortunately, several confound- ataxia type 6 [SCA6], spinocerebellar ataxia type 8 [SCA8]
ing factors must be borne in mind when interpreting such and idiopathic cerebellar cortical atrophy) demonstrated
studies. The first problem is that performance on the neu- impairments of visual association learning independent of
ropsychological tests utilized may be adversely affected by the separately manipulated motor output requirements of
requirements for rapid verbal output, motor accuracy and the task.
speed or by dependence on rapid visual scanning. A corre- The second problem affecting patient studies is that of
lation between test results and ataxia severity will not nec- choice of the patient type. The ideal patient group for such
essarily indicate that such requirements are confounding a study would have widespread/diffuse involvement of cer-
the results; however, motor and cognitive function would ebellar corticonuclear microcomplex units with complete
be expected to decline in diffuse cerebellar degeneration in sparing of all other central nervous system (CNS) struc-
parallel, whether the observed cognitive dysfunction was tures. Unfortunately, such patients probably do not exist,
secondary to the cerebellar disorder or was merely second- at least to the extent of satisfying those sceptical of the idea
ary to the resultant motor impairment. of cerebellar contributions to cognition. Even relatively
Several types of neuropsychological tasks would be ‘pure’ cerebellar ataxias, such a SCA6 and SCA8 often
expected a priori to limit the potential for confounding of have mild non-cerebellar features (e.g. pyramidal signs).
The cerebellum and cognitive impairment 815

Other degenerative cerebellar diseases studied in this Auditory Serial Addition Test (PASAT) and the Stroop
context, such as spinocerebellar ataxias (SCAs) 1, 2 and colour–word interference test. These abnormalities were
3, are even more problematic, as they are known to mani- not accompanied by a clinically apparent ‘frontal’ neuro-
fest extracerebellar involvement of structures potentially psychiatric syndrome. Block design was also affected, the
or actually contributing to cognition, such as the cere- deficit of which the authors likened to a mild parietal syn-
bral cortex, the striatum and substantia nigra (Koeppen, drome, although problems with block design can also be
2002). On the other hand, focal cerebellar lesions, such as a feature of executive dysfunction. Interestingly, infarcts
infarcts and resected tumours, may be ‘pure’ but there is in any of the three cerebellar arterial territories produced
no guarantee that cerebellar region(s) potentially impor- similar deficits, although the numbers in each group were
tant for cognition will be involved, unless a range of small. This is in contrast to the findings of Schmahmann
lesions in different locations is studied. Pooled results may and Sherman (1998) and Exner et al. (2004), the latter
then conceal regional differences in cognitive contribu- reporting that posterior inferior, but not superior, cerebel-
tion unless sufficient subjects are available in each group lar artery infarcts resulted in cognitive deficits. As might
for comparison. A further potential limitation, of which be expected with infarcts, the deficits reported by Neau
to be aware, concerns functional changes due to neural et al. (2000) tended to have improved at the 1 year assess-
plasticity in patients with chronic focal lesions (Timmann ment. The authors plausibly suggest that this improve-
et al., 2009). While controlling these potential confounds ment may have been partly responsible for the failure of
is difficult, such studies do perhaps provide the most con- Beldarrain et al. (1997) to demonstrate consistent defi-
vincing clinical evidence, as outlined below. cits in a patient population including subjects with old
cerebellar infarcts, a finding since confirmed by Richter
et al. (2007a). Hokkanen et al. (2006) reported 26 Finnish
subjects with cerebellar lesions, assessed acutely before
and after 3 months. Those with left cerebellar lesions were
70.5 ALTERED COGNITION IN ADULTS
slow on a visuospatial task, whereas those with right-sided
WITH FOCAL CEREBELLAR
damage showed impairments of verbal episodic and work-
LESIONS ing memory. These deficits improved over the course of
time, such that all but one patient eventually returned to
In a seminal report, Schmahmann and Sherman (1998) work and 77 % had done so after 3 months. The reasons for
described the features of an entity they named the ‘cer- this discrepancy in long-term outcome from the Swedish
ebellar cognitive affective syndrome’ in 20 adults with study of Malm et al. (1998) are unclear.
isolated cerebellar lesions: strokes, resected tumours Using the lesion-symptom correlation method with 39
not treated with radiotherapy and post-viral cerebellitis. patients, following an acute/subacute cerebellar infarct, a
Posterior (and roughly equivalent to lateral neocerebellar) recent study demonstrated that a focal but quite large lesion
lobe lesions produced executive dysfunction with impair- to the cerebellum is neither necessary nor sufficient condi-
ment in planning, set shifting, verbal fluency by semantic tion for motor impairment and provided clinical support
category and abstract reasoning. There was also impair- for the notion that there are cerebellar regions devoted to
ment of spatial cognition including visual organization non-motor function (Schmahmann et al., 2009). Following
and visual memory. Lesions of the posterior vermis tended assessment with a modified International Cooperative
to produce personality change with blunting and disinhi- Ataxia Rating Scale (MICARS), patients whose stroke
bition, while anterior lobe lesions resulted only in motor avoided the anterior lobes (lobules I–V) and was confined
deficit. Similar findings were reported by Malm et al. to parts of lobules VII–X (in particular lobule VII), demon-
(1998) in 24 consecutive young adult patients with cere- strated no signs of the cerebellar motor syndrome (i.e. gait
bellar infarcts who were compared with 14 age-matched ataxia, appendicular ataxia or dysarthria). Minimal motor
controls. These patients performed less on working mem- impairment was identified if lobule VI was involved. These
ory and cognitive flexibility tasks, while intelligence (The findings were consistent with a later review of functional
Wechsler Adult Intelligence Scale – Revised [WAIS-R] imaging and clinical data, which concluded that cogni-
scores) and episodic memory were seen to be unaffected. tive impairment may occur with lesions impacting lobules
Interestingly, while most made a good motor recovery, VI, VII (principally Crus I, Crus II) and lobule VIIIB and
only half of them returned to work. The broad conclu- suggesting disruption of ‘cognitive’ cerebro-cerebellar
sions of these studies have since been confirmed by Neau connectivity with the association cortices (Stoodley and
et al. (2000), who reported the neuropsychological profile Schmahmann, 2010).
of 15 consecutive patients with recent, isolated cerebellar The specific role of the right lateral cerebellum in lan-
infarcts compared with 15 demographically matched con- guage, outlined in the section on functional neuroimag-
trols. A further assessment was undertaken 1 year after ing above, was studied in control subjects and subjects
the infarcts. These authors demonstrated impairments with left lateral or right lateral cerebellar infarcts (Gebhart
on a range of tasks tapping aspects of executive function- et al., 2002). Those with right lateral cerebellar damage
ing, such as verbal fluency, working memory on the Paced were impaired only on antonym generation and not on
816 Dementia

semantic (category) noun fluency or verb selection. This in intra- or inter-dimensional non-spatial attentional shift-
argues in favour of a cerebellar role in producing word asso- ing, again failing to confirm earlier functional neuroim-
ciations and not just in ‘imaging’ verb action to a noun cue. aging studies in a clinical population. Moreover, Ravizza
Helmuth et al. (1997) also failed to demonstrate impaired and Ivry (2001) found that, while patient groups with
noun-triggered verb generation in subjects with right cer- Parkinson’s disease (PD) and with cerebellar lesions each
ebellar lesions, opposing the findings from earlier func- showed impairment of attentional shifting, reducing the
tional neuroimaging studies. In contrast, verbal fluency was motor demands of the task, improved the performance of
the only impaired function in subjects with chronic focal only the cerebellar patients. Neglect may be regarded as a
cerebellar lesions, being most evident in those with right- disorder of lateralized spatial attention. Conflicting results
sided lesions (Richter et al., 2007b). Exploring the role of have been obtained in line bisection tasks in subjects with
the cerebellum in verbal fluency further, Schweizer et al. lateralized cerebellar lesions (Daini et al., 2008; Kim et al.,
(2010) compared the performance of patients with chronic 2008) with the minority in whom neglect was demonstrable
unilateral cerebellar lesions (right versus left) and healthy not always showing this ipsilateral to the lesion, as might
controls on phonemic (letter) and semantic (category) flu- be expected a priori (Kim et al., 2008). The question of a
ency tasks. Performance on the phonemic fluency task was cerebellar contribution to various forms of attention has
more impaired for the right cerebellar lesion group than for generated conflicting results and, while this remains to be
either the left cerebellar lesion group or controls and was settled conclusively, the evidence so far is not convincing
characterized by executive and attention deficits normally when potential oculomotor/motor confounds are removed
expected with left prefrontal lesions. Semantic fluency per- (Haarmeier and Thier, 2007).
formance was worse relative only to controls for the right The role of the cerebellum in sequencing – an aspect of
cerebellar lesion group. executive functioning – has also been highlighted (Leggio
The potential role of the cerebellum in more complex et al., 2008). Analogous with its role in sensory and motor
rather than simpler tasks has recently been extended to sequence detection and production, cerebellar damage also
reading: Ben-Yehudah and Fiez (2008) showed that focal disturbs verbal (sentence) and spatial sequencing (Leggio
cerebellar damage impaired a visual rhyme judgement task et al., 2008). Furthermore, there is an effect of lesion lateral-
for irregularly spelt words, but not for phonetically regular ity with right-sided lesions impairing verbal and left-sided
words, and impaired working memory only for non-words lesions impairing spatial sequencing.
rather than for real words. Reading fluency and comprehen- The ecological validity of executive function impairment
sion were unaffected. identified on neuropsychological tests following cerebellar
Verbal working memory – the ability to manipulate lesions was addressed by Manes et al. (2009). Eleven patients
phonological data in consciousness (e.g. reversing digit with focal cerebellar vascular lesions variously affecting pos-
strings) – is one aspect of executive function often found terior inferior cerebellar artery, superior cerebellar artery
to be mildly affected in subjects with cerebellar lesions and mixed territories were assessed with a neuropsycholog-
(Bellebaum and Daum, 2007). A parsimonious explanation ical test battery, with significant differences from controls
would be that the cerebellum has a role in subvocal articu- found on tests sensitive to frontal lobe function (including
latory rehearsal. However, Ravizza et al. (2006) provide the WCST and the flexibility scale of The Test of Attentional
evidence that the cerebellum’s role is actually in initial pho- Performance). Ecological validity – the degree to which neu-
nological encoding and/or in strengthening memory traces. ropsychological performance corresponds with real world
Episodic memory has generally been reported as relatively behaviour – was measured using the Multiple Errands Test
preserved with focal cerebellar lesions, but a relatively Hospital Version (MET-HV) with scores reflecting difficul-
recent study of patients after removal of cerebellar tumours ties with timing, task planning and organization in the cer-
revealed episodic memory deficits in most, although there ebellar infarct patients. Roldan Gerschcovich et al. (2011),
was no clear correlation of modality with lesion laterality presenting a case study of a patient with extensive, bilateral
(de Ribaupierre et al., 2008). cerebellar infarction without supratentorial involvement,
Various attentional processes have also been studied noted mild deficits in verbal fluency (particularly to phone-
in subjects with cerebellar lesions with varying results. mic cues) and working memory, moderate deficits in story
Orienting visuospatial attention has been found to be recall, conceptualization, executive function and cogni-
markedly delayed in such subjects, independent of motor tive flexibility and impairment on tests of decision making
responses (Townsend et al., 1999), while the speed of visual (e.g. ABCD Iowa Gambling Task [IGT], Game of Dice Task
attention is also slowed, independent of eye movements [GDT], Stimulus Reward Learning, Reversal and Extinction
(Schweizer et al., 2007). Gottwald et al. (2003) compared Task [SRLRET]). This patient also demonstrated significant
16 subjects with cerebellar tumours or haematomas to 11 difficulties on TOM tasks. A later report of the study con-
matched control subjects and to demographically adjusted ducted by Manes et al. (2009) commented that, in contrast
results from normative studies and detected deficits in to Roldan Gerschcovich et al. (2011), their 11 patients with
divided attention and working memory, but not in sus- focal cerebellar vascular lesions performed no differently to
tained directed attention. However, Helmuth et al. (1997) controls on a TOM test of their ability to detect social faux
did not demonstrate deficits in spatial attention shifting or pas (Roca et al. 2013).
The cerebellum and cognitive impairment 817

Studies in children with resected cerebellar tumours Kish et al. (1994) studied the performance of 43 sub-
have produced similar results to those in adults (Levisohn jects with undefined dominant cerebellar degenerations of
et al., 2000). varying ataxia severity on a number of neuropsychological
measures. Impaired performance on the WCST correlated
with ataxia severity; none of those with mild ataxia showed
70.6 EVIDENCE FOR COGNITIVE deficits while all those with severe ataxia were impaired.
Some of the latter subjects also showed evidence of mild
IMPAIRMENT IN DEGENERATIVE
generalized cognitive deficit. The authors concluded that
CEREBELLAR DISEASE the dominant ataxias are not homogeneous with respect to
cognitive involvement and indeed the topography of extra-
Evidence for cognitive impairment in degenerative cerebel- cerebral neuropathological involvement does vary consid-
lar disease comes from both neurophysiological and clinical erably between these disorders (Rüb et al., 2013). Trojano
neuropsychological studies. The best known neurophysi- et al. (1998) also studied a mixed population of subjects
ological method of studying cognition involves measuring with dominant ataxias of varying severity, although 15 out
the latency of the P300 event-related cerebral potential. This of their 22 subjects had SCA2. They attempted to avoid con-
occurs with conscious registration of an unusual stimulus, founding effects of motor impairment on performance by
such as an occasional (oddball) high-pitched tone inserted restricting their investigation to tests with untimed verbal
into a series of usual stimuli, such as many low-pitched response modes. The domains examined were predomi-
tones. It may be regarded as a neurophysiological measure nantly verbal memory and non-verbal reasoning (Raven’s
of selective attention and processing speed and is delayed Progressive Matrices). None of the four genetically con-
in various forms of dementia (Goodin, 2003). Tachibana firmed but asymptomatic subjects performed poorly on any
et al. (1999) measured the P300 during a visual discrimi- of the tests, while most of the symptomatic subjects showed
nation task and found it to be delayed in 15 subjects with impairment on at least one measure, most commonly, the
idiopathic late-onset cerebellar ataxia, when compared with progressive matrices. Some showed widespread deficits on
10 controls. Single-photon emission computed tomogra- a large scale but there was no correlation with ataxia sever-
phy (SPECT) scanning in the ataxic subjects showed corre- ity. This might have reflected the small sample size and the
lated cerebellar/frontal cortex hypoperfusion. Tanaka et al. contaminating effects of inclusion of several SCA subtypes.
(2003) did not find any differences in P300 timing or scalp Several studies have assessed subjects with different SCA
distribution to the classical auditory ‘oddball’ paradigm in types, singe or placed separately from other types, thereby
13 subjects with cerebellar cortical atrophy when compared avoiding this particular confounding factor. Maruff et al.
to 13 demographically matched controls. However, P300 (1996) compared six patients with SCA3 (Machado–Joseph
latency was prolonged and its peak attenuated frontally, in disease [MJD]) to 15 matched controls using a touchscreen
the ‘No-Go’ condition of the ‘Go/No-Go’ paradigm. The testing system: the Cambridge Neuropsychological Test
authors suggested that the cerebellum contributes to the Automated Battery (CANTAB). Learning and visual mem-
frontal inhibitory system. ory were unaffected, but SCA3 subjects displayed impaired
The neuropsychological consequences of degenerative visual information processing speed on high-demand tasks
cerebellar disease have been studied extensively. In an and impaired attentional switching between visual stimu-
early study, Grafman et al. (1992) compared nine patients lus dimensions. Executive dysfunction in SCA3 was con-
with ‘pure cortical cerebellar atrophy’ with 12 controls. firmed by Zawacki et al. (2002). Braga-Neto et al. (2012)
There were no differences on paired associate learning, compared 38 molecularly and clinically proven patients
verbal fluency or procedural (skill) learning. However, the with SCA3 to 31 controls on measures of neuropsychologi-
ataxic subjects performed significantly worse on the Tower cal function, ataxia rating scales, depression and anxiety.
of Hanoi planning task (ToH), which is conceptually simi- Patients with SCA3 reported significantly more symptoms
lar to the Tower of London (TOL) test. Botez-Marquard of depression and anxiety than controls, but performances
and Botez (1997) showed that cerebellar degenerations on tests of visuospatial abilities and executive function were
resulted in impaired executive functioning, visuospatial significantly impaired independent of psychiatric scores
organization, working memory and spontaneous retrieval and demographic factors. Results failed to confirm a visual
of episodic memories. Cerebellar strokes produced simi- memory deficit in this patient group. In a SCA3 sample
lar patterns of impairment, but these cognitive deficits group of 32 patients, Lopes et al. (2013) found mild impair-
resolved in 2–5 months. In both types of subjects, SPECT ments in episodic and working memory and executive func-
scanning showed reverse cerebellar/frontoparietal dias- tion relative to demographically matched controls, deficits
chisis. Drepper et al. (1999) studied nine subjects with that were not associated with CAG expansion size, disease
‘isolated’ cerebellar degeneration and 10 controls. The duration or ataxia severity. Neuroimaging with 3T MRI and
ataxic subjects showed a significant impairment in novel DTI revealed reduced grey matter volume in the temporal,
associative learning (colours with numbers) that did not parietal and frontal lobes as well as the cerebellum, which
correlate with either simple reaction time or visual scan- was associated with episodic memory impairment and
ning time. executive dysfunction in the patient group. There was also
818 Dementia

a positive correlation between fractional anisotropy values anxiety or positive symptoms. The SCA2 group of patients
in the brain stem and forwards digit span. Progressive cog- performed worse on cognitive tests than the SCA1 group.
nitive dysfunction, notably in verbal and figural memory, Memory was not impaired and only three patients out of
was also observed over 3½ years by Roescke et al. (2013) in a total of 42 at baseline scored below 24/30 on the Mini-
a small group of SCA3 patients with a range of CAG repeat Mental State Examination (MMSE), suggesting dementia
expansion sizes. While motor deterioration was observed, was not a feature of these SCA types in the moderate stage
the memory tests were independent of motor impairment of the disease. A 2-year follow-up assessment of small sub-
and cognitive changes could not be attributed to progressive sets of both patient groups (9 SCA1 and 11 SCA2) was car-
cerebellar ataxia. ried out and revealed that only performances on attentional
SCA2 may be associated with cortical atrophy and clini- matrices in the SCA2 group had deteriorated significantly.
cally with obvious dementia in some pedigrees (Dürr et al., There was a significant increase in motor impairment and
1995; Geschwind et al., 1997). Although it is not ideal for the a trend towards worsening of functional abilities in this
purpose it has been studied by several groups. Gambardella group, compared to the SCA1 group. Although findings
et al. (1998) and Storey et al. (1999) demonstrated impair- were limited to small sample groups for follow-up and the
ments of executive function on the WCST and the Stroop short follow-up duration was to be taken into account, the
test. Bürk et al. (1999) found that 25% of their subjects were researchers proposed their study as supporting dissociation
demented, while the others showed impairments of execu- between cognitive and motor impairment in SCA2.
tive function and verbal memory that were independent of Social cognition was investigated by Sokolovsky et al.
the severity of motor impairment. In contrast, Le Pira et al. (2010) in common types of SCA, which included SCAs 1, 2
(2002) reported that the impairments of executive function, and 7 and compared with previously investigated cognitive
attention and verbal memory in their SCA2 patients were and social cognitive characteristics in the SCA3 and SCA6,
partly related to ataxia severity. using an identical assessment battery (Garrard et al., 2008).
The pattern of neuropsychological impairment in SCA1 SCA2 patients demonstrated the most cognitive impairment,
patients compared to matched controls was reported by notably in attention, non-verbal intellectual abilities and
Bürk et al. (2001). Evidence of executive and verbal memory executive function and at a degree similar to SCA3 patient
dysfunction was found, while visuospatial memory and group. SCA1 patients had relatively intact cognitive profiles
attention were unaffected. The deficits did not correlate with and the SCA7 group patients had selective mild deficits in
CAG repeat length or with disease duration, which may be executive function. On social cognition tests, SCA2 and
regarded as surrogate markers of disease severity in differ- SCA7 patients demonstrated impairment on a test of social
ent dimensions. attribution (providing a one-word description of a character’s
While the studies on SCAs 1, 2 and 3 cited above paint a feeling in an emotional situation, e.g. happiness, sadness,
fairly consistent picture of executive dysfunction and verbal disgust, fear, anger, embarrassment). Only one patient with
memory impairment, the effects of differences in the pattern SCA1 was impaired on a TOM test, a deficit which had been
and extent of extracerebellar neuropathological involve- previously observed in SCA3 and SCA6 patients (Garrard
ment, if any, can only be addressed adequately by direct et al., 2008). The results of these studies again bring into focus
comparison. This was undertaken by Bürk et al. (2003), the need to consider the wide range of non-motor deficits
who found that executive dysfunction was more marked that may afflict patients with cerebellar disease and the addi-
in SCA1 than in SCAs 2 or 3, while mild verbal memory tional impact beyond motor disability on daily functioning.
impairment was present in all of them. On the basis of these Perhaps the best way of clarifying the relative contribu-
findings, the authors suggested that the cognitive deficits tions of cerebellar and extracerebellar pathology to cog-
were likely to result from disruption of cerebro-cerebellar nitive dysfunction in the SCAs is to study those disorders
circuitry at the pontine rather than at the cerebellar level. characterized by relatively pure cerebellar involvement, such
Given that SCA2 causes more severe pontine atrophy than as SCA6 and to a lesser extent, SCA8. Although McMurtray
SCA1, the reason for this conclusion is unclear. A prob- et al. (2006) found that SCA6 patients, in contrast to those
lem with this study is the lack of control for ataxia sever- with other SCA types, tended not to show memory deficits,
ity. Klinke et al. (2010) also found poorer performances by Suenaga et al. (2008) reported significant impairments of
SCA1 patients on cognitive tests when compared to SCA2 verbal fluency and immediate visual memory in SCA6, inde-
and 3, with attentional and executive dysfunction predomi- pendent of motor dysfunction. In a group of 27 patients with
nantly present in these three types. Semantic and episodic genetically confirmed SCA6, Cooper et al. (2010) reported
memory were essentially preserved. Cognitive domains of an absence of memory decline and identified significant
deficit were partly correlated with ataxia severity and fine impairment in executive function on tests of cognitive flex-
motor coordination. Fancellu et al. (2013) examined the ibility, response inhibition, verbal reasoning and abstrac-
progression of cognitive, psychiatric, motor and functional tion. A later study by this research group (Cooper et al.,
abilities of patients with SCA1(9) and SCA2(11). Baseline 2012) used high resolution structural MRI to demonstrate
cognitive measures showed mild deficits in both groups a relationship between verbal working memory based on
in attention and executive function, with the most notable phonological storage and retrieval and grey matter density
psychiatric feature being apathy rather than depression, in bilateral lobules VI and Crus I as well as in the inferior
The cerebellum and cognitive impairment 819

lobe (bilateral lobules VIIIA and VIIIB and right lobule IX) Balsters, J.H., Whelan, C.D., Robertson, I.H. and
of 15 patients with SCA6. Two groups have reported a dysex- Ramnani, N. (2013). Cerebellum and cognition:
ecutive syndrome (DES) in SCA8 (Lilja et al., 2005; Torrens Evidence for encoding of higher-order rules. Cerebral
et al., 2008) with the first group also demonstrating deficits Cortex, 23: 1433–1443.
in attention and information processing. Beldarrain, M.G., Garcia-Monco, J.C., Quintana, J.M.
et al. (1997). Diaschisis and neuropsychological perfor-
mance after cerebellar stroke. European Neurology,
37: 82–89.
70.7 CONCLUSIONS Bellebaum, C. and Daum, I. (2007). Cerebellar involvement
in executive control. The Cerebellum, 6 (3): 184–192.
There is converging evidence from neuroanatomical, neu- Ben-Yehudah, G. and Fiez, J.A. (2008). Impact of
roimaging, neurophysiological and neuropsychological cerebellar lesions on reading and phonological
patient studies that the lateral cerebellum is involved in processing. Annals of the New York Academy of
network(s) subservient to a range of cognitive functions, Sciences, 1145: 260–274.
including aspects of language and executive functioning. Bischoff-Grethe, A., Ivry, R.B. and Grafton, S.T. (2002).
The exact nature of the cerebellar contributions to cogni- Cerebellar involvement in response reassignment
tion and especially to attention, are still being debated rather than attention. Journal of Neuroscience, 22:
and ­elucidated. Nevertheless, the broad picture is suffi- 546–553.
ciently well established that clinicians should consider the Botez-Marquard, T. and Botez, M.I. (1997).
possibility of cognitive dysfunction in patients with overt Olivopontocerebellar atrophy and Friedreich’s ataxia:
cerebellar disorders. In this context, executive dysfunction Neuropsychological consequences of bilateral versus
is both the most likely to be overlooked in the structured unilateral cerebellar lesions. International Review of
clinical setting and potentially, the most disruptive to the Neurobiology, 41: 387–410.
patients’ ability to compensate for impaired motor func- Braga-Neto, P., Pedroso, J.L., Alessi, H. et al. (2012).
tioning. Clinicians should also bear in mind that obvious Cerebellar cognitive affective syndrome in
cognitive dysfunction may result from cerebellar disease, at Machado Joseph disease: Core clinical features. The
the least from degenerative disorders and acute focal lesions, Cerebellum, 11: 549–556.
and such dysfunction does not necessarily signify that a sec- Buckner, R.L. (2013). The cerebellum and cognitive func-
ond supratentorial disorder has been overlooked. tion: 25 Years of insight from anatomy and neuroimag-
ing. Neuron, 80: 807–815.
Buckner, R.L., Krienen, F.M., Castellanos, A. et al. (2011).
REFERENCES The organization of the human cerebellum by instrinsic
functional connectivity. Journal of Neurophysiology,
Alexander, G.E., DeLong, P.L. and Strick, P.L. (1986). 106: 2322–2345.
Parallel organization of functionally segregated circuits Bürk, K., Bösch, S., Globas, C. et al. (2001). Executive
linking basal ganglia and cortex. Annual Review of dysfunction in spinocerebellar ataxia type 1. European
Neuroscience, 9: 357–381. Neurology, 46: 43–48.
Allen, G., Buxton, R.B., Wong, E.C. et al. (1997). Bürk, K., Globas, C., Bösch, S. et al. (1999). Cognitive
Attentional activation of the cerebellum independent deficits in spinocerebellar ataxia 2. Brain: A Journal of
of motor involvement. Science, 275: 1940–1943. Neurology, 122 (Pt 4): 769–777.
Allen, G., McColl, R., Barnard, H. et al. (2005). Magnetic Bürk, K., Globas, C., Bösch, S. et al. (2003). Cognitive defi-
resonance imaging of cerebellar-prefrontal and cer- cits in spinocerebellar ataxia type 1, 2, and 3. Journal
ebellar-parietal functional connectivity. NeuroImage, of Neurology, 250: 207–211.
28: 39–48. Cabeza, R. and Nyberg, L. (2000). Imaging cognition II: An
Andreasen, N.C., O’Leary, D.S., Paradiso, S. et al. empirical review of 275 PET and fMRI studies. Journal
(1999). The cerebellum plays a role in conscious of Cognitive Neuroscience, 12: 1–47.
episodic memory retrieval. Human Brain Mapping, Calarge, C., Andreasen, N.C. and O’Leary, D.S. (2003).
8: 226–234. Visualising how one brain understands another: A
Azevedo, F.A.C., Carvalho, L.R.B., Grinberg, L.T. et al. PET study of theory of mind. The American Journal of
(2009). Equal numbers of neuronal and nonneuronal Psychiatry, 160 (11): 1954–1964.
cells make the human brain an isometrically scaled-up Cooper, F.E., Grube, M., Elsegood, K.J. et al. (2010). The
primate brain. The Journal of Comparative Neurology, contribution of the cerebellum to cognition in spi-
513: 532–541. nocerebellar ataxia type 6. Behavioural Neurology,
Baillieux, H., De Smet, H.J., Paquier, P.F. et al. (2008). 23: 3–15.
Cerebellar neurocognition: Insights into the bottom Cooper, F.E., Grube, M., Von Kriegstein, K. et al. (2012).
of the brain. Clinical Neurology and Neurosurgery, Distinct critical subregions for components of verbal
110: 763–773. working memory. Neuropsychologia, 50: 189–197.
820 Dementia

Daini, R., Arduino, L.S., Di Menza, D. et al. (2008). Line Gottwald, B., Mihajlovic, Z., Wilde, B. and Mehdorn, H.M.
bisection and cerebellar damage. Cognitive and (2003). Does the cerebellum contribute to specific
Behavioural Neurology, 21: 214–220. aspects of attention? Neuropsychologia, 41: 1452–1460.
de Ribaupierre, S., Ryser, C., Villemure, J.G. et al. (2008). Grafman, J., Litvan, I., Massaquoi, S. et al. (1992).
Cerebellar lesions: Is there a lateralisation effect on Cognitive planning deficit in patients with cerebellar
memory deficits? Acta Neurochirurgica, 150: 545–550. atrophy. Neurology, 42: 1493–1496.
Desmond, J.E., Gabrieli, J., Wagner, A. et al. (1997). Haarmeier, T. and Thier, P. (2007). The attentive cerebel-
Lobular patterns of cerebellar activation in ver- lum – Myth or reality? The Cerebellum, 6: 177–183.
bal working-memory and fingertapping tasks as Habas, C., Kamdar, N., Nguyen, D. et al. (2009). Distinct
revealed by functional MRI. Journal of Neuroscience, cerebellar connections to intrinsic connectivity net-
17: 9675–9685. works. The Journal of Neuroscience,
Drepper, J., Timmann, D., Kolb, F.P. and Diener, H.C. 29: 8586–8594.
(1999). Non-motor associative learning in patients Helmuth, L.L., Ivry, R.B. and Shimizu, N. (1997). Preserved
with isolated degenerative cerebellar disease. Brain: performance by cerebellar patients on tests of word
A Journal of Neurology, 122: 87–97. generation, discrimination learning, and attention.
Dürr, A., Smadja, D., Cancel, G. et al. (1995). Autosomal Learning and Memory, 3: 456–474.
dominant cerebellar ataxia type I in Martinique (French Hokkanen, L.S., Kauranen, V., Roine, R.O. et al. (2006).
West Indies): Clinical and neuropathological analysis of Subtle cognitive deficits after cerebellar infarcts.
53 patients from three unrelated SCA2 families. Brain: European Journal of Neurology, 13: 161–170.
A Journal of Neurology, 118: 1573–1581. Ito, M. (1993). Movement and thought: Identical con-
Exner, C., Weniger, G. and Irle, E. (2004). Cerebellar trol mechanisms by the cerebellum. Trends in
lesions in the PICA but not SCA territory impair cogni- Neuroscience, 16: 448–450.
tion. Neurology, 63: 32–35. Keren-Happuch, E., Chen, S.A., Ho, M.R. and Desmond, J.E.
Fancellu, R., Paridi, D., Tomasello, C. et al. (2013). (2014). A meta-analysis of cerebellar contributions to
Longitudinal study of cognitive and psychiatric func- higher cognition from PET and fMRI studies. Human
tions in spinocerebellar ataxia types 1 and 2. Journal Brain Mapping, 35: 593–615.
of Neurology, 260 (12): 3134–3143. Kim, E.J., Choi, K.D., Han, M.K. et al. (2008). Hemispatial
Fliessbach, K., Trautner, P., Quesada, C.M. et al. (2007). neglect in cerebellar stroke. Journal of the
Cerebellar contributions to episodic memory encoding Neurological Sciences, 275: 133–138.
as revealed by fMRI. Neuroimage, 35: 1330–1337. Kim, S., Ugurbil, K. and Strick, P. (1994). Activation of a
Fullbright, R.K., Jenner, A.R., Mencl, W.E. et al. (1999). cerebellar output nucleus during cognitive processing.
The cerebellum’s role in reading: A functional MR Science, 265: 949–951.
imaging study. American Journal of Neuroradiology, Kish, S.J., el-Awar, M., Stuss, D. et al. (1994).
20: 1925–1930. Neuropsychological test performance in patients
Gambardella, A., Annesi, G., Bono, F. et al. (1998). CAG with dominantly inherited spinocerebellar ataxia:
repeat length and clinical features in three Italian Relationship to ataxia severity. Neurology,
families with spinocerebellar ataxia type 2 (SCA2): 44: 1738–1746.
Early impairment of Wisconsin Card Sorting Test and Klinke, I., Minnerop, M., Schmitz-Hübsch, T. et al. (2010).
saccade velocity. Journal of Neurology, 245: 647–652. Neuropsychological features of patients with spi-
Garrard, P., Martin, N.H., Giunti, P. and Cipollati, L. (2008). nocerebellar ataxia (SCA) types 1, 2, 3, and 6. The
Cognitive and social cognitive functioning in spinocer- Cerebellum, 9: 433–442.
ebellar ataxia: A preliminary characterization. Journal Koeppen, A.H. (2002). Neuropathology of the inher-
of Neurology, 255: 398–405. ited ataxias. In Manto, M.U., Pandolfo, M. (eds.), The
Gebhart, A.L., Petersen, S.E. and Thach, W.T. (2002). Role Cerebellum and Its Disorders. Cambridge, United
of the posterolateral cerebellum in language. Annals Kingdom: Cambridge University Press, pp. 387–405.
of the New York Academy of Sciences, 978: 318–333. Koziol, L.F., Budding, D., Andreasen, N. et al. (2014).
Geschwind, D.H., Perlman, S., Figueroa, C.P. et al. (1997). Consensus paper: The cerebellum’s role in movement
The prevalence and wide clinical spectrum of the and cognition. The Cerebellum, 13: 151–177.
spinocerebellar ataxia type 2 trinucleotide repeat in Le, T.H., Pardo, J.V. and Hu, X. (1998). 4T-fMRI study
patients with autosomal dominant cerebellar ataxia. of nonspatial shifting of selective attention:
American Journal of Human Genetics, 60: 842–850. Cerebellar and parietal contributions. Journal of
Goodin, D.S. (2003). Long-latency event-related poten- Neurophysiology, 79: 1535–1548.
tials. In Ebersole, J.S., Pedley, T.A. (eds.), Current Leggio, M.G., Tedesco, A.M., Chiricozzi, F.R. et al. (2008).
Practice of Clinical Electroencephalography, Third Cognitive sequencing impairment in patients with
Edition. Philadelphia, PA: Lippincott Williams & focal or atrophic cerebellar damage. Brain: A Journal
Wilkins, pp. 923–935. of Neurology, 131: 1332–1343.
The cerebellum and cognitive impairment 821

Le Pira, F., Zappala, G., Saponara, R. et al. (2002). Middleton, F.A. and Strick, P.L. (2000). Basal ganglia and
Cognitive findings in spinocerebellar ataxia type 2: cerebellar loops: Motor and cognitive circuits. Brain
Relationship to genetic and clinical variables. Journal Research Reviews, 31: 236–250.
of the Neurological Sciences, 201: 53–57. Middleton, F.A. and Strick, P.L. (2001). Cerebellar projec-
Levisohn, L., Cronin-Golomb, A. and Schmahmann, J.D. tions to the prefrontal cortex of the primate. Journal of
(2000). Neuropsychological consequences of cerebel- Neuroscience, 21: 700–712.
lar tumour resection in children: Cerebellar cogni- Moberget, T., Gullesen, E.H., Andersson, S. et al. (2014).
tive affective syndrome in a paediatric population. Generalized role for the cerebellum in encoding inter-
Brain: A Journal of Neurology, 123: 1041–1050. nal models: Evidence from semantic processing. The
Lilja, A., Hamalainen, P., Kaitaranta, E. et al. (2005). Journal of Neuroscience, 34 (8): 2871–2878.
Cognitive impairment in spinocerebellar ataxia type 8. Murdoch, B.E. (2010). The cerebellum and language:
Journal of the Neurological Sciences, 237: 31–38. Historical perspective and review. Cortex, 46: 858–868.
Lopes, T.M., D’Abreu, A., Franca, M.C. Jr. et al. (2013). Nagahama, Y., Fukuyama, H., Yamauchi, H. et al. (1996).
Widespread neuronal damage and cognitive dys- Cerebral activation during performance of a card sort-
function in spinocerebellar ataxia type 3. Journal of ing test. Brain: A Journal of Neurology, 119: 1667–1675.
Neurology, 260: 2370–2379. Neau, J. P., Arroyo-Anllo, E., Bonnaud, V. et al. (2000).
MacLeod, C.E., Zilles, K., Schleicher, A. et al. (2003). Neuropsychological disturbances in cerebellar infarcts.
Expansion of the neocerebellum in Hominoidea. Acta Neurologica Scandinavica, 102 (6): 363–370.
Journal of Human Evolution, 44: 401–429. O’Halloran, C.J., Kinsella, G.J. and Storey, E. (2012). The
McMurtray, A.M., Clark, D.G., Flood, M.K. et al. (2006). cerebellum and neuropsychological functioning.
Depressive and memory symptoms as present- Journal of Clinical and Experimental Neuropsychology,
ing features of spinocerebellar ataxia. Journal of 34: 35–56.
Neuropsychiatry and Clinical Neurosciences, 18: O’Reilly, J.X., Beckmann, C.F., Tomassini, V. et al. (2009).
420–422. Distinct and overlapping functional zones in the cer-
Malm, J., Kristensen, B., Karlsson, T. et al. (1998). ebellum defined by resting state functional connectiv-
Cognitive impairment in young adults with nfratento- ity. Cerebral Cortex, 20: 953–965.
rial infarcts. Neurology, 51: 433–440. Premack, D. and Woodruff, G. (1978). Does the chim-
Manes, F., Villamil, A.R., Ameriso, S. et al. (2009). ‘Real life’ panzee have a theory of mind? Behavioural and Brain
executive deficits in patients with focal vascular lesions Sciences 4: 515–629.
affecting the cerebellum. Journal of Neurological Ramnani, N., Behrens, T.E.J., Johansen-Berg, H. et al.
Sciences, 283: 95–98. (2006). The evolution of prefrontal inputs to the
Mariën, P., Achermann, H., Adamaszek, M. et al. (2014). cortico-pontine system: Diffusion imaging evidence
Consensus paper: Language and the cerebellum: An from macaque monkeys and humans. Cerebral Cortex,
ongoing enigma. The Cerebellum, 13: 386–410. 16: 811–818.
Mariën, P., Engelborghs, S., Fabbro, F. and De Ravizza, S.M. and Ivry, R.B. (2001). Comparison of the
Deyn, P.P. (2001). The lateralized linguistic cer- basal ganglia and cerebellum in shifting attention.
ebellum: A review and a new hypothesis. Brain and Journal of Cognitive Neuroscience, 13: 285–297.
Language, 79: 580–600. Ravizza, S.M., McCormick, C.A., Schlerf, J.E. et al. (2006).
Maruff, P., Tyler, P., Burt, T. et al. (1996). Cognitive deficits Cerebellar damage produces selective deficits in
in Machado–Joseph disease. Annals of Neurology, verbal working memory. Brain: A Journal of Neurology,
40: 421–427. 129: 306–320.
Marvel, C.L. and Desmond, J.E. (2010). The contributions Richter, S., Aslan, B., Gerwig, M. et al. (2007a). Patients
of cerebro-cerebellar circuitry to executive verbal with chronic focal cerebellar lesions show no
working memory. Cortex, 46: 880–895. ­cognitive abnormalities in a bedside test. Neurocase,
Massaquoi, S.G. and Topka, H. (2002). Models of cerebel- 13: 25–36.
lar function. In Manto, M.U., Pandolfo, M. (eds.), The Richter, S., Gerwig, M., Aslan, B. et al. (2007b). Cognitive
Cerebellum and its Disorders. Cambridge, , United functions in patients with MR-defined chronic
Kingdom: Cambridge University Press, pp. 69–94. focal cerebellar lesions. Journal of Neurology,
Matano, S. (2001). Brief communication: Proportions of 254: 1193–1203.
the ventral half of the cerebellar dentate nucleus in Roca, M., Gleichgerrcht, E., Ibánez, A. et al. (2013).
humans and great apes. American Journal of Physical Cerebellar stroke impairs executive functions but not
Anthropology, 114: 163–165. theory of mind. Journal of Neuropsychiatry and Clinical
Middleton, F.A. and Strick, P.L. (1997). Cerebellar output Neurosciences, 25: E48–E49.
channels. In Schmahmann, J.D. (ed.), The Cerebellum Roescke, S., Filla, I., Heim, S. et al. (2013). Progressive
and Cognition. San Diego, CA: Academic Press, pp. cognitive dysfunction in spinocerebellar ataxia type 3.
61–82 (International Review of Neurobiology, Vol. 41). Movement Disorders, 28: 1435–1438.
822 Dementia

Roldan Gerschcovich, E., Cerquetti, D., Tenca, E. et al. Tachibana, H., Kawabata, K., Tomino, Y. and
(2011). The impact of bilateral cerebellar damage Sugita, M. (1999). Prolonged P3 latency and
on theory of mind, empathy and decision making. decreased brain perfusion in cerebellar degenera-
Neurocase 17: 270–275. tion. Acta Neurologica Scandinavica, 100: 310–316.
Rüb, U., Schöls, L., Paulson, H. et al. (2013). Clinical Tanaka, H., Harada, M., Arai, M. and Hirata, K. (2003).
features, neurogenetics and neuropathology of the Cognitive dysfunction in cortical cerebellar atrophy
polyglutamine spinocerebellar ataxias type 1, 2, 3, 6 correlates with impairment of the inhibitory system.
and 7. Progress in Neurobiology, 104: 38–66. Neuropsychobiology, 47: 206–211.
Schmahmann, J.D. (ed.) (1997). The Cerebellum Thürling, M., Küper, M., Stafanescu, R. et al. (2011).
and Cognition. San Diego, CA: Academic Press Activation of the dentate nucleus in a verb generation
(International Review of Neurobiology, Vol. 41). task: A 7T MRI study. NeuroImage, 57: 1184–1191.
Schmahmann, J.D., MacMore, J. and Vangel, M. (2009). Timmann, D. and Daum, I. (2007). Cerebellar contributions
Cerebellar stroke without motor deficit: Clinical evi- to cognitive functions: A progress report after two
dence for motor and non-motor domains within the decades of research. The Cerebellum, 6: 159–162.
human cerebellum. Neuroscience, 162: 852–861. Timmann, D., Drepper, J., Maschke, M. et al. (2002). Motor
Schmahmann, J.D. and Pandya, D.N. (1997). The cere- deficits cannot explain impaired cognitive associative
brocerebellar system. International Review of learning in cerebellar patients. Neuropsychologia,
Neurobiology, 41:31–60. 40: 788–800.
Schmahmann, J.D. and Sherman, J.C. (1998). The cerebel- Timmann, D., Konczak, J., Ilg, W. et al. (2009). Current
lar affective cognitive syndrome. Brain: A Journal of advances in lesion-symptom mapping of the human
Neurology, 121: 561–579. cerebellum. Neuroscience, 162: 836–851.
Schweizer, T.A., Alexander, M.P., Cusimano, M. et al. Torrens, L., Burns, E., Stone, J. et al. (2008).
(2007). Fast and efficient visuotemporal atten- Spinocerebellar ataxia type 8 in Scotland: Frequency,
tion requires the cerebellum. Neuropsychologia, neurological, neuropsychological and neuropsychiatric
45: 3068–3074. findings. Acta Neurologica Scandinavica, 117: 41–48.
Schweizer, T.A., Alexander, M.P., Gillingham, S. et al. Townsend, J., Courchesne, E., Covington, J. et al. (1999).
(2010). Lateralized cerebellar contributions to word Spatial attention deficits in patients with acquired
generation: A phonemic and semantic fluency study. or developmental cerebellar abnormality. Journal of
Behavioural Neurology, 23: 31–37. Neurosciences, 19: 5632–5643.
Sokolovsky, N., Cook, A., Hunt, H. et al. (2010). A pre- Trojano, L., Chiacchio, L., Grossi, D. et al. (1998).
liminary study of cognition and social cognition in Determinants of cognitive disorders in autosomal
spinocerebellar ataxia types 2, 1, and 7. Behavioural dominant cerebellar ataxia type 1. Journal of the
Neurology, 23: 17–29. Neurological Sciences, 157: 162–167.
Stoodley, C.J. (2012). The cerebellum and cognition: Van Overwalle, F., Baetens, K., Mariën, P. and
Evidence from functional imaging studies. The Vandekerckhove, M. (2014). Social cognition and the
Cerebellum, 11: 352–365. cerebellum: A meta-analysis of over 350 fMRI studies.
Stoodley, C.J. and Schmahmann, J.D. (2009). Functional NeuroImage, 86: 554–572.
topography in the human cerebellum: A meta- White, M., Lalonde, R. and Botez-Marquard, T. (2000).
analysis of neuroimaging studies. NeuroImage, Neuropsychologic and neuropsychiatric characteristics
44 (2): 489–501. of patients with Friedreich’s ataxia. Acta Neurologica
Stoodley, C.J. and Schmahmann, J.D. (2010). Evidence for Scandinavica, 102: 222–226.
topographic organization in the cerebellum of motor Wollmann, T., Barroso, J., Monton, F. and Nieto, A. (2002).
control versus cognitive and affective processing. Neuropsychological test performance of patients with
Cortex, 46: 831–844. Friedreich’s ataxia. Journal of Clinical and Experimental
Storey, E., Forrest, S.M., Shaw, J.H. et al. (1999). Neuropsychology, 24: 677–686.
Spinocerebellar ataxia type 2: Clinical features of a Xiang, H., Lin, C., Ma, X. et al. (2003). Involvement of the
pedigree displaying prominent frontal-executive dys- cerebellum in semantic discrimination: An fMRI study.
function. Archives of Neurology, 56: 43–50. Human Brain Mapping, 18: 208–214.
Strick, P.L., Dum, R.P. and Fiez, J.A. (2009). Cerebellum Zawacki, T.M., Grace, J., Friedman, J.H. and Sudarsky, L.
and nonmotor function. Annual Reviews Neuroscience, (2002). Executive and emotional dysfunction in
32: 413–434. Machado–Joseph disease. Movement Disorders,
Suenaga, M., Kawai, Y., Watanabe, H. et al. (2008). 17: 1004–1010.
Cognitive impairment in spinocerebellar ataxia type
6. Journal of Neurology, Neurosurgery, and Psychiatry,
79: 496–499.
71
Delirium: A clinical overview

ANDREW TEODORCZUK AND DANIEL DAVIS

akin to be a ‘first rank symptom’ without which a diagno-


71.1 INTRODUCTION sis cannot be made.

Despite delirium being described in the Greek literature as 71.2.1 DIAGNOSTIC CRITERIA


far back as 400 bc, current levels of knowledge and practice,
in comparison to other medical conditions, lag considerably In 2013, new DSM-5 criteria for delirium were developed
behind. However, over the past 20 years, there has been sig- (American Psychiatric Association, 2013). These criteria
nificant progress in the way that we understand, conceptu- are outlined in Box 71.1. In comparison to Diagnostic and
alize and manage delirium (Maclullich et al., 2013). Statistical Manual of Mental Disorders, Fourth Edition
This chapter provides an overview of the clinical pre- (DSM-IV), which had been adopted widely within clinical
sentation and current diagnostic criteria (Diagnostic and practice, DSM-5 is less inclusive and more phenomenologi-
Statistical Manual of Mental Disorders, Fifth Edition [DSM- cally precise. Though DSM-5 elevates attention as a feature
5]), epidemiology and prevalence of delirium. In addition, of delirium, if the criteria are precisely applied there will be
clinical principles are described in relation to assessment, cases of people who had DSM-IV criteria delirium and who
management (non-pharmacological and pharmacological) would no longer qualify for delirium by DSM-5 standards.
and prevention of delirium. An up-to-date understanding of This is because it specifies the need for both disturbances
the pathophysiology of delirium and aetiology of delirium is in awareness and attention when pragmatically it may
outlined. The chapter concludes with current thoughts on not be possible to undertake tests of awareness in patients
the association between dementia and delirium, challenges who may have a low level of arousal (Meagher et al., 2014).
for researchers and possible future directions. Hence, in clinical practice, a relaxed approach to diagnosis
is advocated.

71.2.2 CLINICAL EXPERIENCE
71.2 CLINICAL FEATURES, CONCEPTS
AND DIAGNOSTIC CRITERIA OF Delirium is a highly distressing disorder for patients, rela-
DELIRIUM tives and indeed care staff (Partridge et al., 2013). To appre-
ciate just how distressing it can be, readers are directed to a
Delirium is a clinical syndrome characterized by the patient experience video available in the European Delirium
cardinal symptoms of inability to focus, shift or sustain Association website (Teodorczuk, 2012). Furthermore, it is
attention, acute change in mental state, fluctuations and not uncommon for patients to suffer post-delirium depres-
altered level of consciousness (Inouye et al., 2014). Other sive symptoms in this context.
supportive symptoms include disorganized thinking,
disturbance of the sleep–wake cycle, hallucinations and 71.2.3 CLINICAL CONCEPTS
delusions. The core features of delirium of inattention
and distractibility can be considered pragmatically as the Previously, delirium was considered to be the manifestation
ability to focus (for example, on a conversation) and is of a medical precipitant not requiring specific management

823
824 Dementia

morbidity, mortality and healthcare costs. Prevention of


BOX 71.1: DSM-5 criteria for delirium delirium therefore, can be a particularly high-yield strategy
(American Psychiatric Association, 2013). (Akunne et al., 2012).
Lastly, delirium is under-recognized in clinical set-
1. A disturbance in attention (i.e. reduced abil- tings (Collins et al., 2010). This potentially is as a conse-
ity to direct, focus, sustain and shift attention) quence of the fact that staff lack knowledge and expertise
AND awareness (reduced orientation to the and the absence of reliable collateral history. Common
environment). diagnostic mistakes include misattributing delirium
2. The disturbance develops over a short period of to depression or behavioural psychological symptoms
time (usually hours to a few days), represents a of dementia (BPSD). Moreover, there is an element of
change from baseline attention and awareness therapeutic nihilism when it comes to treating delirium
AND tends to fluctuate in severity during the (Teodorczuk et al., 2012). Furthermore, though there
course of the day. are many different assessment tools in practice, none
3. An additional disturbance in cognition (e.g. mem- are widely used and arguably, delirium screening should
ory deficit, disorientation, language, visuospatial form a core part of nursing assessments of the older con-
ability or perception). fused patients. Commonly used tools in practice include
4. The disturbances in criteria 1 and 3 are not better the Confusion Assessment Method (CAM), Confusion
explained by a pre-existing established or evolving Assessment Method for the Intensive Care Unit (CAM-
neurocognitive disorder and do not occur in the ITU), Delirium Observation Screening Scale (DOSS),
context of a severely reduced level of arousal, such evaluating the performance of the 4 ‘A’s Test (4AT),
as coma. Nursing delirium screening scale (NuDESC) and The
5. There is evidence from history, physical examina- Neelon and Champagne Confusion Scale (NEECHAM)
tion or laboratory findings that the disturbance (Wong et al., 2010).
is a direct physiological consequence of another
medical condition, substance intoxication or
withdrawal.

71.3 AETIOLOGY
beyond treatment of the underlying cause. However, delir-
ium is now best considered as a diagnostic treatment tar- As described above, delirium is considered conceptually as
geted in its own right (MacLullich and Hall, 2011; Meagher decompensation of the brain acutely. This is important in
et al., 2013). It can be conceptualized as acute brain failure order to understand the settings where prevalence would
in contrast to dementia (chronic brain failure). Expanding be expected to be high, such as in areas of high frailty (e.g.
on this concept further, delirium can be viewed as a marker nursing homes) and/or high acute illness burden (e.g. criti-
of brain vulnerability (the opposite of cognitive reserve) cal care). The Stress Vulnerability Model has been proposed
(Davis et al., 2012). and is widely accepted (Figure 71.1). In essence, any patient
Importantly, it should be recognized that delirium is the may become delirious if sufficiently insulted. Reciprocally,
harbinger of an acute medical problem, and hence an emer- the extent of vulnerability factors may allow delirium to
gency. It is a sensitive (but highly non-specific) marker of result from a relatively trivial precipitant.
illness in older persons and so can represent any number of Therefore, it follows that patients with dementia, who
occult presentation symptoms. are in a high vulnerability state due to their pre-existing
Diagnosis is made clinically and rests firmly on iden- cognitive impairment, will require a less severe insult
tifying the characteristic features as outlined above. than patients without dementia. Other important risk
Electroencephalography (EEG) changes, such as diffuse factors that increase vulnerability include age, frailty,
slowing in activity and increased theta and delta activity sensory impairment, polypharmacy and co-morbidity
can support the diagnosis in some cases (Kooi et al., 2012). (Table 71.1). Similarly, in settings such as ITU or surgery,
However, EEG is not routinely used in practice. where a patient has had a severer insult, they are more
Increasingly, it is recognized that delirium is both treat- likely to be delirious. Important noxious insults include
able and preventable; however, the body of evidence in sup- pain, constipation, medication, dehydration, nutritional
port of strategies to prevent delirium is considerably weaker deficiencies.
than that concerning treatment (MacLullich and Hall, 2011). A fundamental consideration that arises from the Stress
It has been reported by Inouye et al. (1999) that as many Vulnerability Model is that in patients with high vulner-
as 30% of cases of delirium could be prevented by means ability there are multiple causes for delirium. Therefore,
of a multicomponent tailored intervention. Though in the treatment should be broadened to identify multiple causes
real world setting this figure may not be an easy target, or insults. Too often a failure of non-pharmacological inter-
the fact that delirium is preventable is particularly impor- ventions is a consequence of inadequate understanding of
tant, as delirium carries a negative impact in both terms of the causal factors and at risk vulnerability state.
Delirium: A clinical overview 825

Precipitating factors Table 71.1 Common risk and precipitating factors in


Acute medical or surgical conditions delirium
caused by drugs, toxics
Insult intensity

Precipitating factors
Vulnerability factors (insults)
Age Infections
Cognitive impairment Medications
Depression Hypoxia
Age cognitive impairment frailty Frailty and functional Metabolic disturbance/

Predisposing
Protective

factors
dependency dehydration
factors

Co-morbidity Physical restraints


Nutritional state Urinary catheterization
Sensory impairment Constipation
Pain
Delirium
Environmental changes

Figure 71.1 Relationship between predisposing, protec- 71.4.3 SUBSYNDROMAL DELIRIUM


tive and precipitating factors in delirium. (From Cerejeira,
J. and Mukaetova-Ladinska, E.B., Nursing Research and
Subsyndromal delirium is an emerging clinical concept
Practice, 2011, 875196, 2011. With permission under CC that refers to a delirium-like illness that does not reach the
attribution.) diagnostic criteria for full delirium. Typically symptoms
may be of a lesser severity. Symptoms include impaired
cognitive function, increased motor activity (restlessness,
71.4 CLINICAL PRESENTATIONS OF agitation, sensitivity to stimuli) and perceptual mood dis-
DELIRIUM turbance. However, circadian rhythm may be preserved and
the degree of fluctuation less in comparison to delirium. A
study by Meagher (2012) demonstrated in a heterogeneous
71.4.1 SUBTYPES group of patients with subsyndromal delirium that the syn-
drome was present in 27% of patients with post-delirium.
Three different subtypes of delirium have been defined
However, the prevalence of subsyndromal delirium across
(O’Keeffe and Lavan, 1999; Santana Santos et al., 2005).
all settings, whether it is commoner in older adults or those
The hypoactive subtype, which is more prevalent in a
with dementia is not fully established.
nursing home care setting accounts for approximately
Studies have shown that patients who develop sub-
one-third of delirium presentations. Patients with this pre-
syndromal delirium after an episode of delirium are less
sentation will be acutely drowsy, perhaps, unable to have
likely to be discharged to the community, have longer hos-
a conversation. Hyperactive subtypes are more common
pital stays and greater medical complications including
in those without dementia, and these patients account
higher risk of falls (Marcantonio et al., 2005). Presently, it
for a further one–third; mixed subtypes account for the
is unclear whether subsyndromal delirium should form a
remainder. Though typically medications are used to treat
distinct treatment target and further studies are warranted.
hyperactive delirium (Morandi et al., 2013), increasingly,
However, non-pharmacological interventions (perhaps sim-
some clinicians’ medication for hypoactive delirium when
ply representing good clinical care) are vital and low-risk
risks are present (Leentjens et al., 2014). In practice, the
in delirium. Given a diagnosis of subsyndromal delirium,
hypoactive subtype is most easily missed (O’Keeffe, 1999)
it is, therefore, prudent to implement non-pharmacological
and may carry a worse prognosis for this reason.
approaches.

71.4.2 PERSISTENT DELIRIUM
Dedicated follow-up of persons with delirium demon-
strate that it may persist in 20% after 1 year (Cole et al.,
71.5 INCIDENT AND PREVALENT
2009). This presentation is associated with worsening out- DELIRIUM
comes though not necessarily greater mortality (Klein
Klouwenberg et al., 2014). Clinically it may be difficult to Delirium can also be differentiated in terms of incident or
differentiate from a dementia illness, however, the cardi- prevalent delirium in a hospital setting (Siddiqi et al., 2007;
nal features of inattention, fluctuations and disorganiza- Young et al., 2010). Prevalent delirium relates to patients
tion to thoughts would help differentiate from dementia, who present to hospital with a delirium syndrome. Incident
which is characterized by clear consciousness and a differ- delirium, however, relates to patients who develop delirium
ent symptom profile. during hospitalization as a result of further insult or insults
826 Dementia

in the hospital. The drive of guidelines, such as The National causes. In addition, a midstream specimen of urine (MSU)
Institute for Health and Care Excellence (NICE) in the test to assess urinary tract infection (UTI) is indicated.
United Kingdom (NICE, 2010), is to prevent the develop- Computed tomography (CT) is only recommended if there
ment of such incident delirium. is a history of falls or neurology on examination or sus-
pected metastases.

71.6 PREVALENCE 71.8.1 ASSESSMENT TOOLS


Increasingly, assessment screening tools are entering
Delirium’s prevalence in different populations alters clinical practice. Commonly used tools include the CAM,
depending upon the level of insult. As such, in a general DOSS and 4AT. Whichever tool is chosen is dependent
hospital upon admission, the prevalence is between 10% and upon sensitivity and specificity for the particular group
40%, depending upon setting (Vasilevskis et al., 2012). This of healthcare professionals using the tool. In general, the
rises to 25%–60% of incident delirium during admission. CAM tends to be better when undertaken by medically
In high-risk areas, such as vascular surgery (Balasundaram trained staff (Lemiengre et al., 2006), whereas the DOSS
and Holmes, 2007) and orthopaedics (Bruce et al., 2007), mainly lends itself to nursing staff for usage (Detroyer
the prevalence of delirium rises even further. Indeed, in et al., 2014).
the ITU population, the incidence can be up to 80% (Pisani Once the patient has been assessed, the delirium sta-
et al., 2003). In the hospital, delirium has been described as tus should be clearly recorded in the notes. This will help
the most common complication. A recent study of all medi- to initiate delirium management pathways, as well as raise
cal admissions aged over 70 found the total prevalence of awareness for other professionals involved in the care of the
delirium was 27% (Whittamore et al., 2014). Delirium was patient. If they are in a high-risk state but do not suffer from
complicated by dementia in 19% and 37% of patients admit- delirium, it is important to put into place measures to pre-
ted with delirium died within 6 months. vent delirium: promote good nursing care, mobility to the
patient and vigilant to the possibility of medical deteriora-
tion are some of them.
71.7 PROGNOSIS

Delirium has been demonstrated in various studies to be 71.9 MANAGEMENT OF DELIRIUM


associated with an increased mortality rate, length of stay
and rates of institutionalization (McCusker et al., 2003; Treatment of an identified delirium falls into three catego-
Witlox et al., 2010). Delirium has also been shown to acceler- ries, which are as follows:
ate cognitive decline in patients with dementia (see Sections
71.8 and 71.9). Interestingly, though lack of apolipoprotein 1. Treating the underlying cause
E (ApoE) ε4 allele, being female and lower levels of insulin- 2. Optimizing brain function by non-pharmacological
like growth factor 1 (IGF-1) have been associated with a full measures
recovery from delirium; severity of delirium, underlying 3. Symptom relief through medication if a patient is
presence of dementia and advanced age do not appear to be distressed or in a high-risk state
associated with a complete recovery (Adamis et al., 2007).
The guiding principle for the treatment of delirium is to
promote non-pharmacological interventions as a treatment
of choice before attempting pharmacological interventions.
71.8 ASSESSMENT OF DELIRIUM Crucially, a clear and comprehensive non-pharmacological
treatment plan involving optimal brain protection follows
Guidelines suggest that all patients who are at high risk, from a good understanding of aetiology of the delirium of
that is to say have vulnerability factors for delirium as listed each patient.
above, should be assessed for delirium within 24 hours of
admission (NICE, 2010). Typically, assessments should 71.9.1 TREATING THE UNDERLYING
involve talking with carers to determine if there has been CAUSE
an acute change in mental status. Also, it is important to be
vigilant for behaviour that may indicate a hypoactive delir- The most common causes for delirium include medica-
ium. Along with a full medical history and examination, tion changes. As such, potentially deliriogenic medications
particular emphasis is needed to ensure an accurate drug should be stopped. Inappropriate medications to remove as
history is taken, including any recently stopped medication. indicated by the American Geriatrics Society Beers Criteria
In terms of investigations for the assessment of delirium, a Update Expert Panel (2012) include tricyclic antidepres-
full screening of blood is necessary to look for physiological sants, anticholinergics, benzodiazepines, chlorpromazine,
Delirium: A clinical overview 827

steroids, pethidine, H2 antagonists, sedative hypnotics and prevention, for example, in preoperative settings, is not
thioradazine. recommended routinely (American Geriatrics Society, 2014).
Other easily identifiable causes for treatment include There is growing evidence that melatonin may as well
pain, nutrition and constipation. Dehydration should be be beneficial in the treatment of delirium (Al-Aama et al.,
attended to, as evidenced by urea and electrolytes. Infection 2011), though the largest trial till date did not show a reduc-
must be treated, in particular UTIs and pneumonias. As tion in delirium incidence in the ICU (de Jonghe et al.,
indicated above, in a vulnerable patient, there are likely to 2014). Melatonin is well tolerated and can help with the
be numerous causes for delirium and a multifactorial treat- sleep–wake cycle.
ment plan addressing all insults should be developed. The place of cholinesterase inhibitors (ChEIs) in the
management of delirium is unclear. In ITU patients initi-
71.9.2 BRAIN OPTIMIZATION ated with rivastigmine for delirium, there was significantly
higher mortality (van Eijk et al., 2010). However, it is unclear
In terms of brain optimization, further non-pharmacolog- as to the extent to which these findings would apply to the
ical measures include orientation of the patient. Involving population outside critical care (Skrobik, 2010). Good prac-
carers to help with this, in addition to managing a patient tice, until further studies are undertaken, would be not to
is essential. Furthermore, environmental measures can be initiate a ChEI in a patient with delirium. If a patient suf-
put into place to promote orientation by means of clocks fers from delirium and is already on a ChEI, then it could
and calendars. Other environmental strategies to promote reasonably be continued. To date there are no studies con-
brain function include appropriate lighting for the time of ducted on the relationship between memantine and delir-
day. Reducing change of hospital or wards is particularly ium. Further studies are warranted.
important in the treatment of delirium. Avoiding catheters
and restraints is to be supported. Addressing risk factors,
such as sensory impairment using hearing aids and glasses
is a particularly high-yield non-pharmacological measure. 71.10 PATHOPHYSIOLOGY
Early and safe mobilization of patients by means of phys-
iotherapy will also help to optimize brain function. Lastly, Delirium pathophysiology is poorly understood. Clearly,
attending to sleep difficulties by means of noise reduction certain pathophysiological processes can result in direct
strategy and promoting good sleep hygiene is indicated brain injury. Examples might include hypoxaemia, stroke,
where possible. hypoglycaemia or medication effects. However, more com-
plex explanations are required to account for the interplay
71.9.3 SYMPTOM RELIEF IN THE between predisposing pathology and acute precipitants.
DISTRESSED PATIENT An emerging strategy is that aberrant stress responses
mediate between peripheral stimuli and the brain
If a patient is particularly distressed, or there are risks to (MacLullich et al., 2008; Cunningham and MacLullich,
their (or others’) safety, it would be appropriate to consider 2013). Systemic inflammation can communicate with
a short course of medication. In England and Wales, the neural substrates through a variety of mechanisms. These
NICE guidelines support the use of antipsychotics, such as include areas where blood-brain barrier is deficient, directly
olanzapine and haloperidol, for a short course of 1 week to through the vagus nerve and where endothelial glial cells
help manage complex delirium (NICE, 2010). It is important directly transmit cytokines into the brain.
to note that there is no indication for prescribing benzodi- There may be a neuroendocrine basis to some of the cog-
azepines to help with the management of a distressed delir- nitive dysfunction seen in delirium and acute illness. The
ium in routine clinical practice of hospital wards (Breitbart normal adaptive stress response is understood to become
et al., 1996). dysregulated in dementia, especially given the higher lim-
The evidence for treating delirium with antipsychotics is bic control of the hypothalamus–pituitary–adrenal axis
weak and initiating antipsychotics involve carefully weigh- (O’Brien et al., 1996). In such situations, relatively trivial
ing the risks and benefits on an individual patient basis. physiological stressors may lead to exaggerated and sus-
Nonetheless, 75% of patients who do receive a short course tained levels of cortisol. In itself, cortisol cytotoxicity could
of antipsychotics will respond (Meagher et al., 2013). It is account for some of the long-term sequelae of delirium.
not clear if delirium itself is being treated or if the psychotic Evidence supporting these hypotheses comes from a small
symptoms are being reduced. In general, atypical antipsy- range of clinical studies and is also available. One strategy
chotics are to be promoted rather than haloperidol due to the that has yielded interesting results has been the examina-
reduced incidence of extrapyramidal side effects (EPSEs). In tion of cerebrospinal fluid in individuals undergoing spinal
patients with Parkinson’s dementia or Lewy body disease, anaesthesia for elective or emergency surgery. A number of
it may be appropriate to treat a severe delirium with low biomarkers have been considered, though, general studies
dose quetiapine, though cautious attendance to worsening conducted have been limited with little overlap (Hall et al.,
of EPSEs is necessary. The evidence for antipsychotic use in 2011). Nevertheless, cortisol, interleukin-1beta (IL-1β), IL-6,
delirium prevention is less clear. As yet, their use in delirium IL-8 and S-100 (β-subunit) all have been demonstrated as
828 Dementia

altered in the cerebrospinal fluid (CSF) of delirium patients been implicated include dopamine (Maldonado, 2008). The
(Pearson et al., 2010; MacLullich et al., 2011; Hall et al., 2013; challenge is that delirium is a highly heterogeneous con-
Westhoff et al., 2013; Cape et al., 2014). These changes are struct and hence, the pathophysiological mechanism(s)
consistent with stress, pro-inflammatory and cause direct may vary markedly, for example, between the ITU setting
injury to astrocytes as mediators of delirium. The degree to and geriatric clinical environments. There is not yet direct
which these results might be confounded by age and pre- evidence that delirium per se causes concurrent neuronal
existing cognitive impairment has not always been fully death by the same mechanisms.
addressed. The largest investigation examining demen-
tia biomarkers directly (beta-amyloid [Aβ] 1-42, tau and
phosphorylated-tau) did not show association to delirium 71.11 RELATIONSHIP BETWEEN
suggesting that delirium pathophysiology might be distinct DELIRIUM AND DEMENTIA
from dementia (Witlox et al., 2011).
The potential for neuroimaging to elucidate delirium
pathophysiology is also of interest, but such studies are The association between delirium and dementia is complex.
difficult to conduct (Soiza et al., 2008). There is a general Studies of individuals with delirium followed up for wors-
limitation arising from the fact that usually only a fraction ened cognition are usually biased by unrecognized (and
of people with delirium can be successfully diagnosed. In a unquantified) pre-existing cognitive deficits. Nonetheless,
cohort of critically ill patients, longer duration of delirium it is clear that delirium does not recover in all persons and
was associated with smaller brain volumes and evidence prospectively linking delirium with permanent decrements
of disruption of the white matter (Gunther et al., 2012; in cognitive function challenges the construct of dementia,
Morandi et al., 2012) was found. These findings were in since it suggests that chronic cognitive impairment may be
turn associated with worse cognition after 1 year follow-up. affected by incomplete recovery from acute illness.
Preoperative leukoaraiosis has been demonstrated to pre- Prospective studies more reliably establish the tempo-
dict post-operative delirium after elective cardiac surgery ral sequence between baseline cognitive function, incident
(Hatano et al., 2013). delirium and subsequent cognitive impairment. These have
Compared to the number of clinical studies, relatively less shown cognition can decline after hospitalization (from any
work has been undertaken using animal models. Finding cause) though these studies have not specifically considered
meaningful cognitive behavioural animal models to explore delirium (Mathews et al., 2013). Equally, it is possible that
possible hypotheses has not been straightforward. Any clini- systemic inflammation may lead to accelerated rates of cog-
cally relevant experimental approach must capture both nitive decline in dementia, even in the absence of delirium
predisposing and precipitating dimensions. Mouse models (Holmes et al., 2009).
for this have been specifically developed using methods for Ascertaining delirium in the context of an epidemiological
mimicking and predisposing dementia (prior pathology), study is challenging. Three cohort studies have investigated
superimposed with inflammatory challenge to simulate bac- this and each could only determine delirium retrospectively.
terial or viral infection (Field et al., 2010; Murray et al., 2012). Nonetheless, the consistent finding is that delirium (or symp-
Here, prior pathology can be modelled by inducing neuro- toms plausibly attributable to delirium) resulted in an acceler-
degeneration or selective partial lesioning of the cholinergic ated degree of cognitive decline, over and above that expected
system (Cunningham et al., 2005; Field et al., 2012). In these from pre-existing cognitive function (Fong et al., 2009; Gross
mice, acute peripheral inflammation leads to acute deficits in et al., 2012; Davis et al., 2012, 2014).
cognition and these behavioural effects are consistent regard-
less of the underlying prior pathology. The pathophysiological
pathways that underpin these relationships is beginning to be 71.12 FUTURE DIRECTIONS FOR
described, perhaps identifying future targets for intervention.
DELIRIUM RESEARCH AND
Emerging findings from mouse models suggest that
microglial priming and exaggerated responses to systemic
PRACTICE
inflammatory events may sometimes play a role in the
pathophysiology of delirium. Microglia express cyclooxy- Acknowledging delirium as a determinant of chronic cog-
genase (COX) and inhibition of this using COX-1 has been nitive impairment positions delirium as a critical event in
shown to be protective against systemic inflammation- the path towards dementia. In recognizing that neurode-
induced deficits (Griffin et al., 2013). Similarly, the same generative processes may interact with delirium and/or its
deficits reproduced in cholinergically-deficient mice could precipitants, future work needs to consider the long-term
be blocked by donepezil (Field et al., 2012). In support, impact of acute illness on the brain.
the role of cholinergic deficiency in aetiology of delirium A greater breadth of experimental work is necessary,
leads to the fact that anticholinergic drugs are particularly even more pressing for delirium as it is for dementia. The
deliriogenic. biological substrates of both these conditions might best be
The pathophysiology of delirium is undoubtedly multi- investigated in tandem rather than separately. Researchers
dimensional. Other important neurotransmitters that have carrying out experiments in dementia may not even be fully
Delirium: A clinical overview 829

aware of delirium as a construct and this is likely leading to Al-Aama, T., Brymer, C., Gutmanis, I. et al. (2011).
missed opportunities for translational collaboration. Melatonin decreases delirium in elderly patients:
Currently, the specification of arousal stated in the A randomized, placebo-controlled trial. International
DSM-5 as an explicit manifestation of inattention is highly Journal of Geriatric Psychiatry, 26: 687–694.
relevant. Acute illness, underlying dementia and superim- American Geriatrics Society (2014). American Geriatrics
posed delirium, each have an impact on the level of arousal. Society abstracted clinical practice guideline for
Therefore, the extent to which each has an impact (individu- postoperative delirium in older adults. Journal of the
ally and in concert) on cognitive and functional outcomes American Geriatrics Society, 63 (1): 142–150.
urgently warrants research. American Geriatrics Society Beers Criteria Update Expert
A number of assessment scales is available for delirium Panel (2012). American Geriatrics Society updated
in clinical practice. Clearly, no single instrument is suitable Beers Criteria for potentially inappropriate medication
for all patient groups. The diagnostic accuracy will vary use in older adults. Journal of the American Geriatrics
with setting. The degree of training required, along with Society, 60 (4): 616–631.
the length of time to administer any such tool is crucial American Psychiatric Association (ed.) (2013). Diagnositic
if widespread improvements in delirium recognition are and Statistical Manual of Mental Disorders, Fifth
to be effective. Change in diagnostic accuracy when there Edition. Washington, DC: American Psychiatric
is underlying dementia is important to measure and war- Association.
rants specific study. Where low arousal states are intrinsic Balasundaram, B. and Holmes, J. (2007). Delirium in
to delirium, formal cognitive testing is usually not possible, vascular surgery. European Journal of Vascular and
though, few instruments allow for this. Individual tools Endovascular Surgery, 34: 131–134.
may be compared in randomized controlled trials and this Breitbart, W., Marotta, R., Platt, M.M. et al.
will help clarify which will work best in practice. In general, (1996). A double-blind trial of haloperidol, chlor-
greater detail reporting the exact operationalization of ref- promazine, and lorazepam in the treatment of delirium
erence standards within validation studies is essential. in hospitalized AIDS patients. American Journal of
While the public health impact of dementia is widely Psychiatry, 153: 231–237.
acknowledged, the relevance of delirium in highlight- Bruce, A.J., Ritchie, C.W., Blizard, R. et al. (2007). The inci-
ing vulnerabilities to cognitive impairment arguably will dence of delirium associated with orthopedic surgery:
become increasingly prominent. Delirium and dementia A meta-analytic review. International Psychogeriatrics,
care within acute hospitals must be closely aligned. The eco- 19: 197–214.
nomic costs of delirium should be a potent driver for change. Cape, E., Hall, R.J., van Munster, B.C. et al. (2014).
Innovations in delirium education may facilitate this as will Cerebrospinal fluid markers of neuroinflammation in
a more conscious approach to knowledge translation. delirium: A role for interleukin-1beta in delirium after
hip fracture. Journal of Psychosomatic Research, 77:
219–225.
71.13 CONCLUSIONS Cerejeira, J. and Mukaetova-Ladinska, E.B.
(2011). A clinical update on delirium: From early recog-
nition to effective management. Nursing Research and
Delirium is a distressing, under-recognized and expensive
Practice, 2011: 875196.
clinical entity that influences cognitive outcomes in both
Cole, M.G., Ciampi, A., Belzile, E. and Zhong, L. (2009).
patients with and without dementia. Treating delirium
Persistent delirium in older hospital patients: A sys-
hinges on addressing the underlying cause, brain optimiza-
tematic review of frequency and prognosis. Age and
tion and symptom relief if distressed. Given that it is pre-
Ageing, 38: 19–26.
ventable and treatable, substantial clinical efforts should be
Collins, N., Blanchard, M.R., Tookman, A. and Sampson,
directed to its identification in high-risk groups and there-
E.L. (2010). Detection of delirium in the acute hospital.
after management in its own right.
Age and Ageing, 39: 131–135.
Cunningham, C. and MacLullich, A.M. (2013). At the
extreme end of the psychoneuroimmunological spec-
REFERENCES
trum: Delirium as a maladaptive sickness behaviour
Adamis, D., Treloar, A., Darwiche, F.Z., Gregson, N., response. Brain, Behavior, and Immunity, 28: 1–13.
Macdonald, A.J., Martin, F.C. (2007). Associations of Cunningham, C., Wilcockson, D.C., Campion, S. et al.
delirium with in-hospital and in 6-months mortality (2005). Central and systemic endotoxin challenges
in elderly medical inpatients. Age and ageing, 36(6): exacerbate the local inflammatory response and
644–649. increase neuronal death during chronic neurodegen-
Akunne, A., Murthy, L. and Young. J. (2012). Cost- eration. The Journal of Neuroscience, 25: 9275–9284.
effectiveness of multi-component interventions to Davis, D.H., Barnes, L.E., Stephan, B.C. et al. (2014). The
prevent delirium in older people admitted to medical descriptive epidemiology of delirium symptoms in a
wards. Age and Ageing, 41 (3): 285–291. large population-based cohort study: Results from
830 Dementia

the Medical Research Council Cognitive Function Holmes, C., Cunningham, C., Zotova, E. et al. (2009).
and Ageing Study (MRC CFAS). BMC Geriatrics, Systemic inflammation and disease progression in
14: 87. Alzheimer disease. Neurology, 73: 768–774.
Davis, D.H., Muniz Terrera, G., Keage, H. et al. (2012). Inouye, S.K., Bogardus, S.T., Jr., Charpentier, P.A. et al.
Delirium is a strong risk factor for dementia in the (1999). A multicomponent intervention to prevent
oldest-old: A population-based cohort study. Brain: delirium in hospitalized older patients. New England
A Journal of Neurology, 135 (Pt 9): 2809–2816. Journal of Medicine, 340: 669–676.
de Jonghe, A., van Munster, B.C., Goslings, J.C. et al. Inouye, S.K., Westendorp, R.G. and Saczynski, J.S. (2014).
(2014). Effect of melatonin on incidence of delirium Delirium in elderly people. Lancet, 383: 911–922.
among patients with hip fracture: A multicentre, Klein Klouwenberg, P.M., Zaal, I.J., Spitoni, C. et al. (2014).
double-blind randomized controlled trial. Canadian The attributable mortality of delirium in critically ill
Medical Association Journal, 186: E547–E556. patients: Prospective cohort study. British Medical
Detroyer, E., Clement, P.M., Baeten, N. et al. (2014). Journal, 349: g6652.
Detection of delirium in palliative care unit patients: Kooi, A.W., Leijten, F.S., van der Wekken, R.J. and Slooter,
A prospective descriptive study of the Delirium A.J. (2012). What are the opportunities for EEG-
Observation Screening Scale administered by bedside based monitoring of delirium in the ICU? The Journal
nurses. Palliative Medicine, 28: 79–86. of Neuropsychiatry and Clinical Neurosciences, 24:
Field, R., Campion, S., Warren, C. et al. (2010). Systemic 472–477.
challenge with the TLR3 agonist poly I:C induces Leentjens, A.F., Molag, M.L., Van Munster, B.C. et al.
amplified IFNalpha/beta and IL-1beta responses in the (2014). Changing perspectives on delirium care:
diseased brain and exacerbates chronic neurodegen- The new Dutch guideline on delirium. Journal of
eration. Brain, Behavior, and Immunity, 24: 996–1007. Psychosomatic Research, 77: 240–241.
Field, R.H., Gossen, A. and Cunningham, C. (2012). Prior Lemiengre, J., Nelis, T., Joosten, E. et al. (2006). Detection
pathology in the basal forebrain cholinergic system of delirium by bedside nurses using the confu-
predisposes to inflammation-induced working memory sion assessment method. Journal of the American
deficits: Reconciling inflammatory and cholinergic Geriatrics Society, 54: 685–689.
hypotheses of delirium. The Journal of Neuroscience, Maclullich, A.M., Anand, A., Davis, D.H. et al. (2013). New
32: 6288–6294. horizons in the pathogenesis, assessment and man-
Fong, T.G., Jones, R.N., Shi, P. et al. (2009). Delirium agement of delirium. Age and Ageing, 42: 667–674.
accelerates cognitive decline in Alzheimer disease. MacLullich, A.M., Edelshain, B.T., Hall, R.J. et al. (2011).
Neurology, 72: 1570–1575. Cerebrospinal fluid interleukin-8 levels are higher in
Griffin, E.W., Skelly, D.T., Murray, C.L. and Cunningham, people with hip fracture with perioperative delirium
C. (2013). Cyclooxygenase-1-dependent prostaglan- than in controls. Journal of the American Geriatrics
dins mediate susceptibility to systemic inflammation- Society, 59: 1151–1153.
induced acute cognitive dysfunction. The Journal of MacLullich, A.M. and Hall, R.J. (2011). Who understands
Neuroscience, 33: 15248–15258. delirium? Age and Ageing, 40: 412–414.
Gross, A.L., Jones, R.N., Habtemariam, D.A. et al. (2012). MacLullich, A.M.J., Ferguson, K.J., Miller, T. et al. (2008).
Delirium and long-term cognitive trajectory among Unravelling the pathophysiology of delirium: A focus
persons with dementia. Archives of Internal Medicine, on the role of aberrant stress responses. Journal of
172: 1324–1331. Psychosomatic Research, 65 (3): 229–238.
Gunther, M.L., Morandi, A., Krauskopf, E. et al. (2012). The Maldonado, J.R. (2008). Pathoetiological model of
association between brain volumes, delirium duration, delirium: A comprehensive understanding of the neu-
and cognitive outcomes in intensive care unit survi- robiology of delirium and an evidence-based approach
vors: The VISIONS cohort magnetic resonance imaging to prevention and treatment. Critical care clinics, 24(4):
study. Critical Care Medicine, 40: 2022–2032. 789–856, ix.
Hall, R.J., Ferguson, K.J., Andrews, M. et al. (2013). Marcantonio, E.R., Kiely, D.K., Simon, S.E. et al. (2005).
Delirium and cerebrospinal fluid S100B in hip fracture Outcomes of older people admitted to postacute
patients: A preliminary study. The American Journal of facilities with delirium. Journal of the American
Geriatric Psychiatry, 21 (12): 1239–1243. Geriatrics Society, 53(6):963–969.
Hall, R.J., Shenkin, S.D. and Maclullich, A.M. (2011). Mathews, S.B., Arnold, S.E. and Epperson, C.N. (2013).
A systematic literature review of cerebrospinal fluid Hospitalization and cognitive decline: Can the nature
biomarkers in delirium. Dementia and Geriatric of the relationship be deciphered? The American
Cognitive Disorders, 32: 79–93. Journal of Geriatric Psychiatry, 22 (5): 465–480.
Hatano, Y., Narumoto, J., Shibata, K. et al. (2013). White- McCusker, J., Cole, M., Dendukuri, N. et al. (2003). The
matter hyperintensities predict delirium after cardiac course of delirium in older medical inpatients: A pro-
surgery. The American Journal of Geriatric Psychiatry, spective study. Journal of General Internal Medicine,
21: 938–945. 18: 696–704.
Delirium: A clinical overview 831

Meagher, D., Adamis, D., Trzepacz, P., Leonard, M. (2012). Santana Santos, F., Wahlund, L.O., Varli, F. et al. (2005).
Features of subsyndromal and persistent delirium. The Incidence, clinical features and subtypes of delirium
British Journal of Psychiatry, 200(1): 37–44. in elderly patients treated for hip fractures. Dementia
Meagher, D.J., McLoughlin, L., Leonard, M. et al. (2013). and Geriatric Cognitive Disorders, 20: 231–237.
What do we really know about the treatment of delir- Siddiqi, N., Stockdale, R., Britton, A.M. and Holmes,
ium with antipsychotics? Ten key issues for delirium J. (2007). Interventions for preventing delirium in
pharmacotherapy. The American Journal of Geriatric hospitalised patients. Cochrane Database Systematic
Psychiatry, 21: 1223–1238. Reviews, (2): CD005563.
Meagher, D.J., Morandi, A., Inouye, S.K. et al. (2014). Skrobik, Y. (2010). Delirium treatment: An unmet chal-
Concordance between DSM-IV and DSM-5 criteria for lenge. Lancet, 376: 1805–1807.
delirium diagnosis in a pooled database of 768 pro- Soiza, R.L., Sharma, V., Ferguson, K. et al. (2008).
spectively evaluated patients using the delirium rating Neuroimaging studies of delirium: A systematic review.
scale-revised-98. BMC Medicine, 12: 164. Journal of Psychosomatic Research, 65: 239–248.
Morandi, A., Davis, D., Taylor, J.K. et al. (2013). Consensus Teodorczuk, A. (2012). Patient Experience Video. Available
and variations in opinions on delirium care: A sur- at http://www.europeandeliriumassociation.com/
vey of European delirium specialists. International patient-video.html. Accessed 12 June 2016.
Psychogeriatrics, 25: 2067–2075. Teodorczuk, A., Reynish, E. and Milisen, K. (2012).
Morandi, A., Rogers, B.P., Gunther, M.L. et al. (2012). The Improving recognition of delirium in clinical practice:
relationship between delirium duration, white mat- A call for action. BMC Geriatrics, 12: 55.
ter integrity, and cognitive impairment in intensive van Eijk, M.M., Roes, K.C., Honing, M.L. et al. (2010).
care unit survivors as determined by diffusion tensor Effect of rivastigmine as an adjunct to usual care with
imaging: The VISIONS prospective cohort magnetic haloperidol on duration of delirium and mortality
resonance imaging study. Critical Care Medicine, 40: in critically ill patients: A multicentre, double-blind,
2182–2189. placebo-controlled randomised trial. Lancet, 376:
Murray, C., Sanderson, D.J., Barkus, C. et al. (2012). 1829–1837.
Systemic inflammation induces acute working memory Vasilevskis, E.E., Han, J.H., Hughes, C.G. and Ely, E.W.
deficits in the primed brain: Relevance for delirium. (2012). Epidemiology and risk factors for delirium
Neurobiology of Aging, 33: 603–616. across hospital settings. Best Practice and Research
NICE (2010). Delirium: Diagnosis, Prevention and Clinical Anaesthesiology, 26: 277–287.
Management. (Clinical Guideline 103). London, United Westhoff, D., Witlox, J. Koenderman, L. et al. (2013).
Kingdom: NICE. Preoperative cerebrospinal fluid cytokine levels and
O’Brien, J.T., Ames, D., Schweitzer, I. et al. (1996). the risk of postoperative delirium in elderly hip frac-
Clinical and magnetic resonance imaging correlates ture patients. Journal of Neuroinflammation, 10: 122.
of h
­ ypothalamic-pituitary-adrenal axis function in Whittamore, K.H., Goldberg, S.E., Gladman, J.R. et al.
depression and Alzheimer’s disease. British Journal of (2014). The diagnosis, prevalence and outcome of
Psychiatry, 168: 679–687. delirium in a cohort of older people with mental health
O’Keeffe, S.T. (1999). Clinical subtypes of delirium in the problems on general hospital wards. International
elderly. Dementia and Geriatric Cognitive Disorders, Journal of Geriatric Psychiatry, 29: 32–40.
10: 380–385. Witlox, J., Eurelings, L.S., de Jonghe, J.F. et al. (2010).
O’Keeffe, S.T. and Lavan, J.N. (1999). Clinical significance Delirium in elderly patients and the risk of postdis-
of delirium subtypes in older people. Age and Ageing, charge mortality, institutionalization, and dementia:
28: 115–119. A meta-analysis. JAMA, 304: 443–451.
Partridge, J.S., Martin, F.C., Harari, D. and Dhesi, J.K. Witlox, J., Kalisvaart, K.J., de Jonghe, J.F. et al. (2011).
(2013). The delirium experience: What is the effect Cerebrospinal fluid beta-amyloid and tau are not
on patients, relatives and staff and what can be done associated with risk of delirium: A prospective cohort
to modify this? International Journal of Geriatric study in older adults with hip fracture. Journal of the
Psychiatry, 28: 804–812. American Geriatrics Society, 59: 1260–1267.
Pearson, A., de Vries, A., Middleton, S.D. et al. (2010). Wong, C.L., Holroyd-Leduc, J., Simel, D.L. and Straus,
Cerebrospinal fluid cortisol levels are higher in S.E. (2010). Does this patient have delirium? Value of
patients with delirium versus controls. BMC Research bedside instruments. JAMA, 304: 779–786.
Notes, 3: 33. Young, J., Murthy, L., Westby, M. et al. (2010). Diagnosis,
Pisani, M.A., McNicoll, L. and Inouye, S.K. (2003). prevention, and management of delirium: Summary
Cognitive impairment in the intensive care unit. Clinics of NICE guidance. British Medical Journal, 341:
in Chest Medicine, 24: 727–737. c3704.
72
Depression with cognitive impairment

SARAH SHIZUKO MORIMOTO, GENEVIEVE S. YUEN, STEPHEN BERES AND


GEORGE S. ALEXOPOULOS

Lesser et al., 1996; Elderkin-Thompson et al., 2004), verbal


72.1 INTRODUCTION fluency and psychomotor speed (Hart et al., 1987; Butters
et al., 2004b), have been reported consistently in late-life
Major depression in the elderly is often accompanied by depression. These impairments, particularly memory
cognitive impairment. Although estimates vary, studies impairments, were attributed to dysfunction in subcortical
show that combined depression and cognitive dysfunction structures related to mood regulation, such as the hippo-
is present in roughly 25% of the subjects (Arve et al., 1999). campus (Hickie et al., 2005). Recent research has focused
In addition, the number of community residents with both on the role of executive functions, such as impaired plan-
depressive symptoms and impaired cognition doubles every ning, organizing, initiating, perseverating, sequencing and
5 years after the age of 70 years. In some cases, the syndromes attentional set-shifting in geriatric depression (Lesser et al.,
of depression and cognitive impairment may be related to the 1996; Alexopoulos et al., 2000; Butters et al., 2000; Murphy
same underlying disorders (e.g. vascular dementia, hypo- and Alexopoulos, 2004; Potter et al., 2004). Executive func-
thyroidism), whereas in other cases, depression and cogni- tion is impaired in depressed older adults relative to healthy
tive impairment may be relatively independent and simply controls, and this impairment persists even when control-
coexist. Differential diagnosis and treatment decisions can ling for processing speed (Dybedal et al., 2013). Further,
be complicated because depressive cognitive changes can be abnormal performance on specific aspects of executive
severe, incipient dementia often can have physical and cog- function predicts both poor and unstable antidepressant
nitive symptoms that overlap with depression and the two response in late-life depression (Kalayam and Alexopoulos,
can coexist (Bayles et al., 1987). The relationships between 1999; Alexopoulos et al., 2000, 2004, 2005; Sneed et al.,
the prominent cerebrovascular changes, other structural 2007; Morimoto et al., 2011), although some disagreement
abnormalities, specific forms of cognitive dysfunction and exists (Butters et al., 2004a). The specifics of this topic are
increased risk for developing dementias in geriatric depres- discussed in Section 72.5.1.
sion have yet to be reconciled. The varied and most current Although focal deficits or even severe global impair-
findings suggest that there are likely multiple pathways to ment are observed in some depressed elderly patients, the
poor cognitive outcomes (Butters et al., 2008). cognitive functioning of others remains intact. Studies of
cognitive response to psychopharmacological treatment
of late-life depression indicate that a substantial number
72.2 COGNITIVE IMPAIRMENT IN of patients continue to experience residual symptoms and
neuropsychological deficits. Executive dysfunction, pro-
NON-DEMENTED DEPRESSED
cessing speed and working memory impairment may per-
PATIENTS sist after remission of geriatric depression (Butters et al.,
2000; Nebes et al., 2000; Aizenstein et al., 2009). Multiple
Neuropsychological impairments spanning many cogni- pathways may lead to depression and cognitive impairment.
tive domains including impairment in executive functions However, clinical as well as structural and functional neu-
(Lockwood et al., 2000, 2002), episodic memory (Beats roimaging studies suggest that depressive symptoms and
et al., 1996; Kramer-Ginsberg et al., 1999; Story et al., 2008), executive impairment originate from related brain dys-
recognition memory, visuospatial skills (Boone et al., 1994; functions (Alexopoulos et al., 2001a; Murphy et al., 2007;

832
Depression with cognitive impairment 833

Alexopoulos et al., 2008; Morimoto et al., 2011, 2012a), cortical glucose metabolism in both anterior and posterior
at least in a subgroup of elderly patients. We described cortical regions in patients with geriatric depression relative
a ‘depression-executive dysfunction (DED) syndrome’ to controls, particularly in areas where there has been cere-
(Alexopoulos, 2001), characterized by symptoms and signs bral atrophy, which may represent a compensatory response
resembling a medial frontal lobe syndrome, including (Smith et al., 2009). In fact, positive correlations between
psychomotor retardation, anhedonia, apathy, mild vegeta- anxiety and depressive symptoms and cortical glucose
tive symptoms and pronounced functional disability dis- metabolism have been observed (Smith et al., 2009).
proportional to the severity of the depressive syndrome
(Alexopoulos et al., 1996, 2000; Lockwood et al., 2002).
72.3 DEPRESSION IN PATIENTS WITH
DEMENTING DISORDERS
72.2.1 NEUROBIOLOGICAL
UNDERPINNINGS OF COGNITIVE
DEFICITS Estimates of the rates of depression in dementing disor-
ders range from 30% to 50% (Taylor et al., 2003). Clinically
The structural and functional abnormalities that contribute significant depressive symptoms occur in 10% to 50% of
to the symptoms of this disorder remain unclear, although Alzheimer’s disease (AD) patients, with estimates of major
several abnormalities have been reported (Alexopoulos depression occurring in 30% of them (Wragg and Jeste,
et al., 2005). Recently, structural and functional neuroim- 1989; Holtzer et al., 2005; Even and Weintraub, 2010;
aging have documented both frontostriatal impairment Enache et al., 2011; Chi et al., 2014). Depression in AD is
and the relationship between frontostriatal impairment notable for increased disturbances in initiation or moti-
and executive dysfunction in geriatric depression (Murphy vation, such as fatigue, psychomotor slowing and apathy
et al., 2007, Aizenstein et al., 2009). (Chemerinski et al., 2001; Janzing et al., 2002; Teng et al.,
Structural abnormalities have been identified in the 2008). Patients with subcortical dementias, including vas-
orbitofrontal cortex (Van Otterloo et al., 2009; Sexton et al., cular dementia (VaD) and Parkinson’s disease (PD), are
2013), particularly in the gyrus rectus bilaterally (Ballmaier more likely to experience depression than patients with
et al., 2004; Yuan et al., 2008), the anterior cingulate (Drevets AD (Sobin and Sackeim, 1997; Enache et al., 2011). Rates
et al., 1997; Ballmaier et al., 2004), the caudate (Krishnan of clinically significant depressive disturbances in PD
et al., 1992; Hannestad et al., 2006), putamen (Husain et al., reach about 35% to 50% (Lagopoulos et al., 2005; Aarsland
1991; Sexton et al., 2013), hippocampus (Sheline et al., 1996; et al., 2012; Marsh, 2013). Depression associated with PD
Lai et al., 2000; Sawyer et al., 2012) and amygdala (Sheline can produce severe negative symptoms and contributes to
et al., 1998; Burke et al., 2011). an increased rate of decline of both cognitive and motor
In addition to grey matter reductions, bilateral white function. Diagnosing depression in PD patients is often
matter hyperintensities are prevalent in geriatric depression challenging due to the overlap of certain somatic and core
(Boone et al., 1992; Steffens et al., 1999; Kumar et al., 2000; depressive symptoms (Lagopoulos et al., 2005; Aarsland
Alexopoulos et al., 2008; Gunning-Dixon et al., 2008) and et al., 2012; Marsh, 2013).
mainly occur in the subcortical structures and their fron- A first episode of depression presenting in late life is,
tal projections (MacFall et al., 2001; Gunning-Dixon et al., sometimes, a prodrome of a dementing disorder. A recent
2008). There is evidence that white matter hyperintensities meta-analysis, which included 23 community-based pro-
disrupt frontostriatal circuits (Hannestad et al., 2006) and spective cohort studies, concluded that late-life depression
have been associated with executive dysfunction (Boone significantly increased the risk of AD incidence 1.65 fold,
et al., 1992; Lesser et al., 1996). Poorer executive function even after controlling all possible cofounders (Chi et al.,
is related to white matter lesions in specific tracts, such as 2014). Another meta-analysis of community-based cohort
the superior longitudinal fasciculus and uncinate fasciculus studies reported individuals with late-life depression had
(Sheline et al., 2008; Mettenburg et al., 2012). Furthermore, a significantly higher risk for incident all-cause dementia,
depressed older adults with impaired response inhibition AD and VaD compared with the elderly controls (Diniz
exhibited reduced white matter integrity, as measured by et al., 2013). Depressed mood has been associated with
fractional anisotropy, in multiple frontostriatal-limbic an increased risk of incident dementia in some samples
regions including areas lateral to the anterior and posterior (Devanand et al., 1996). For example, depressive symptoms
cingulate cortex, and prefrontal, insular and parahippo- predicted cognitive decline of elderly women during a 4-year
campal regions (Murphy et al., 2007). follow-up study (Yaffe et al., 1999). In addition, in patients
Abnormal metabolism has also been noted in limbic with subthreshold cognitive symptoms, those suffering con-
regions, including the amygdala (Wu et al., 1992; Drevets, current depressive symptoms are more likely to progress to
1999; Drevets et al., 2002), the pregenual and subgenual ante- dementia (Ritchie et al., 1999). However, more recent data
rior cingulate (Drevets, 1998), the posterior orbital cortex suggest that there is a significant decline in functioning that
(Drevets et al., 1992), the posterior cingulate and the medial precedes late-onset depressive symptoms in AD and that
cerebellum (Bench et al., 1992). Recent research demonstrates cognition declines subsequently (Holtzer et al., 2005).
834 Dementia

The prevalence of depressive symptoms in AD decreases the hypothalamic–pituitary–adrenal (HPA) axis, which in
over time as patients exhibit fewer affective symptoms, turn can result in hypercortisolaemia (Carroll et al., 1981).
increased agitation and psychomotor slowing. Data suggest Depression disrupts the fast feedback control of corti-
that rates of depression in this population remain stable for sol secretion (Young et al., 1991) at various levels, includ-
roughly the first 3 years of follow-up. Rates appear to drop ing limbic structures such as the hippocampus (Young
in the fourth and fifth year of follow-up by as much as 30% and Vazquez, 1996). The precise relationship between
(Holtzer et al., 2005). hypercortisolaemia, hippocampal volume loss and hip-
The National Institute of Mental Health’s Provisional pocampal inhibitory control is unknown, although ani-
Diagnostic Criteria for Depression of AD were developed mal studies show that neurotoxic tissue damage may be
to promote research on the mechanisms and treatment of one possible mechanism. Another theory postulates that
this disorder (Olin et al., 2002). The criteria require diag- excess and chronic secretion of glucocorticoid hormones
nosis of AD and clinically significant depressive symp- can reduce neurotrophic factors, inhibit neurogenesis and
toms. The criteria specify that the patient must have three render neurones vulnerable to the toxic effect of amyloid.
(reduced from five) or more of the following symptoms These changes may then compound the neuropathologi-
during the same 2-week period (reduced from ‘most of cal changes of AD and accelerate the clinical expression of
the day, nearly every day’) and represent a change from dementia (Zhu et al., 2005).
previous functioning: depressed mood, anhedonia, social One such neurotrophic factor is the brain-derived neu-
isolation, appetite disturbance, sleep disturbance, psy- rotrophic factor (BDNF), which is widely distributed in the
chomotor changes, irritability, fatigue or loss of energy, adult brain and has been implicated in structural abnor-
worthlessness, hopelessness or inappropriate or excessive malities of the human hippocampus (Pezawas et al., 2004;
guilt and recurrent thoughts of death or suicidal ideation, Bueller et al., 2006). Several antidepressants elevate BDNF
plan or attempt. At least one of the symptoms must be in the rat hippocampus. Recent research suggests that
depressed mood or anhedonia. Symptoms due to a medi- BDNF protein is rapidly elevated by antidepressant treat-
cal condition other than AD, or as a direct result of non- ments through post-transcriptional mechanisms (Musazzi
mood-related dementia symptoms (e.g. loss of weight due et al., 2009). Through this action, antidepressants may pre-
to difficulties with food intake), should not be used in vent stress-induced inhibition of neurogenesis and increase
making the diagnosis of depression or AD. These diag- dendritic branching (Duman et al., 1997). Whether this
nostic criteria have less stringent guidelines about the action delays the onset or inhibits the progression of AD
duration and number of symptoms and, therefore, are is unclear; systematic studies have not yet explored this
likely more sensitive than other diagnostic approaches question.
(Engedal et al., 2011).

72.5 COGNITIVE IMPAIRMENT AND


72.4 DEPRESSION AS A RISK FACTOR THE COURSE OF GERIATRIC
FOR DEMENTIA DEPRESSION

Individual studies attempting to link geriatric depression The neuropsychological impairments, seen frequently
with subsequent dementia have mixed results. Although in geriatric depression, span multiple cognitive domains
one recent meta-analysis showed an almost twofold risk (Lockwood et al., 2002). These impairments, particularly
for the development of AD with previous history of depres- memory impairments, were attributed to dysfunction in
sion (Ownby et al., 2006), another found this association subcortical structures related to mood regulation, such as
only when depressive symptoms had appeared within the hippocampus (Hickie et al., 2005).
the 10 years before onset of dementia (Jorm et al., 1991). Recent research has focused on the role of executive
Other individual studies have failed to find this association functions, such as impaired planning, organizing, initiat-
(Ganguli et al., 2006). Recent research suggests that only the ing, perseverating, sequencing and attention set shifting
most severe cases of depression increase risk for later devel- in the clinical course of geriatric depression (Boone et al.,
opment of dementia (Chen et al., 2008). 1994; Lesser et al., 1996; Alexopoulos et al., 2000; Murphy
Some data suggest that the experience of multiple and Alexopoulos, 2004; Potter et al., 2004; Morimoto et al.,
depressive episodes may promote the clinical expression 2011). Abnormal performance on some tests of executive
of dementia in patients with AD. Some volumetric studies function predicts both poor and unstable antidepressant
have found reductions in hippocampal volumes in patients response (Kalayam and Alexopoulos, 1999; Alexopoulos
with recurrent major depression (Sheline et al., 1996, 2003). et al., 2000; Morimoto et al., 2011, 2012a), as well as cur-
Lifetime duration of depression correlated with hippo- rent and future disability (Kiosses et al., 2001; Alexopoulos
campal volume reduction and with behavioural measures et al., 2005). In addition, executive dysfunction predicts
of hippocampal function, such as verbal memory (Sheline suicidality, even after controlling for co-morbid conditions
et al., 1999). Depression leads to an acute dysregulation of (Dombrovski et al., 2008, 2010).
Depression with cognitive impairment 835

The specific structural and functional abnormalities Potter et al., 2004; Sneed et al., 2007, 2008; Story et al., 2008;
that contribute to the symptoms of depression remain to Morimoto et al., 2011 2012). Dysfunction in the neural
be identified, although several abnormalities have been circuits related to remission may produce multiple down-
reported (Alexopoulos et al., 2004). Recently, structural and stream cognitive abnormalities that are similar (i.e. seman-
functional neuroimaging have documented both frontostri- tic strategy deficits vs. response inhibition deficits) but not
atal impairment and a relationship between frontostriatal identical, perhaps because of individual differences in net-
impairment and executive dysfunction in geriatric depres- work abnormalities, premorbid network characteristics and
sion (Murphy et al., 2007; Aizenstein et al., 2009). network interactions with other brain systems necessary for
Structural abnormalities have been identified in the orbi- completion of tasks (Morimoto et al., 2012b).
tofrontal cortex (Van Otterloo et al., 2009), particularly the Decrements in performance on the Initiation/
gyrus rectus bilaterally (Ballmaier et al., 2004; Yuan et al., Perseveration (I/P) domain of the Dementia Rating Scale
2008), the anterior cingulate (Drevets et al., 1997; Ballmaier (DRS) predict poor response to antidepressant treatment
et al., 2004), the caudate head (Krishnan et al., 1992), puta- in geriatric depression (Kalayam and Alexopoulos, 1999;
men (Husain et al., 1991), hippocampus (Sheline et al., 1996; Alexopoulos, 2003b). A meta-analysis examining the rela-
Lai et al., 2000) and amygdala (Sheline et al., 1998). tionship between pretreatment cognitive impairment and
In addition to grey matter reductions, bilateral white response to antidepressants demonstrated that the DRS
matter hyperintensities are prevalent in geriatric depres- I/P domain was a reliable predictor of poor antidepres-
sion (Boone et al., 1992; Kumar et al., 2000; Alexopoulos sant treatment response (McLennan and Mathias, 2010).
et al., 2008; Gunning-Dixon et al., 2008), and mainly occur Approximately 42% of elders with major depression have
in the subcortical structures and their frontal projections abnormal I/P scores as measured by the Mattis Dementia
(MacFall et al., 2001; Gunning-Dixon et al., 2008). White Rating Scale (MDRS) (Alexopoulos et al., 2002). Though not
matter hyperintensitites disrupt frontostriatal circuits a classic test of executive function, the I/P domain of the
(Hannestad et al., 2006) and have been associated with DRS tests multiple coordinated cognitive skills that require
executive dysfunction (Boone et al., 1992; Lesser et al., 1996, executive functioning.
Murphy et al., 2007). Among the functions tested by the I/P, only the Complex
Abnormal metabolism has also been noted in limbic Verbal portion (CV/IP) predicted remission during treat-
regions, including the amygdala (Wu et al., 1992; Drevets, ment with escitalopram (Morimoto et al., 2011). The CV/IP
1999; Drevets et al., 2002), the pregenual and subgenual tests word generation ability by asking patients to name all
anterior cingulate (Drevets, 1998), the posterior orbital cor- of the items they can think of in a supermarket (i.e. seman-
tex (Drevets et al., 1992), the posterior cingulate and the tic fluency). Performance on speeded verbal tasks such as
medial cerebellum (Bench et al., 1992). Recent research has the CV/IP can be improved by the use of strategies, such
shown increased cortical glucose metabolism in both ante- as the organization of responses into super ordinate verbal
rior and posterior cortical regions in patients with geriatric categories (semantic organization), e.g. replying with words
depression relative to controls, particularly in areas where belonging to the same category (fruit), e.g. grapes, strawber-
there has been cerebral atrophy, which may represent a ries, bananas, oranges, rather than separate categories, e.g.
compensatory response (Smith et al., 2009). Cortical glu- bread, bananas, milk. Completing strategic semantic tasks
cose metabolism was correlated with anxiety and depressive such as the CV/IP also requires selection of words from
symptoms (Smith et al., 2009). multiple activated responses and suppression of semanti-
cally or phonemically related but inapplicable words. In
the aforementioned study, the use of semantic strategy,
72.5.1 NEUROCOGNITIVE DEFICITS explained performance differences between remitters and
INFLUENCING TREATMENT non-remitters (Morimoto et al., 2011).
RESPONSE OF GERIATRIC These findings were followed by a second study demon-
DEPRESSION strating the use of the same executive function; semantic
organizational strategy, explained both verbal memory
Abnormal performance on select executive function performance and remission rates of geriatric depression
tests predicted poor and/or slow antidepressant response (Morimoto et al., 2012a). These studies defined a single exec-
(Alexopoulos et al., 2002b; Sneed et al., 2007, 2008; utive function that is predictive of remission with antide-
Morimoto et al., 2011, 2012a) and a higher level of func- pressant drug treatment, regardless of the task by which the
tional disability (Kiosses et al., 2001) in geriatric depression. function is elicited. If these findings are replicated, testing
These include processing speed, perseveration, seman- semantic strategy could be a useful tool added to the clini-
tic strategy and response inhibition, which were associ- cal assessment of depressive symptoms. Results could help
ated with both poor and unstable antidepressant response improve the clinician’s ability to identify patients at risk for
and low remission rate in non-demented elderly patients poor antidepressant response (Morimoto et al., 2011).
with major depression treated with ‘adequate’ dosages of These neuropsychological findings parallel structural
various antidepressants (Simpson et al., 1998; Kalayam and functional neuroanatomical changes associated both
and Alexopoulos, 1999; Alexopoulos et al., 2000, 2004; with cognitive control dysfunction and with poor response
836 Dementia

of late-life depression to antidepressants. Structural and they often have a later age of onset of illness (Alexopoulos
functional imaging findings, including white matter hyper- et al., 1993). When compared to AD patients with concomi-
intensities (Gunning-Dixon et al., 2010), microstructural tant depression, pseudodementia patients have more psy-
white matter changes (Alexopoulos et al., 2008), low volume chic and somatic anxiety, early morning awakening and loss
of the anterior cingulate (Gunning et al., 2009) and reduced of libido (Reynolds et al., 1986).
resting functional connectivity of the cognitive control Neuropsychological assessment is generally considered
network (Alexopoulos et al., 2012), are observed in late- the gold standard in differentiating between depression
life depression. Taken together, these findings lend support and the early stages of dementia. Decrements in memory,
to the hypothesis that depression with cognitive control attention, visuospatial ability, processing speed and execu-
dysfunction is a distinct syndrome of late-life depression tive functioning have been reported in pseudodemen-
(Alexopoulos, 2001) with poor antidepressant response. tia. Although these deficits are also seen in AD, cognitive
impairments seen in early AD are more severe in nearly
every cognitive domain. For example, depressed patients
perform better than AD patients on measures of initial
72.6 PSEUDODEMENTIA learning and recall as well as delayed recall (Kang et al.,
2014). In addition, although memory may be impaired in
The concept of reversible dementia was introduced in the both conditions, AD patients will often exhibit ‘rapid for-
mid-nineteenth century (Berrios, 1985; Emery, 1988) and getting’ (Gray et al., 1986) pointing to deficits in memory
became the focus of clinical attention in the early 1960s systems, as well as difficulty with recognition memory. AD
when the term ‘pseudodementia’ was used to describe a patients also produce more ‘false-positive’ errors or ‘intru-
broad range of reversible cognitive impairments associated sions’, whereas depressed elderly patients tend to produce
with psychiatric syndromes (Kiloh, 1961). The current con- more false negatives (Whitehead, 1973; Kang et al., 2014).
cept of pseudodementia is that of an initially reversible cog- Patients with true AD tend to present with additional spe-
nitive impairment that both occurs in the context of diverse cific impairments, such as impaired temporal orientation,
psychiatric disorders and influences their course. abstraction, calculation, visual recognition, visuoconstruc-
The clinical presentation of depression with cognitive tion and language functions (Chaves and Izquierdo, 1992).
impairment is heterogeneous; the signs and symptoms are As a rule, the symptoms of depression also precede those of
principally influenced by the patients’ age and underly- cognitive dysfunction in patients with true pseudodemen-
ing psychiatric disorders (Kiloh, 1961; Wells, 1979; Rabins tia, making it important to take a thorough history of the
et al., 1984; Alexopoulos, 1990; Emery and Oxman, 1992). symptoms.
In a non-geriatric series, pseudodementia was reported Studies of pseudodementia in depressed geriatric
in patients with Ganser syndrome, personality disorders, patients challenge the concept of pseudodementia as a truly
melancholic depression, hypomania, atypical psychosis, reversible dementia syndrome. Notably, although cognitive
paraphrenia, catatonia, depersonalization and malinger- impairment associated with depressive pseudodementia
ing (Kiloh, 1961; Wells, 1971). Non-geriatric patients with may diminish upon remission of depression, follow-up stud-
pseudodementia in this sample had a history of prior psy- ies show that a large percentage of these patients progress
chiatric disorders, a recent and abrupt onset of current ill- into irreversible dementia within 2–3 years of onset. This
ness, complained about their cognitive loss and experienced supports the claim that reversible cognitive impairment
distress and were able both to identify precisely the onset associated with pseudodementia is more likely the result of
and describe in detail the course of their illness (Kiloh, a preclinical or early-stage dementing disorder exacerbated
1961; Wells, 1979). On cognitive tests, these patients often by concomitant depression (Alexopoulos, 2003).
said they did not know the correct answers even though the
tasks were within their abilities. Performance on tasks of
similar difficulty was markedly variable and accompanied
72.7 DIAGNOSTIC ASSESSMENT AND
by an overdramatization of failures.
As many younger patients with pseudodementia have
TREATMENT PLANNING
psychiatric syndromes other than depression, their clinical
presentation is dissimilar from that of geriatric patients with Identifying and characterizing the cognitive impairment of
pseudodementia in whom depression is the most common depressed elderly patients has important clinical implica-
diagnosis. Depressed older patients with pseudodemen- tions. Depressed elders often develop delirium in response
tia usually have a severe depression syndrome, but a mild to drug side effects, dehydration, infections and other fac-
dementia. The clinical profile of major depression accom- tors. Therefore, cognitive examination should focus on
panying pseudodementia of older adults is characterized by manifestations of delirium, including inattention, fluctuat-
motor retardation, depressive delusions, hopelessness and ing state of consciousness and sleep–wake disturbances. Of
helplessness (Alexopoulos and Abrams, 1991). Geriatric course, making a diagnosis of delirium does not exclude a
inpatients with major depression and pseudodementia dif- diagnosis of dementia. As dementing disorders predispose
fer from those with uncomplicated major depression in that to delirium, delirious patients should be re-examined for an
Depression with cognitive impairment 837

underlying dementia after the delirium resolves. Identifying patients with the DED syndrome of late life require carefully
treatable causes of dementia (e.g. dementia due to drug planned psychopharmacological treatment and vigilant fol-
intoxication, organ failure, endocrinopathies, B12 deficiency, low-up, since they are at high risk for relapse or recurrence
normal pressure hydrocephalus and space-occupying (Alexopoulos et al., 2000). Non-pharmacological interven-
lesions) is important, as treatment of these conditions may tions should be considered, particularly in medication non-
reverse or arrest the progress of dementia. responders. These are further discussed in Section 72.7.2.
The identification of depression or cognitive impairment Evaluation of depressive syndromes in cognitively
in the elderly should lead to an examination for identifying impaired patients is complicated by the symptom overlap
possibly undetected vascular disease and to consideration with dementia, the instability of depressive manifestations
of factors that might predispose to vascular disease, because over time and the poor ability of elderly patients to report
both of these conditions are associated with cerebrovascu- their symptoms. If criteria for one of the depressive syn-
lar disease (Thomas et al., 2004; Roman, 2006; Kelly and dromes are met, an antidepressant treatment trial should
Alexopoulos, 2009). Most brain infarcts do not manifest be offered. The Expert Consensus Guideline recommends
with neurological signs (Fried et al., 1991; Longstreth et al., antidepressant drug therapy combined with psychosocial
1998; Vermeer et al., 2002), so depression and/or cognitive intervention as the treatment of choice for geriatric depres-
impairment can be the first sign of cerebrovascular disease. sion (Alexopoulos et al., 2001). Variability in the course of
The relationship between depression and cardiovascular elders with depression and cognitive impairment suggests
disease appears to be bidirectional (Thomas et al., 2004). the need for careful follow-up. About 40% of these patients
Therefore, care should be coordinated to ensure that both are expected to receive the diagnosis of dementia within 2
psychiatric and cardiovascular health issues are addressed years after the diagnosis of pseudodementia. Prompt iden-
(Alexopoulos et al., 2002). Since many medications, includ- tification and treatment of the dementing disorder may
ing psychotropic medications, increase cardiovascular risk increase the time during which the patient can function
factors, including hyperlipidaemia, hyperglycaemia, hyper- independently.
tension and obesity, a careful overview of the patient’s regi-
men is needed (Kelly and Alexopoulos, 2009). 72.7.1 ASSESSMENT AND DIAGNOSIS:
When dementia is identified, the clinical character- NEUROPSYCHOLOGICAL AND
ization of the dementia syndrome can guide treatment. CLINICAL EXAMINATION
Among the dementia syndromes, subcortical dementia
is the syndrome most likely to be complicated by depres- Tests of executive function should be part of the initial
sion. Subcortical dementia is characterized by significant assessment of late-life depression. Clinicians can employ
memory impairment, executive dysfunction and psycho- bedside screening tests for executive dysfunction, such as
motor retardation. Disorders causing subcortical dementias a verbal fluency task (e.g. ask patients to name as many
include mixed AD and VaD, VaD, PD and dementia with items as possible that can be purchased in a supermarket,
Lewy bodies (DLB). These disorders require specific treat- in 1 minute) or the Stroop Colour Word test of cognitive
ments, e.g. VaD is often treated with aspirin, statins and inhibition. Abnormal performance on each of these tests
management of hypertension, PD with dopamine-acting may predict poor antidepressant response, relapse and
agents, and DLB with cholinesterase inhibitors. Unlike greater functional disability (Baldwin et al., 2004; Potter et
subcortical dementias, cortical dementias manifest broader al., 2004; Alexopoulos et al., 2005; Kiosses and Alexopoulos,
impairment of cognitive functions, including memory 2005; Sneed et al., 2007; Morimoto et al., 2011).
impairment, apraxia, aphasia with paraphasic errors and A more comprehensive neuropsychological assessment
graphomotor construction problems. The most common can aid the differential diagnosis of patients likely to expe-
cause of cortical dementia is AD for which cholinester- rience cognitive decline from depressed patients who are
ase inhibitors and memantine are the available treatments likely to remain cognitively stable. Rushing et al. found that
(see Chapter 52). Frontal lobe syndromes are characterized out of a battery of multiple cognitive measures, impaired
by rather mild memory impairment and pronounced per- recall of verbal contextual information (short stories) pre-
sonality changes, apathy, socially inappropriate behaviour dicted conversion to dementia in a study of older adults with
and disinhibition, often resulting in irritability. Frontal lobe major depression who developed AD dementia 13 years
syndromes may be due to frontotemporal dementia, head later (Rushing et al., 2014).
trauma or stroke and no specific treatments are available. Initial evaluation of the depressed older adult with
Identification of executive dysfunction is important even cognitive impairment should also include a physical and
in non-demented depressed elderly patients. Depressed neurological examination with laboratory tests (including
patients with psychomotor retardation, reduced interest in comprehensive chemistry screen, complete blood count,
activities, suspiciousness and disability are likely to have thyroid stimulating hormone levels, electrocardiogram,
executive dysfunction (Alexopoulos et al., 2002). Depression serum B12 levels, folate levels, urinalysis and drug blood
with executive dysfunction may have a poor and unstable levels) to rule out medical causes of cognitive impairment.
response to antidepressants (Kalayam and Alexopoulos, Identifying treatable causes of cognitive dysfunction (e.g.
1999; Alexopoulos et al., 2000, 2005). For this reason, drug or medication intoxication/withdrawal, organ failure,
838 Dementia

endocrinopathies, B12 deficiency, normal pressure hydro- depressed (Culang et al., 2009). Second-line psychotropic
cephalus and space-occupying lesions) is important, as options may include bupropion, mirtazapine and tricyclic
treatment of these conditions may reverse or arrest progres- antidepressants (TCAs). TCAs, however, do not improve
sion to dementia. cognitive function in depressed older adults. In fact, nor-
Coexisting depression and cognitive impairment may triptyline compromised verbal learning performance com-
have a common aetiology, most frequently vascular disease. pared to placebo (Meyers et al., 1991).
Therefore, physicians should examine for previously unde- Antidepressant treatment improves symptoms in some
tected cardiovascular disease and risk factors that predis- but not all patients, and those who benefit may not return to
pose to cardiovascular disease. The ‘vascular depression’ the cognitive and functional level of non-depressed elderly.
hypothesis postulates that cerebrovascular disease can pre- (Butters et al., 2000; Frasch et al., 2000; Beekman et al.,
dispose, precipitate or perpetuate a depressive syndrome in 2002; Doraiswamy et al., 2003; Nebes et al., 2003, Murphy
older adults. Patients with vascular depression often have and Alexopoulos, 2004; Bhalla et al., 2006; Lee et al., 2007).
impairment of executive function, psychomotor slowing, Therefore, separate treatment for cognitive impairment may
apathy, impaired insight, disability disproportional to the be necessary to improve outcomes in late-life depression.
severity of their depression and poor response to antide- Antidepressant augmentation with existing cognitive
pressants (Alexopoulos et al., 1997). Neuroimaging in these enhancing drugs has received little research attention.
cases will often reveal white matter hyperintensities, likely Open-label and placebo-controlled trials of the combination
caused by cerebrovascular lesions (Taylor et al., 2013). of citalopram with the psychostimulant methylphenidate,
Management of depression with cognitive impairment showed improved depressive symptoms in older depressed
should therefore include optimization of vascular risk fac- patients, but these studies did not report effects on cogni-
tors by carefully controlling hypertension, hypercholes- tion (Lavretsky and Kumar, 2001; Lavretsky et al., 2003,
terolaemia, hyperglycaemia and obesity as well as using 2006). A small placebo-controlled study found no effect of
aspirin or other anticoagulants in patients with atrial fibril- memantine, on mood or functional independence (Lenze
lation (Alexopoulos et al., 2002; Avari et al., 2014). One et al., 2012) and augmentation of an antidepressant with
study suggested that augmentation of fluoxetine with the donepezil modestly improved cognition but increased the
calcium channel blocker nimodipine results in more effec- risk of depression recurrence (Reynolds et al., 2011).
tive treatment of vascular depression and lower recurrence The cholinergic system is well established as important
rates (Taragano et al., 2005). to cognition. Cholinesterase inhibitors increase acetylcho-
line, while negatively affecting mood (Reynolds et al., 2011),
72.7.2 TREATMENTS: PHARMACOLOGIC selective stimulation of nicotinic acetylcholine receptors
AND TARGETED PSYCHOTHERAPY may benefit cognition without worsening mood (Zurkovsky
et al., 2013). As reviewed by Newhouse and colleagues
If criteria for depression are met, an antidepressant treat- (Zurkovsky et al., 2013), some studies of nicotinic receptor
ment trial should be offered. Beyond the benefits of alleviat- agonists show promise in improving cognitive performance
ing the suffering and complications of depression, remission in patients with AD dementia or mild cognitive impairment
of the depressive syndrome can increase the clinician’s (MCI). However, this has not yet been demonstrated in a
ability to evaluate the severity of the remaining cognitive population of depressed elderly with cognitive impairment.
impairment and plan for further treatment and follow-up. Given the poor and unstable response to antidepres-
The Expert Consensus Guideline recommends antidepres- sants that frequently characterizes depression with execu-
sant drug therapy combined with a psychosocial inter- tive dysfunction (Alexopoulos et al., 2000, 2005), adjunctive
vention as the treatment of choice for geriatric depression non-pharmacological interventions should be considered
(Alexopoulos et al., 2001). (Alexopoulos et al., 2001). Targeted therapies can improve
Several antidepressants are available for the treat- the behavioural deficits of depressed elders with cogni-
ment of unipolar, non-psychotic, late-life major depres- tive impairments that are associated with poor response to
sion. Selective serotonin reuptake inhibitors (SSRIs) or antidepressant. Problem-solving therapy, as modified by
serotonin-norepinephrine reuptake inhibitors (SNRIs) are Alexopoulos and Arean to address the behavioural deficits
first-line agents (Alexopoulos et al., 2001). Some SSRIs can of depression with executive dysfunction, is efficacious in
improve cognitive function but mainly in patients whose reducing depression and disability in cognitively unim-
depressive symptoms respond to the SSRI antidepressant. paired depressed older adults and in older adults with cog-
Treatment with sertraline in depressed elders with cogni- nitive impairment (Alexopoulos et al., 2003, 2008; Arean
tive impairment, improved performance on tests of atten- et al., 2010; Kiosses et al., 2011b). Other targeted therapies
tion, episodic memory and executive function, but again for depressed elderly patients incorporate modification of
only in patients whose depression responded to this antide- the patient’s environment or ‘ecosystem’, which includes the
pressant (Devanand et al., 2003). Treatment with citalopram patient, caregiver and home environment. We have devel-
improved psychomotor speed and visuos

You might also like