You are on page 1of 418

https://t.

me/MBS_MedicalBooksStore
Molecular Hematology
Molecular
Hematology
FOURTH EDITION

Edited by
Drew Provan MD FRCP FRCPath
Emeritus Reader in Autoimmune Haematology
Department of Haematology
Barts and The London School of Medicine and Dentistry
Queen Mary University of London, UK

John G. Gribben MD DSc FRCP FRCPath FMedSci


Professor of Medical Oncology
Barts Cancer Institute
Barts and The London School of Medicine and Dentistry
Queen Mary University of London, UK
This edition first published 2020
© 2020 by John Wiley & Sons
Edition History [3rd edition, 2010]
All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or
transmitted, in any form or by any means, electronic, mechanical, photocopying, recording or
otherwise, except as permitted by law. Advice on how to obtain permission to reuse material from this
title is available at http://www.wiley.com/go/permissions.
The right of Drew Provan to be identified as the author of editorial in this work has been asserted in
accordance with law.
Registered Office(s)
John Wiley & Sons, Inc., 111 River Street, Hoboken, NJ 07030, USA
John Wiley & Sons Ltd, The Atrium, Southern Gate, Chichester, West Sussex, PO19 8SQ, UK
Editorial Office
9600 Garsington Road, Oxford, OX4 2DQ, UK
For details of our global editorial offices, customer services, and more information about Wiley
products visit us at www.wiley.com.
Wiley also publishes its books in a variety of electronic formats and by print-on-demand. Some content
that appears in standard print versions of this book may not be available in other formats.
Limit of Liability/Disclaimer of Warranty
The contents of this work are intended to further general scientific research, understanding, and
discussion only and are not intended and should not be relied upon as recommending or promoting
scientific method, diagnosis, or treatment by physicians for any particular patient. In view of ongoing
research, equipment modifications, changes in governmental regulations, and the constant flow of
information relating to the use of medicines, equipment, and devices, the reader is urged to review and
evaluate the information provided in the package insert or instructions for each medicine, equipment,
or device for, among other things, any changes in the instructions or indication of usage and for added
warnings and precautions. While the publisher and authors have used their best efforts in preparing this
work, they make no representations or warranties with respect to the accuracy or completeness of the
contents of this work and specifically disclaim all warranties, including without limitation any implied
warranties of merchantability or fitness for a particular purpose. No warranty may be created or
extended by sales representatives, written sales materials or promotional statements for this work. The
fact that an organization, website, or product is referred to in this work as a citation and/or potential
source of further information does not mean that the publisher and authors endorse the information or
services the organization, website, or product may provide or recommendations it may make. This work
is sold with the understanding that the publisher is not engaged in rendering professional services. The
advice and strategies contained herein may not be suitable for your situation. You should consult with a
specialist where appropriate. Further, readers should be aware that websites listed in this work may have
changed or disappeared between when this work was written and when it is read. Neither the publisher
nor authors shall be liable for any loss of profit or any other commercial damages, including but not
limited to special, incidental, consequential, or other damages.
Library of Congress Cataloging-in-Publication Data
Names: Provan, Drew, editor. | Gribben, John editor.
Title: Molecular hematology / edited by Drew Provan, John G. Gribben.
Description: Fourth edition. | Hoboken, NJ : Wiley-Blackwell, 2020. | Includes bibliographical
references and index.
Identifiers: LCCN 2019034376 (print) | LCCN 2019034377 (ebook) | ISBN 9781119252870 (hardback) |
ISBN 9781119252955 (adobe pdf) | ISBN 9781119252931 (epub)
Subjects: MESH: Hematologic Diseases | Molecular Biology–methods
Classification: LCC RC636 (print) | LCC RC636 (ebook) | NLM WH 120 | DDC 616.1/5–dc23
LC record available at https://lccn.loc.gov/2019034376
LC ebook record available at https://lccn.loc.gov/2019034377
Cover Design: Wiley
Cover Image: © JUAN GAERTNER/SCIENCE PHOTO LIBRARY/Getty Images
Set in 10/12pt MinionPro by Aptara Inc., New Delhi, India

10 9 8 7 6 5 4 3 2 1
Dedication

We would like to dedicate this book to two people: our dear friend and colleague, Professor Sir David Weatherall, who sadly
passed away on 8 December 2018. He was truly a pioneer of molecular biology and was the first physician scientist to use
molecular techniques to study hematological disease. We will all miss him very much.
In addition, we would like to dedicate the book to Val Provan. Always in our thoughts and much missed.
Contents

Contributors, ix 13 The molecular basis of iron metabolism, 161


Preface to the fourth edition, xiii Nancy C. Andrews & Tomas Ganz

Further reading, xv 14 Hemoglobinopathies due to structural mutations, 173


D. Mark Layton & Steven Okoli
Acknowledgments, xvi
15 Molecular pathogenesis of malaria, 193
David J. Roberts, Arnab Pain & Chetan E. Chitnis
1 Beginnings: the molecular pathology of hemoglobin, 1
16 Molecular coagulation and thrombophilia, 207
David Weatherall
Björn Dahlbäck & Andreas Hillarp
2 Stem cells, 21
17 The molecular basis of hemophilia, 221
David T. Scadden
Daniel P. Hart & Paul L.F. Giangrande
3 The genetics of acute myeloid leukemias, 37
18 The molecular basis of von Willebrand disease, 235
Amy M. Trottier & Carolyn J. Owen
Luciano Baronciani
4 Molecular diagnostics and risk assessment in
19 Platelet disorders, 251
myeloid malignancies, 49
Kenneth J. Clemetson
Christian Scharenberg & Torsten Haferlach
20 The molecular basis of blood cell alloantigens, 267
5 Molecular basis of acute lymphoblastic leukemia, 59
Cristina Navarrete, Louise Tilley, Winnie Chong
Bela Patel & Fiona Fernando
& Colin J. Brown
6 Chronic myeloid leukemia, 71
21 Functions of blood group antigens, 285
Hagop Kantarjian, Jorge Cortes, Elias Jabbour &
Jonathan S. Stamler & Marilyn J. Telen
Susan O’Brien
22 Autoimmune hematological disorders, 297
7 Myeloproliferative neoplasms, 87
Drew Provan & John W. Semple
Jyoti Nangalia, Anthony J. Bench, Anthony R.
Green & Anna L. Godfrey 23 Molecular therapeutics in hematology: gene
therapy, 319
8 Lymphoma genetics, 101
William M. McKillop & Jeffrey A. Medin
Jennifer L. Crombie, Anthony Letai & John G. Gribben
24 Pharmacogenomics, 339
9 The molecular biology of chronic lymphocytic
Leo Kager & William E. Evans
leukemia, 111
John G. Gribben 25 History and development of molecular biology, 353
Paul Moss
10 The molecular biology of multiple myeloma, 121
Wee Joo Chng & P. Leif Bergsagel 26 Cancer stem cells, 363
Sara Ali & Dominique Bonnet
11 The molecular basis of bone marrow failure
syndromes and red cell enzymopathies, 131 27 Molecular basis of transplantation, 373
Deena Iskander, Lucio Luzzatto & Anastasios Francesco Dazzi & Antonio Galleu
Karadimitris
12 Anemia of chronic disease, 155 Index, 389
Tomas Ganz

vii
Contributors

Sara Ali MD Kenneth J. Clemetson PhD, ScD, CChem, FRSC


Haematopoietic Stem Cell Laboratory, The Francis Crick Department of Haematology, Inselspital, University of Berne,
Institute, London, UK Berne, Switzerland

Nancy C. Andrews MD, PhD Jorge Cortes MD


Duke University School of Medicine, Durham, NC, USA Department of Leukemia, University of Texas MD Anderson
Cancer Center, Houston, TX, USA
Luciano Baronciani PhD
Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlin- Jennifer L. Crombie MD
ico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Department of Medical Oncology, Dana-Farber Cancer
Center, Milan, Italy Institute, Harvard Medical School, Boston, MA, USA

Anthony J. Bench MA, PhD Björn Dahlbäck MD, PhD


Laboratory Medicine, NHS Lothian, Edinburgh, UK Department of Translational Medicine, Section of Clinical
Chemistry, Lund University, University Hospital, Malmö,
P. Leif Bergsagel MD Sweden
Division of Hematology-Oncology, Comprehensive Cancer
Center, Mayo Clinic Arizona, Scottsdale, AZ, USA Francesco Dazzi MD, PhD
School of Cancer & Pharmaceutical Sciences, King’s College
Dominique Bonnet PhD London; King’s Health Partners Cancer Research UK Centre,
Haematopoietic Stem Cell Laboratory, The Francis Crick London, UK
Institute, London, UK
William E. Evans PharmD
Colin J. Brown PhD, FRCPath St Jude Children’s Research Hospital, Memphis, TN, USA
Histocompatibility and Immunogenetics Laboratory, NHS
Blood and Transplant; Faculty of Life Sciences & Medicine,
Fiona Fernando MD
King’s College London, London, UK
Centre of Haemato-Oncology, Barts Cancer Institute, Queen
Mary University of London, London, UK
Chetan E. Chitnis MSc, MA, PhD
Malaria Group, Pasteur Institute, Paris, France
Antonio Galleu MD, PhD
School of Cancer & Pharmaceutical Sciences, King’s College
Wee Joo Chng MB ChB, MRCP, FRCPath London; King’s Health Partners Cancer Research UK Centre,
National University Cancer Institute, National Univer- London, UK
sity Health System of Singapore; University of Singapore,
National University Hospital, Singapore
Tomas Ganz PhD, MD
Department of Medicine, David Geffen School of Medicine
Winnie Chong PhD
at UCLA, Los Angeles, CA, USA
Histocompatibility and Immunogenetics Service Develop-
ment Laboratory, NHS Blood and Transplant, London, UK

ix
x Contributors

Paul L.F. Giangrande MD, FRCP, FRCPath, FRCPCH Anthony Letai MD, PhD
Formerly of Oxford Haemophilia and Thrombosis Centre, Department of Medical Oncology, Dana-Farber Cancer
Churchill Hospital, Oxford, UK Institute, Harvard Medical School, Boston, MA, USA

Anna L. Godfrey PhD, MRCP, FRCPath Lucio Luzzatto MD


Department of Haematology, Cambridge University Hospi- Department of Haematology and Blood Transfusion,
tals NHS Foundation Trust, Cambridge, UK Muhimbili University College of Health Sciences, Dar-es-
Salaam, Tanzania
Anthony R. Green PhD, FRCP, FRCPath, FMedSci
Department of Haematology, Cambridge Institute for Medi- William M. McKillop PhD
cal Research; Wellcome Medical Research Council Stem Cell Department of Pediatrics, Medical College of Wisconsin,
Institute, Cambridge, UK Milwaukee, WI, USA

John G. Gribben MD, DSc, FRCP, FRCPath, FMedSci Jeffrey A. Medin PhD
Barts Cancer Institute, Barts and The London School of Departments of Pediatrics and Biochemistry, Medical Col-
Medicine and Dentistry, Queen Mary University of London, lege of Wisconsin, Milwaukee, WI, USA
London, UK
Paul Moss MD, PhD
Torsten Haferlach MD School of Cancer Sciences, University of Birmingham, Birm-
MLL Munich Leukemia Laboratory, Munich, Germany ingham, UK
Daniel P. Hart FRCP, FRCPath, PhD
Jyoti Nangalia PhD, MRCP, FRCPath
The Royal London Hospital Haemophilia Centre, Barts and
Welcome Sanger Institute, Hinxton; Department of Haema-
The London School of Medicine and Dentistry, Queen Mary
tology, University of Cambridge; Wellcome-Medical
University of London, London, UK
Research Council Cambridge Stem Cell Institute, Cam-
bridge, UK
Andreas Hillarp PhD
Department of Clinical Chemistry and Transfusion
Cristina Navarrete PhD, FRCPath
Medicine, Halland County Hospital, Halmstad, Sweden
Histocompatibility and Immunogenetics Service Develop-
ment Department, NHS Blood and Transplant; Department
Deena Iskander MD, PhD, MRCP
of Immunology and Molecular Pathology, University College
Centre for Haematology, Imperial College London, Ham-
London, London, UK
mersmith Hospital, London, UK

Elias Jabbour MD Susan O’Brien MD


Department of Leukemia, University of Texas MD Anderson Department of Leukemia, University of Texas MD Anderson
Cancer Center, Houston, TX, USA Cancer Center, Houston, TX, USA

Leo Kager MD Steven Okoli MBChB, FRCP, FRCPath


Department of Pediatrics, St. Anna Children’s Hospital, Center for Hematology, Imperial College London, London,
Medical University Vienna, Austria UK

Hagop Kantarjian MD Carolyn J. Owen MD, MDres(UK), FRCPC


Department of Leukemia, University of Texas MD Anderson Division of Hematology and Hematological Malignancies,
Cancer Center, Houston, TX, USA University of Calgary, Foothills Medical Centre, Calgary,
Canada
Anastasios Karadimitris PhD, MRCP, FRCPath
Department of Haematology and Blood Transfusion, Arnab Pain PhD
Muhimbili University College of Health Sciences, Dar-es- Biological and Environmental Sciences and Engineering
Salaam, Tanzania (BESE) Division, King Abdullah University of Science and
Technology, Jeddah, Saudi Arabia; Nuffield Division of Clin-
D. Mark Layton MB BS, FRCP, FRCPCH ical Laboratory Sciences (NDCLS), The John Radcliffe Hos-
Center for Hematology, Imperial College London, London, pital, University of Oxford, Headington, Oxford, UK
UK
Contributors xi

Bela Patel MD, FRCPath, MD(res) Jonathan S. Stamler MD


Centre of Haemato-Oncology, Barts Cancer Institute, Queen Harrington Discovery Institute and Institute of Transfor-
Mary University of London, London, UK mative Molecular Medicine, University Hospitals Cleveland
Medical Center and Case Western Reserve University, Cleve-
Drew Provan MD, FRCP, FRCPath land, OH, USA
Blizard Institute, Barts and The London School of Medicine
and Dentistry, Queen Mary University of London, London, Marilyn J. Telen MD
UK Department of Medicine, Division of Hematology, Duke
University Medical Center, Durham, NC, USA
David J. Roberts DPhil, MRCP, FRCPath
National Health Service Blood and Transplant (Oxford), The Louise Tilley PhD
John Radcliffe Hospital, Oxford, UK International Blood Group Reference Laboratory, NHS
Blood and Transplant, Bristol, UK
David T. Scadden MD
Department of Stem Cell and Regenerative Biology, Harvard Amy M. Trottier MSc, MD, FRCPC
Stem Cell Institute, Harvard University; Center for Regenera- Division of Hematology and Hematological Malignancies,
tive Medicine, Massachusetts General Hospital, Boston, MA, University of Calgary, Foothills Medical Centre, Calgary,
USA Canada

Christian Scharenberg MD, PhD David Weatherall MD, FRCP, FRS


Department of Hematology, Skaraborgs Hospital, Skövde; Formerly of Weatherall Institute of Molecular Medicine, The
Department of Cell and Molecular Biology, Karolinska Insti- John Radcliffe Hospital, Oxford, UK
tute, Stockholm, Sweden

John W. Semple PhD


Division of Hematology and Transfusion Medicine, Lund
University, Lund, Sweden
Preface to the fourth edition

Hematology is a fast-moving discipline with innovation both ative neoplasms, myeloma, and myelodysplastic syndromes.
diagnostically and therapeutically. In the 19 years since the The huge array of biological markers makes stratification and
first edition of Molecular Hematology was published, many treatment much more sophisticated than ever before. All the
advances have been made. Molecular techniques have helped chapters dealing with malignant blood diseases have been
explain the basis of many diseases, starting initially with red thoroughly revised and brought up to date.
cell disorders and hemostasis. However, thanks to the use of However, despite the growing complexity in terms of
molecular biology we can now diagnose and stratify patients pathogenesis, diagnosis, and management of patients with
with diseases such as leukemia, myeloma, myeloproliferative blood diseases, the ethos of the book remains the same –
neoplasms, and others. Such advances in technology have not namely, to provide a succinct account of the molecular biol-
only helped explain the underlying basis of the diseases, but ogy of hematological disease written at a level where it should
have also provided targets for treatment. be of benefit to both the seasoned molecular biologist and
The world of red cells started the whole specialty of molec- the practicing clinician alike. We have retained the original
ular medicine and, using molecular biology techniques, structure for the chapters, high-quality artwork, and Further
many of the phenotypic features of red cell disorders have Reading sections in order to make the book visually appealing
been explained. This is discussed eloquently by the late Pro- and relevant to modern hematology practice.
fessor Sir David Weatherall at the beginning of the book. We very much hope you enjoy this edition and, as always,
Other non-malignant areas which have been updated include we welcome any comments or suggestions from readers,
the Functions of Blood Group Antigens, von Willebrand Dis- which we will attempt to incorporate into the next edition.
ease, and Platelet Disorders.
Undoubtedly, hemato-oncology has seen the biggest Drew Provan
explosion in terms of understanding the molecular basis of John Gribben
diseases such as leukemias, lymphomas, the myeloprolifer-

xiii
Further reading

Anderson, K.C. and Ness, P.M. (eds.) (2000). Scientific Basis of Transfu- Mullis, K.B. (1990). The unusual origin of the polymerase chain reac-
sion Medicine: Implications for Clinical Practice, 2e. Philadelphia, PA: tion. Scientific American 262: 56–65.
WB Saunders. Roitt, I. (2001). Roitt’s Essential Immunology, 10e. Oxford: Blackwell Sci-
Beutler, E. and Lichtman, M.A. (eds.). Williams’ Hematology, 6e. New ence.
York: McGraw-Hill. Stamatoyannopoulos, G., Nienhuis, A.W., Majerus, P.W., and Varmus,
Cooper, G.M. (1997). The Cell: A Molecular Approach. Washington, DC: H. (eds.) (2000). The Molecular Basise of Blood Diseases, 2e. Philadel-
ASM Press. phia, PA: W.B. Saunders.
Cox, T.M. and Sinclair, J. (1997). Molecular Biology in Medicine. Oxford: Watson, J.D., Gilman, M., Witkowski, J., and Zoller, M. (eds.) (1992).
Blackwell Science. Recombinant DNA, 2e. New York: Scientific American Books.
Jameson, J.L. (ed.) (1998). Principles of Molecular Medicine. New York:
Humana Press.

xv
Acknowledgments

We would like to express thanks to Claire Bonnett, Publisher, and Deirdre Barry, Senior Editorial Assistant, for their help with
this work.

xvi
Chapter 1 Beginnings: the molecular
pathology of hemoglobin
David Weatherall
Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK

Historical background, 1 Molecular aspects of the high frequency of the hemoglobin variants, 17
The structure, genetic control, and synthesis of normal hemoglobin, 2 Molecular aspects of the prevention and management of the
The molecular pathology of hemoglobin, 6 hemoglobin disorders, 18
Genotype–phenotype relationships in the thalassemias, 12 Postscript, 18
Structural hemoglobin variants, 16 Further reading, 18

within bacteria. The steady improvement in the proper-


Historical background ties of cloning vectors made it possible to generate libraries
of human DNA growing in bacterial cultures. Ingenious
Linus Pauling first used the term “molecular disease” in 1949,
approaches were developed to scan the libraries to detect
after the discovery that the structure of sickle cell hemoglobin
genes of interest; once pinpointed, the appropriate bacte-
differed from that of normal hemoglobin. Indeed, it was this
rial colonies could be grown to generate larger quantities of
seminal observation that led to the concept of molecular
DNA carrying a particular gene. Later it became possible
medicine, the description of disease mechanisms at the level
to sequence these genes, persuade them to synthesize their
of cells and molecules. However, until the development of
products in microorganisms, cultured cells, or even other
recombinant DNA technology in the mid-1970s, knowledge
species, and hence to define their key regulatory regions.
of events inside the cell nucleus, notably how genes function,
The early work in the field of human molecular genet-
could only be the subject of guesswork based on the structure
ics focused on diseases in which there was some knowl-
and function of their protein products. However, as soon as
edge of the genetic defect at the protein or biochemical level.
it became possible to isolate human genes and to study their
However, once linkage maps of the human genome became
properties, the picture changed dramatically.
available, following the identification of highly polymorphic
Progress over the last 30 years has been driven by techno-
regions of DNA, it was possible to search for any gene for a
logical advances in molecular biology. At first it was possible
disease, even where the cause was completely unknown. This
only to obtain indirect information about the structure and
approach, first called reverse genetics and later rechristened
function of genes by DNA/DNA and DNA/RNA hybridiza-
positional cloning, led to the discovery of genes for many
tion; that is, by probing the quantity or structure of RNA
important diseases.
or DNA by annealing reactions with molecular probes. The
As methods for sequencing were improved and automated,
next major advance was the ability to fractionate DNA into
thoughts turned to the next major goal in this field, which was
pieces of predictable size with bacterial restriction enzymes.
to determine the complete sequence of the bases that con-
This led to the invention of a technique that played a central
stitute our genes and all that lies between them: the Human
role in the early development of human molecular genetics,
Genome Project. This remarkable endeavor was finally com-
called Southern blotting after the name of its developer, Edwin
pleted in 2006. The further understanding of the functions
Southern. This method allowed the structure and organiza-
and regulation of our genes will require multidisciplinary
tion of genes to be studied directly for the first time and led
research encompassing many different fields. The next stage
to the definition of a number of different forms of molecular
in the Human Genome Project, called genome annotation,
pathology.
entails analyzing the raw DNA sequence in order to deter-
Once it was possible to fractionate DNA, it soon became
mine its biological significance. One of the main ventures
feasible to insert the pieces into vectors able to divide
in the era of functional genomics will be in what is termed
proteomics, the large-scale analysis of the protein products of
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben.
genes. The ultimate goal will be to try to define the protein
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.

1
2 Molecular Hematology

complement, or proteome, of cells and how the many dif-


ferent proteins interact with one another. To this end, large-
The structure, genetic control, and
scale facilities are being established for isolating and purify-
synthesis of normal hemoglobin
ing the protein products of genes that have been expressed
in bacteria. Their structure can then be studied by a vari- Structure and function
ety of different techniques, notably X-ray crystallography
The varying oxygen requirements during embryonic, fetal,
and nuclear magnetic resonance spectroscopy. The crys-
and adult life are reflected in the synthesis of different
tallographic analysis of proteins is being greatly facilitated
structural hemoglobins at each stage of human development.
by the use of X-ray beams from a synchrotron radiation
However, they all have the same general tetrameric struc-
source.
ture, consisting of two different pairs of globin chains, each
In the last few years both the utility and extreme com-
attached to one heme molecule. Adult and fetal hemoglobins
plexity of the fruits of the genome project have become
have α chains combined with β chains (Hb A, α2 β2 ),
apparent. The existence of thousands of single-nucleotide
δ chains (Hb A2 , α2 δ2 ), and γ chains (Hb F, α2 γ2 ). In
polymorphisms (SNPs) has made it possible to search for
embryos, α-like chains called ζ chains combine with γ
genes of biological or medical significance. The discovery of
chains to produce Hb Portland (ζ2 γ2 ), or with ε chains to
families of regulatory RNAs and proteins is starting to shed
make Hb Gower 1 (ζ2 ε2 ), while α and ε chains form Hb
light on how the functions of the genome are controlled,
Gower 2 (α2 ε2 ). Fetal hemoglobin is heterogeneous; there
and studies of acquired changes in its structure, epigenetics,
are two varieties of γ chain that differ only in their amino
promise to provide similar information. Recent develop-
acid composition at position 136, which may be occupied by
ments in new-generation sequencing of DNA and RNA are
either glycine or alanine; γ chains containing glycine at this
also providing invaluable information about many aspects
position are called G γ chains, those with alanine A γ chains
of gene regulation.
(Figure 1.1).
During this remarkable period of technical advance, con-
The synthesis of hemoglobin tetramers consisting of two
siderable progress has been made toward an understanding
unlike pairs of globin chains is absolutely essential for the
of the pathology of disease at the molecular level. This has
effective function of hemoglobin as an oxygen carrier. The
had a particular impact on hematology, leading to advances
classical sigmoid shape of the oxygen dissociation curve,
in the understanding of gene function and disease mecha-
which reflects the allosteric properties of the hemoglobin
nisms in almost every aspect of the field.
molecule, ensures that, at high oxygen tensions in the lungs,
The inherited disorders of hemoglobin – the thalassemias
oxygen is readily taken up and later released effectively at the
and structural hemoglobin variants, the commonest human
lower tensions encountered in the tissues. The shape of the
monogenic diseases – were the first to be studied systemati-
curve is quite different to that of myoglobin, a molecule that
cally at the molecular level and a great deal is known about
consists of a single globin chain with heme attached to it,
their genotype–phenotype relationships. This field led the
which, like abnormal hemoglobins that consist of homote-
way to molecular hematology and, indeed, to the develop-
tramers of like chains, has a hyperbolic oxygen dissociation
ment of molecular medicine. Thus, even though the genet-
curve.
ics of hemoglobin is complicated by the fact that different
The transition from a hyperbolic to a sigmoid oxygen
varieties are produced at particular stages of human devel-
dissociation curve, which is absolutely critical for normal
opment, the molecular pathology of the hemoglobinopathies
oxygen delivery, reflects cooperativity between the four
provides an excellent model system for understanding any
heme molecules and their globin subunits. When one of
monogenic disease and the complex interactions between
them takes on oxygen, the affinity of the remaining three
genotype and environment that underlie many multigenic
increases markedly; this happens because hemoglobin can
disorders.
exist in two configurations, deoxy(T) and oxy(R), where T
In this chapter I consider the structure, synthesis, and
and R represent the tight and relaxed states, respectively.
genetic control of the human hemoglobins, describe the
The T configuration has a lower affinity than the R for
molecular pathology of the thalassemias, and discuss briefly
ligands such as oxygen. At some point during the addition
how the complex interactions of their different genotypes
of oxygen to the hemes, the transition from the T to the R
produce a remarkably diverse family of clinical phenotypes;
configuration occurs and the oxygen affinity of the partially
the structural hemoglobin variants are discussed in more
liganded molecule increases dramatically. These allosteric
detail in Chapter 14. Readers who wish to learn more about
changes result from interactions between the iron of the
the methods of molecular genetics, particularly as applied to
heme groups and various bonds within the hemoglobin
the study of hemoglobin disorders, are referred to the reviews
tetramer, which lead to subtle spatial changes as oxygen is
cited at the end of this chapter.
taken on or given up.
Beginnings: the molecular pathology of hemoglobin 3

1 Kb

31 32 99 100 30 31 104 105

ζ ψζ ψα2 ψα1 α2 α1 θ1 ε Gγ Aγ ψβ δ β
16 11

ζ2ε2 ζ2γ2 α2ε2 α2γ2 α2β2 α2δ2


Hb Gower 1 Hb Portland Hb Gower 2 HbF HbA HbA2

Embryo Fetus Adult


Fig. 1.1 The genetic control of human hemoglobin production in embryonic, fetal, and adult life. The standard names for these genes
are as follows: Alpha genes HBA1 and HBA2, Beta gene HBB, Gamma genes HBG1 and HBG2, Delta gene HBD, and the embryonic genes HBE1
and HBZ.

The precise tetrameric structures of the different human In short, oxygen transport can be modified by a variety
hemoglobins, which reflect the primary amino acid of adaptive features in the red cell that include interactions
sequences of their individual globin chains, are also vital between the different heme molecules, the effects of CO2 , and
for the various adaptive changes that are required to ensure differential affinities for 2,3-BPG. These changes, together
adequate tissue oxygenation. The position of the oxygen with more general mechanisms involving the cardiorespira-
dissociation curve can be modified in several ways. For tory system, provide the main basis for physiological adapta-
example, oxygen affinity decreases with increasing CO2 ten- tion to anemia.
sion (the Bohr effect). This facilitates oxygen loading to the
tissues, where a drop in pH due to CO2 influx lowers oxygen
affinity; the opposite effect occurs in the lungs. Oxygen affin- Genetic control of hemoglobin
ity is also modified by the level of 2,3-diphosphoglycerate The α- and β-like globin chains are the products of two dif-
(2,3-BPG) in the red cell. Increasing concentrations shift ferent gene families which are found on different chromo-
the oxygen dissociation curve to the right (i.e. they reduce somes (Figure 1.1). The β-like globin genes form a linked
oxygen affinity), while diminishing concentrations have the cluster on chromosome 11, spread over approximately 60 kb
opposite effect. 2,3-BPG fits into the gap between the two β (kilobase or 1000 nucleotide bases). The different genes that
chains when it widens during deoxygenation, and interacts form this cluster are arranged in the order 5′ –ε–G γ–A γ–
with several specific binding sites in the central cavity of ψβ–δ–β–3′ . The α-like genes also form a linked cluster, in
the molecule. In the deoxy configuration the gap between this case on chromosome 16, in the order 5′ –ζ–ψζ–ψα1–
the two β chains narrows and the molecule cannot be α2–α1–3′ . The ψβ, ψζ, and ψα genes are pseudogenes; that
accommodated. With increasing concentrations of 2,3-BPG, is, they have strong sequence homology with the β, ζ, and
which are found in various hypoxic and anemic states, α genes, but contain a number of differences that prevent
more hemoglobin molecules tend to be held in the deoxy them from directing the synthesis of any products. They may
configuration and the oxygen dissociation curve is therefore reflect remnants of genes that were functional at an earlier
shifted to the right, with more effective release of oxygen. stage of human evolution.
Fetal red cells have greater oxygen affinity than adult The structure of the human globin genes is, in essence,
red cells, although, interestingly, purified fetal hemoglobin similar to that of all mammalian genes. They consist of long
has an oxygen dissociation curve similar to that of adult strings of nucleotides that are divided into coding regions,
hemoglobin. These differences, which are adapted to the oxy- or exons, and non-coding inserts called intervening sequences
gen requirements of fetal life, reflect the relative inability of (IVSs) or introns. The β-like globin genes contain two
Hb F to interact with 2,3-BPG compared with Hb A. This is introns, one of 122–130 base pairs between codons 30 and 31
because the γ chains of Hb F lack specific binding sites for and one of 850–900 base pairs between codons 104 and 105
2,3-BPG. (the exon codons are numbered sequentially from the 5′ to
4 Molecular Hematology

the 3′ end of the gene, i.e. from left to right). Similar, though promoter elements: enhancers (regulatory sequences that
smaller, introns are found in the α and ζ globin genes. These increase gene expression despite being located at a con-
introns and exons, together with short non-coding sequences siderable distance from the genes) and “master” regulatory
at the 5′ and 3′ ends of the genes, represent the major func- sequences called, in the case of the β globin gene cluster, the
tional regions of the particular genes. However, there are also locus control region (LCR) and, in the case of the α genes,
extremely important regulatory sequences which subserve HS40 (a nuclease-hypersensitive site in DNA 40 kb from the
these functions that lie outside the genes themselves. α globin genes). Each of these sequences has a modular struc-
At the 5′ non-coding (flanking) regions of the globin ture made up of an array of short motifs that represent the
genes, as in all mammalian genes, there are blocks of binding sites for transcriptional activators or repressors.
nucleotide homology. The first, the ATA box, is about 30
bases upstream (to the left) of the initiation codon; that is, the
Gene action and globin synthesis
start word for the beginning of protein synthesis (see later).
The second, the CCAAT box, is about 70 base pairs upstream The flow of information between DNA and protein is sum-
from the 5′ end of the genes. About 80–100 bases further marized in Figure 1.2. When a globin gene is transcribed,
upstream there is the sequence GGGGTG, or CACCC, which messenger RNA (mRNA) is synthesized from one of its
may be inverted or duplicated. These three highly conserved strands, a process which begins with the formation of a tran-
DNA sequences, called promoter elements, are involved in scription complex consisting of a variety of regulatory pro-
the initiation of transcription of the individual genes. Finally, teins together with an enzyme called RNA polymerase (see
in the 3′ non-coding region of all the globin genes there later). The primary transcript is a large mRNA precursor
is the sequence AATAAA, which is the signal for cleavage which contains both intron and exon sequences. While in
and polyA addition to RNA transcripts (see Gene Action and the nucleus, this molecule undergoes a variety of modifica-
Globin Synthesis). tions. First, the introns are removed and the exons are spliced
The globin gene clusters also contain several sequences together. The intron/exon junctions always have the same
that constitute regulatory elements, which interact to pro- sequence: GT at their 5′ end, and AG at their 3′ end. This
mote erythroid-specific gene expression and coordination appears to be essential for accurate splicing; if there is a muta-
of the changes in globin gene activity during develop- tion at these sites this process does not occur. Splicing reflects
ment. These include the globin genes themselves and their a complex series of intermediary stages and the interaction

A
C C A
A C T T
C A A A T A
C A T T A A
C T A G A A
Flanking IVS 1 IVS 2 Flanking Gene
NC GT AG GT AG NC
5' 3' mRNA precursor

5' CAP Excision of introns


AAAA-A Splicing of exons
AAAA-A Processed mRNA
Nucleus
Cytoplasm

AAAA-A Translation
AUG UG
Ribosome ACC UUC UAA
U AC G AAG

Transfer RNA

Amino
acid Growing
chain
Finished
chain
Processed Fig. 1.2 The mechanisms of globin gene
chain transcription and translation.
Beginnings: the molecular pathology of hemoglobin 5

of a number of different nuclear proteins. After the exons are the mRNA from left to right, and the growing peptide chain
joined, the mRNAs are modified and stabilized; at their 5′ end is transferred from one incoming tRNA to the next; that is,
a complex CAP structure is formed, while at their 3′ end a the mRNA is translated from 5′ to 3′ . During this time the
string of adenylic acid residues (polyA) is added. The mRNA tRNAs are held in appropriate steric configuration with the
processed in this way moves into the cytoplasm, where it mRNA by the two ribosomal subunits. There are specific
acts as a template for globin chain production. Because of initiation (AUG) and termination (UAA, UAG, and UGA)
the rules of base pairing – that is, cytosine always pairs with codons. When the ribosomes reach the termination codon,
thymine, and guanine with adenine – the structure of the translation ceases, the completed globin chains are released,
mRNA reflects a faithful copy of the DNA codons from which and the ribosomal subunits are recycled. Individual globin
it is synthesized; the only difference is that, in RNA, uracil (U) chains combine with heme, which has been synthesized
replaces thymine (T). through a separate pathway, and then interact with one like
Amino acids are transported to the mRNA template on chain and two unlike chains to form a complete hemoglobin
carriers called transfer RNAs (tRNAs); there are specific tetramer.
tRNAs for each amino acid. Furthermore, because the genetic
code is redundant (i.e. more than one codon can encode a
Regulation of hemoglobin synthesis
particular amino acid), for some of the amino acids there are
several different individual tRNAs. Their order in the globin The regulation of globin gene expression is mediated mainly
chain is determined by the order of codons in the mRNA. at the transcriptional level, with some fine tuning during
The tRNAs contain three bases, which together constitute an translation and post-translational modification of the gene
anticodon; these anticodons are complementary to mRNA products. DNA that is not involved in transcription is held
codons for particular amino acids. They carry amino acids tightly packaged in a compact, chemically modified form that
to the template, where they find the appropriate positioning is inaccessible to transcription factors and polymerases and is
by codon–anticodon base pairing. When the first tRNA is heavily methylated. Activation of a particular gene is reflected
in position, an initiation complex is formed between several by changes in the structure of the surrounding chromatin,
protein initiation factors together with the two subunits that which can be identified by enhanced sensitivity to nucleases.
constitute the ribosomes. A second tRNA moves in along- Erythroid lineage-specific nuclease-hypersensitive sites are
side and the two amino acids that they are carrying form found at several locations in the β globin gene cluster. Four
a peptide bond between them; the globin chain is now two are distributed over 20 kb upstream from the ε globin gene
amino acid residues long. This process is continued along in the region of the β globin LCR (Figure 1.3). This vital

–20 0 20 40 60 kb

5' HS 3' HS
4 3 2 1 1

ε Gγ Aγ δ β
Chromosome 11
LCR

–40 –20 0 20 40 kb

HS–40

ζ α2 α1
Chromosome 16
Fig. 1.3 The positions of the major regulatory regions in the β and α globin gene clusters. The arrows indicate the position of the
erythroid lineage-specific nuclease-hypersensitive sites. HS, hypersensitive.
6 Molecular Hematology

regulatory region is able to establish a transcriptionally active Regulation of developmental changes


domain spanning the entire β globin gene cluster. Several in globin gene expression
enhancer sequences have been identified in this cluster. A
During development, the site of red cell production moves
variety of regulatory proteins bind to the LCR, and to the
from the yolk sac to the fetal liver and spleen, and thence
promoter regions of the globin genes and to the enhancer
to bone marrow in the adult. Embryonic, fetal, and adult
sequences. It is thought that the LCR and other enhancer
hemoglobin synthesis is approximately related in time to
regions become opposed to the promoters to increase
these changes in the site of erythropoiesis, although it is quite
the rate of transcription of the genes to which they are
clear that the various switches, between embryonic and fetal
related.
and between fetal and adult hemoglobin synthesis, are beau-
These regulatory regions contain sequence motifs for
tifully synchronized throughout these different sites. Fetal
various ubiquitous and erythroid-restricted transcription
hemoglobin synthesis declines during the later months of
factors. Binding sites for these factors have been identified in
gestation, and Hb F is replaced by Hb A and Hb A2 by the
each of the globin gene promoters and at the hypersensitive
end of the first year of life.
site regions of the various regulatory elements. A number
Although the exact mechanism of the switch from fetal to
of the factors which bind to these areas are found in all cell
adult hemoglobin is still not understood, recent studies of
types. They include Sp1, Yy1, and Usf. In contrast, a number
patients with unusually high levels of HbF and genome-wide
of transcription factors have been identified, including
association studies (GWAS) have yielded extremely promis-
GATA-1, EKLF, and NF-E2, which are restricted in their
ing information about some of the regulatory genes involved.
distribution to erythroid cells and, in some cases, megakary-
They include BCL11A, MYB, and KLF1. It is clear from stud-
ocytes, and mast cells. The overlapping of erythroid-specific
ies of these and related genes that they are involved directly in
and ubiquitous-factor binding sites in several cases suggests
the regulation of hemoglobin switching, work which is yield-
that competitive binding may play an important part in
ing great promise for the development of future technology
the regulation of erythroid-specific genes. Another binding
for increasing HbF synthesis to modify the phenotype of tha-
factor, SSP, the stage selector protein, appears to interact
lassemias and sickle cell anemia.
specifically with ε and γ genes. Several elements involving
the chromatin and histone acetylation required for access of
these regulatory proteins have been identified.
The binding of hematopoietic-specific factors activates the The molecular pathology
LCR, which renders the entire β globin gene cluster tran- of hemoglobin
scriptionally active. These factors also bind to the enhancer
and promoter sequences, which work in tandem to regu- As is the case for many monogenic diseases, the inherited dis-
late the expression of the individual genes in the clusters. It orders of hemoglobin fall into two major classes. First, there
is likely that some of the transcriptional factors are devel- are those that result from reduced output of one or other
opmental stage specific, and hence may be responsible for globin genes, the thalassemias. Second, there is a wide range
the differential expression of the embryonic, fetal, and adult of conditions that result from the production of structurally
globin genes. The α globin gene cluster also contains an ele- abnormal globin chains; the type of disease depends on how
ment, HS40, which has some structural features in common the particular alteration in protein structure interferes with
with the β LCR, although it is different in aspects of its struc- its stability or function. Of course, no biological classifica-
ture. A number of enhancer-like sequences have been identi- tion is entirely satisfactory and those which attempt to define
fied, although it is becoming clear that there are fundamental the hemoglobin disorders are no exception. There are some
differences in the pattern of regulation of the two globin gene structural hemoglobin variants which happen to be synthe-
clusters. sized at a reduced rate and hence are associated with a clini-
In addition to the different regulatory sequences outlined, cal picture similar to thalassemia. And there are other classes
there are also sequences which may be involved specifically of mutations which simply interfere with the normal tran-
with “silencing” of genes, notably those for the embryonic sition from fetal to adult hemoglobin synthesis, a family of
hemoglobins, during development. conditions given the general title hereditary persistence of fetal
Some degree of regulation is mediated by differences in hemoglobin (HPFH). Furthermore, because these diseases are
the rates of initiation and translation of the different mRNAs, all so common and occur together in particular populations,
and at the post-transcriptional level by differential affinity it is not uncommon for an individual to inherit a gene for
for different protein subunits. However, this kind of post- one or other form of thalassemia and a structural hemoglobin
transcriptional fine tuning probably plays a relatively small variant. The heterogeneous group of conditions that results
role in determining the overall output of the globin gene from these different mutations and interactions is summa-
products. rized in Table 1.1.
Beginnings: the molecular pathology of hemoglobin 7

Table 1.1 The thalassemias and related disorders


Defective β globin gene transcription
There are a variety of mechanisms that interfere with nor-
α Thalassemia γ Thalassemia mal transcription of the β globin genes. First, the genes may
α0 be either completely or partially deleted. Overall, deletions
α+ δ Thalassemia
of the β globin genes are not commonly found in patients
Deletion (−α)
with β thalassemia, with one exception: a 619-bp deletion
Non-deletion (αT ) εγδβ Thalassemia
involving the 3′ end of the gene is found frequently in the
β Thalassemia Hereditary persistence of Sind populations of India and Pakistan, where it constitutes
β0 fetal hemoglobin about 30% of the β thalassemia alleles. Other deletions are
β+ Deletion
extremely rare.
Normal Hb A2 (δβ)0
A much more common group of mutations, which results
“Silent” Non-deletion
Dominant Linked to β globin genes
in a moderate decrease in the rate of transcription of the
β globin genes, involves single-nucleotide substitutions in
δβ Thalassemia G γβ+
or near the TATA box at about −30 nucleotides (nt) from
(δβ)+ A γβ+
the transcription start site, or in the proximal or distal pro-
(δβ)0 Unlinked to β globin genes
moter elements at −90 and −105 nt. These mutations result
(A γδβ)0
in decreased β globin mRNA production, ranging from 10%
to 25% of the normal output. Thus, they are usually associ-
Over recent years, determination of the molecular pathol- ated with the mild forms of β+ thalassemia. They are particu-
ogy of the two common forms of thalassemia, α and β, has larly common in African populations, an observation which
provided a remarkable picture of the repertoire of mutations explains the unusual mildness of β thalassemia in this racial
that can underlie human monogenic disease. In the sections group. One particular mutation, C → T at position −101 nt
that follow I describe, in outline, the different forms of molec- to the β globin gene, causes an extremely mild deficit of β
ular pathology that underlie these conditions. globin mRNA. Indeed, this allele is so mild that it is com-
pletely silent in carriers and can only be identified by its inter-
The β thalassemias action with more severe β thalassemia alleles in compound
heterozygotes.
There are two main classes of β thalassemia, β0 thalassemia,
in which there is an absence of β globin chain production,
Mutations that cause abnormal processing
and β+ thalassemia, in which there is a variable reduction
of mRNA
in the output of β globin chains. As shown in Figure 1.4,
mutations of the β globin genes may cause a reduced output As mentioned earlier, the boundaries between exons and
of gene product at the level of transcription or mRNA introns are marked by the invariant dinucleotides GT at the
processing, or translation, or through the stability of the donor (5′ ) site and AG at the acceptor (3′ ) site. Mutations that
globin gene product. affect either of these sites completely abolish normal splicing

Deletions

I IVSI 2 IVS 2 3

PR C I FS SPL SPL FS SPL SPL FS Poly A


NS NS NS

100 bp
Point mutations
Fig. 1.4 The mutations of the β globin gene that underlie β thalassemia. The heavy black lines indicate the length of the deletions. The
point mutations are designated as follows: PR, promoter; C, CAP site; I, initiation codon; FS, frameshift and nonsense mutations; SPL, splice
mutations; Poly A, poly A addition site mutations.
8 Molecular Hematology

and produce the phenotype of β0 thalassemia. The transcrip- globin mRNA and hence in the clinical phenotype of severe
tion of genes carrying these mutations appears to be normal, β thalassemia. Interestingly, mutations at codons 19, 26, and
but there is complete inactivation of splicing at the altered 27 result in both reduced production of normal mRNA (due
junction. to abnormal splicing) and an amino acid substitution when
Another family of mutations involves what are called splice the mRNA which is spliced normally is translated into pro-
site consensus sequences. Although only the GT dinucleotide tein. The abnormal hemoglobins produced are Hb Malay, Hb
is invariant at the donor splice site, there is conservation E, and Hb Knossos, respectively. All these variants are associ-
of adjacent nucleotides and a common, or consensus, ated with a mild β+ thalassemia-like phenotype. These muta-
sequence of these regions can be identified. Mutations tions illustrate how sequence changes in coding rather than
within this sequence can reduce the efficiency of splicing IVS influence RNA processing, and underline the impor-
to varying degrees, because they lead to alternate splicing tance of competition between potential splice site sequences
at the surrounding cryptic sites. For example, mutations of in generating both normal and abnormal varieties of β globin
the nucleotide at position 5 of IVS-1 (the first intervening mRNA.
sequence), G → C or T, result in a marked reduction of Cryptic splice sites in introns may also carry mutations
β chain production and in the phenotype of severe β+ that activate them even though the normal splice sites remain
thalassemia. On the other hand, the substitution of C for T intact. A common mutation of this kind in Mediterranean
at position 6 in IVS-1 leads to only a mild reduction in the populations involves a base substitution at position 110 in
output of β chains. IVS-1. This region contains a sequence similar to a 3′ accep-
Another mechanism that leads to abnormal splicing tor site, though it lacks the invariant AG dinucleotide. The
involves cryptic splice sites. These are regions of DNA which, change of the G to A at position 110 creates this dinucleotide.
if mutated, assume the function of a splice site at an inappro- The result is that about 90% of the RNA transcript splices to
priate region of the mRNA precursor. For example, a variety this particular site and only 10% to the normal site, again pro-
of mutations activate a cryptic site which spans codons 24– ducing the phenotype of severe β+ thalassemia (Figure 1.5).
27 of exon 1 of the β globin gene. This site contains a GT Several other β thalassemia mutations have been described
dinucleotide, and adjacent substitutions that alter it so that it which generate new donor sites within IVS-2 of the β globin
more closely resembles the consensus donor splice site result gene.
in its activation, even though the normal splice site is intact. A Another family of mutations that interferes with β globin
mutation at codon 24 GGT → GGA, though it does not alter gene processing involves the sequence AAUAAA in the 3′
the amino acid which is normally found in this position in untranslated regions, which is the signal for cleavage and
the β globin chain (glycine), allows some splicing to occur polyadenylation of the β globin gene transcript. Somehow,
at this site instead of the exon–intron boundary. This results these mutations destabilize the transcript. For example, a
in the production of both normal and abnormally spliced β T → C substitution in this sequence leads to only one-tenth of

Normal splicing

GT IVS1 AG GT IVS2 AG
β gene

TTGGTCT

10%
β+ thalassemia Fig. 1.5 The generation of a new splice site in
an intron as the mechanism for a form of β+
90% thalassemia. For details see text.
Beginnings: the molecular pathology of hemoglobin 9

the normal amount of β globin mRNA transcript, and hence absence of mRNA from the cytoplasm of red cell precursors.
to the phenotype of a moderately severe β+ thalassemia. This appears to be an adaptive mechanism, called nonsense-
Another example of a mutation which probably leads to mediated decay, whereby abnormal mRNA of this type is not
defective processing of the function of β globin mRNA is the transported to the cytoplasm, where it would act as a tem-
single-base substitution A → C in the CAP site. It is not yet plate for the production of truncated gene products. How-
understood how this mutation causes a reduced rate of tran- ever, in the case of exon III mutations, apparently because
scription of the β globin gene. this process requires the presence of an intact upstream exon,
There is another small subset of rare mutations that involve the abnormal mRNA is transported into the cytoplasm and
the 3′ untranslated region of the β globin gene, and these are hence can act as a template for the production of unstable
associated with relatively mild forms of β thalassemia. It is β globin chains. The latter precipitate in the red cell pre-
thought that these interfere in some way with transcription, cursors together with excess α chains to form large inclu-
but the mechanism is unknown. sion bodies, and hence there is enough globin chain imbal-
ance in heterozygotes to produce a moderately severe degree
of anemia.
Mutations that result in abnormal
translation of β globin mRNA
The α thalassemias
There are three main classes of mutations of this kind.
Base substitutions that change an amino acid codon to a The molecular pathology of the α thalassemias is more com-
chain termination codon prevent the translation of β globin plicated than that of the β thalassemias, simply because there
mRNA and result in the phenotype of β0 thalassemia. Several are two α globin genes per haploid genome. Thus, the normal
mutations of this kind have been described; the common- α globin genotype can be written αα/αα. As in the case of β
est, involving codon 17, occurs widely throughout South- thalassemia, there are two major varieties of α thalassemia,
east Asia. Similarly, a codon 39 mutation is encountered fre- α+ and α0 thalassemia. In α+ thalassemia one of the linked α
quently in the Mediterranean region. globin genes is lost, either by deletion (−) or mutation (T); the
The second class involves the insertion or deletion of heterozygous genotype can be written –α/αα or αT α/αα.
one, two, or four nucleotides in the coding region of the β In α0 thalassemia the loss of both α globin genes nearly
globin gene. These disrupt the normal reading frame, cause always results from a deletion; the heterozygous genotype is
a frameshift, and hence interfere with the translation of β therefore written − −/αα. In populations where specific dele-
globin mRNA. The end result is the insertion of anomalous tions are particularly common, Southeast Asia (SEA) or the
amino acids after the frameshift until a termination codon Mediterranean region (MED), it is useful to add the appropri-
is reached in the new reading frame. This type of mutation ate superscript as follows: – –SEA /αα or – –MED /αα. It follows
always leads to the phenotype of β0 thalassemia. that when we speak of an “α thalassemia gene,” what we are
Finally, there are several mutations which involve the β really referring to is a haplotype; that is, the state and function
globin gene initiation codon and which, presumably, reduce of both of the linked α globin genes.
the efficiency of translation.
α0 thalassemia
Unstable β globin chain variants
Three main molecular pathologies, all involving deletions,
Some forms of β thalassemia result from the synthesis of have been found to underlie the α0 thalassemia phenotype.
highly unstable β globin chains which are incapable of form- The majority of cases result from deletions that remove both
ing hemoglobin tetramers and which are rapidly degraded, α globin genes and a varying length of the α globin gene
leading to the phenotype of β0 thalassemia. Indeed, in many cluster (Figure 1.6). Occasionally, however, the α globin gene
of these conditions no abnormal globin chain product can be cluster is intact, but is inactivated by a deletion which involves
demonstrated by protein analysis, and the molecular pathol- the major regulatory region HS40, 40 kb upstream from the
ogy has to be interpreted simply on the basis of a derived α globin genes, or the α globin genes may be lost as part of a
sequence of the variant β chain obtained by DNA analysis. truncation of the tip of the short arm of chromosome 16.
Recent studies have provided some interesting insights As well as providing us with an understanding of the
into how complex clinical phenotypes may result from the molecular basis for α0 thalassemia, detailed studies of these
synthesis of unstable β globin products. For example, there deletions have yielded more general information about the
is a spectrum of disorders that result from mutations in exon mechanisms that underlie this form of molecular pathology.
3 which give rise to a moderately severe form of β tha- For example, it has been found that the 5′ breakpoints of a
lassemia in heterozygotes. It has been found that nonsense or number of deletions of the α globin gene cluster are located
frameshift mutations in exons I and II are associated with the approximately the same distance apart and in the same order
10 Molecular Hematology

–50 –10 0 10 20 30

ζ2 ψζ1 ψα2 ψα1 α2 α1 θ1

Inter-ζHVR 3'HVR

Fig. 1.6 Some of the deletions that underlie α0


and α+ thalassemia. The colored rectangles
beneath the α globin gene cluster indicate the
lengths of the deletions. The unshaded regions
indicate uncertainty about the precise breakpoints.
The three small deletions at the bottom of the figure
represent the common α+ thalassemia deletions.
HVR, highly variable regions.

along the chromosome as their respective 3′ breakpoints; α globin gene region, a finding that provides a completely
similar findings have been observed in deletions of the β new mechanism for genetic disease. In another case of α0
globin gene cluster. These deletions seem to have resulted thalassemia, in which no molecular defects could be detected
from illegitimate recombination events, which have led to the in the α globin gene cluster, a gain-of-function regulatory
deletion of an integral number of chromatin loops as they polymorphism was found in the region between the α globin
pass through their nuclear attachment points during chro- genes and their upstream regulatory elements. This alteration
mosomal replication. Another long deletion has been charac- creates a new promoter-like element that interferes with the
terized in which a new piece of DNA bridges the two break- normal activation of all downstream α-like globin genes.
points in the α globin gene cluster. The inserted sequence In short, detailed analysis of the molecular pathology of
originates upstream from the α globin gene cluster, where the α0 thalassemias has provided valuable evidence not only
normally it is found in an inverted orientation with respect about how large deletions of gene clusters are caused, but also
to that found between the breakpoints of the deletion. Thus about some of the complex mechanisms that may underlie
it appears to have been incorporated into the junction in a cases in which the α gene clusters remain intact, but in which
way that reflects its close proximity to the deletion breakpoint their function is completely suppressed.
region during replication. Other deletions seem to be related
to the family of Alu-repeats, simple repeat sequences that
α+ thalassemia
are widely dispersed throughout the genome; one deletion
appears to have resulted from a simple homologous recombi- As mentioned earlier, the α+ thalassemias result from the
nation between two repeats of this kind that are usually 62 kb inactivation of one of the duplicated α globin genes, by either
apart. deletion or point mutation.
A number of forms of α0 thalassemia result from terminal 𝛼 + Thalassemia due to gene deletions. There are two com-
truncations of the short arm of chromosome 16 to a site about mon forms of α+ thalassemia that are due to loss of one
50 kb distal to the α globin genes. The telomeric consensus or other of the duplicated α globin genes, −α3.7 and −α4.2 ,
sequence TTAGGGn has been added directly to the site of the where 3.7 and 4.2 indicate the size of the deletions. The way
break. Since these mutations are stably inherited, it appears in which these deletions have been generated, approximately
that telomeric DNA alone is sufficient to stabilize the ends of 4 kb long, was probably generated by an ancient duplication
broken chromosomes. event. The homologous regions, which are divided by small
Quite recently, two other molecular mechanisms have inserts, are designated X, Y, and Z. The duplicated Z boxes
been identified as the cause of α0 thalassemia which, though are 3.7 kb apart and the X boxes are 4.2 kb apart. The result
rare, may have important implications for an understand- of misalignment reflects the underlying structure of the α
ing of the molecular pathology of other genetic diseases. In globin gene complex (Figure 1.7). Each α gene lies within
one case, a deletion in the α globin gene cluster resulted a boundary of homology and reciprocal crossover between
in a widely expressed gene (LUC7L) becoming juxtaposed these segments at meiosis, a chromosome is produced with
to a structurally normal α globin gene. Although the latter either a single (−α) or triplicated (ααα) α globin gene. As
retained all its important regulatory elements, its expression shown in Figure 1.7, if a crossover occurs between homol-
was silenced. It was found in a transgenic mouse model that ogous Z boxes 3.7 kb of DNA are lost, an event which is
transcription of antisense RNA mediated the silencing of the described as a rightward deletion, −α3.7 . A similar crossover
Beginnings: the molecular pathology of hemoglobin 11

ψα1 α2 α1

X Y Z X Y Z
(a)
ψα1 α2 α1
3.7
αααanti

ψα1 α2 α1
3.7
–α

(b) Rightward crossover

ψα1 α2 α1
4.2
αααanti
Fig. 1.7 Mechanisms of the generation of the
common deletion forms of α+ thalassemia. (a)
The normal arrangement of the α globin genes, ψα1 α2 α1
4.2
with the regions of homology X, Y, and Z. (b) The –α
crossover that generates the −α3.7 deletion. (c) The
crossover that generates the −α4.2 deletion. (c) Leftward crossover

between the two X boxes deletes 4.2 kb, the leftward deletion gene, although there is considerable evidence that it in some
−α4.2 . The corresponding triplicated α gene arrangements way destabilizes the mRNA.
are called αααanti 3.7 and αααanti 4.2 . A variety of different
points of crossing over within the Z boxes give rise to differ-
α thalassemia/mental retardation syndromes
ent length deletions, still involving 3.7 kb.
Non-deletion types of 𝛼 + thalassemia. These disorders There is a family of mild forms of α thalassemia which is
result from single or oligonucleotide mutations of the par- quite different to that described in the previous section and
ticular α globin gene. Most of them involve the α2 gene but, which is associated with varying degrees of mental retarda-
since the output from this locus is two to three times greater tion. Recent studies indicate that there are two quite different
than that from the α1 gene, this may simply reflect ascertain- varieties of this condition, one encoded on chromosome 16
ment bias due to the greater phenotypic effect and, possibly, (ATR-16) and the other on the X chromosome (ATR-X).
a greater selective advantage. The ATR-16 syndrome is characterized by relatively mild
Overall, these mutations interfere with α globin gene mental disability with a variable constellation of facial and
function in a similar way to those that affect the β globin skeletal dysmorphisms. These individuals have long dele-
genes. They affect the transcription, translation, or post- tions involving the α globin gene cluster, but removing
translational stability of the gene product. Since the prin- at least 1–2 Mb. This condition can arise in several ways,
ciples are the same as for β thalassemia, we do not need including unbalanced translocation involving chromosome
to describe them in detail, with one exception, a mutation 16, truncation of the tip of chromosome 16, and the loss of
which has not been observed in the β globin gene cluster. the α globin gene cluster and parts of its flanking regions by
It turns out that there is a family of mutations that involves other mechanisms.
the α2 globin gene termination codon, TAA. Each specifi- The ATR-X syndrome results from mutations in a gene on
cally changes this codon so that an amino acid is inserted the X chromosome, Xq13.1–q21.1. The product of this gene
instead of the chain terminating. This is followed by “read- is one of a family of proteins involved in chromatin-mediated
through” of α globin mRNA, which is not normally trans- transcriptional regulation. It is expressed ubiquitously dur-
lated until another in-phase termination codon is reached. ing development and at interphase it is found entirely within
The result is an elongated α chain with 31 additional residues the nucleus in association with pericentromeric heterochro-
at the C-terminal end. Five hemoglobin variants of this type matin. In metaphase, it is similarly found close to the cen-
have been identified. The commonest, Hb Constant Spring, tromeres of many chromosomes but, in addition, occurs at
occurs at a high frequency in many parts of Southeast Asia. the stalks of acrocentric chromosomes, where the sequences
It is not absolutely clear why the read-through of normally for ribosomal RNA are located. These locations provide
untranslated mRNAs leads to a reduced output from the α2 important clues to the potential role of this protein in the
12 Molecular Hematology

establishment and/or maintenance of methylation of the are left intact and the deletion simply removes the δ and β
genome. Although it is clear that ATR-X is involved in α globin genes; in these cases both the G γ and the A γ globin
globin transcription, it also must be an important player in gene remain functional. For some reason, these long dele-
early fetal development, particularly of the urogenital system tions allow persistent synthesis of the γ globin genes at a
and brain. Many different mutations of this gene have been relatively high level during adult life, which helps to com-
discovered in association with the widespread morphologi- pensate for the absence of β and δ globin chain production.
cal and developmental abnormalities which characterize the They are classified according to the kind of fetal hemoglobin
ATR-X syndrome. that is produced, and hence into two varieties, G γ(A γδβ)0
and G γA γ(δβ)0 thalassemia; in line with other forms of tha-
lassemia, they are best described by what is not produced:
α thalassemia and the myelodysplastic syndrome
(A γδβ)0 and (δβ)0 thalassemia, respectively. Homozygotes
Since the first description of Hb H (see later section) in the red produce only fetal hemoglobin, while heterozygotes have a
cells of a patient with leukemia, many examples of this asso- thalassemic blood picture together with about 5–15% Hb F.
ciation have been reported. The condition usually is reflected
in a mild form of Hb H disease, with typical Hb H inclusions
Hereditary persistence of fetal hemoglobin
in a proportion of the red cells and varying amounts of Hb H
demonstrable by hemoglobin electrophoresis. The hemato- Genetically determined persistent fetal hemoglobin synthe-
logical findings are usually those of one or other form of the sis in adult life is of no clinical importance, except that its
myelodysplastic syndrome. The condition occurs predomi- genetic determinants can interact with the β thalassemias or
nantly in males in older age groups. Very recently it has been structural hemoglobin variants; the resulting high level of Hb
found that some patients with this condition have mutations F production often ameliorates these conditions. The differ-
involving ATR-X. The relationship of these mutations to the ent forms of HPFH result from either long deletions involv-
associated myelodysplasia remains to be determined. ing the δβ globin gene cluster, similar to those that cause
(δβ)0 thalassemia, or point mutations that involve the pro-
moters of the G γ or A γ globin gene. In the former case there
Rarer forms of thalassemia and related
is no β globin chain synthesis and therefore these condi-
disorders
tions are classified as (δβ)0 HPFH. In cases in which there
There are a variety of other conditions that involve the β are promoter mutations involving the γ globin genes, there is
globin gene cluster which, although less common than the increased γ globin chain production in adult life associated
β thalassemias, provide some important information about with some β and δ chain synthesis in cis (i.e. directed by the
mechanisms of molecular pathology and therefore should be same chromosome) to the HPFH mutations. Thus, depend-
mentioned briefly. ing on whether the point mutations involve the promoter of
the G γ or A γ globin gene, these conditions are called G γ β+
HPFH and A γ β+ HPFH, respectively.
The δβ thalassemias
There is another family of HPFH-like disorders in which
Like the β thalassemias, the δβ thalassemias, which result the genetic determinant is not encoded in the β chain clus-
from defective δ and β chain synthesis, are subdivided into ter. In one case the determinant encodes on chromosome 6,
the (δβ)+ and (δβ)0 forms. although its nature has not yet been determined.
The (δβ)+ thalassemias result from unequal crossover It should be pointed out that all these conditions are very
between the δ and β globin gene loci at meiosis with the heterogeneous and that many different deletions or point
production of δβ fusion genes. The resulting δβ fusion mutations have been discovered that produce the rather sim-
chain products combine with α chains to form a family of ilar phenotypes of (δβ)0 or G γ or A γ β+ HPFH.
hemoglobin variants called the hemoglobin Lepores, after the
family name of the first patient of this kind to be discovered.
Because the synthesis of these variants is directed by genes Genotype–phenotype relationships in
with the 5′ sequences of the δ globin genes, which have defec- the thalassemias
tive promoters, they are synthesized at a reduced rate and
result in the phenotype of a moderately severe form of δβ It is now necessary briefly to relate the remarkably diverse
thalassemia. molecular pathology described in the previous sections to
The (δβ)0 thalassemias nearly all result from long dele- the phenotypes observed in patients with these diseases.
tions involving the β globin gene complex. Sometimes they It is not possible to describe all these complex issues here.
involve the A γ globin chains and hence the only active locus Rather, I shall focus on those aspects that illustrate the more
remaining is the G γ locus. In other cases the G γ and A γ loci general principles of how abnormal gene action is reflected
Beginnings: the molecular pathology of hemoglobin 13

in a particular clinical picture. Perhaps the most important membrane causes alterations in its structure, and their degra-
question that I will address is why patients with apparently dation products, notably heme, hemin (oxidized heme), and
identical genetic lesions have widely differing disorders, iron, result in oxidative damage to the red cell contents
a problem that still bedevils the whole field of medical and membrane. These interactions result in intramedullary
genetics, even in the molecular era. destruction of red cell precursors and in shortened survival
of such cells as they reach the peripheral blood. The end
result is anemia of varying severity. This, in turn, causes tis-
The β thalassemias sue hypoxia and the production of relatively large amounts of
As we have seen, the basic defect that results from the 200 erythropoietin; this leads to a massive expansion of the inef-
or more different mutations that underlie these conditions is fective bone marrow, resulting in bone deformity, a hyperme-
reduced β globin chain production. Synthesis of the α globin tabolic state with wasting and malaise, and bone fragility.
chain proceeds normally and hence there is imbalanced A large proportion of hemoglobin in the blood of β tha-
globin chain output with an excess of α chains (Figure 1.8). lassemics is of the fetal variety. Normal individuals produce
Unpaired α chains precipitate in both red cell precursors and about 1% of Hb F, unevenly distributed among their red cells.
their progeny with the production of inclusion bodies. These In the bone marrow of β thalassemics, any red cell precursors
interfere with normal red cell maturation and survival in a that synthesize γ chains come under strong selection because
variety of complex ways. Their attachment to the red cell they combine with α chains to produce fetal hemoglobin, and

α
Ex
ce
γ ss
β

α2γ2 Denaturation
HbF Degradation

Selective survival of
HbF-containing Hemolysis Destruction of RBC
precursors precursors

Increased levels of Splenomegaly Ineffective


HbF in red cells (pooling, plasma erythropoiesis
volume expansion)

High
oxygen affinity of Anemia
red cells
R
O ed u e
c su a
2 d
eli ed Tis poxi
ve h y
ry
Erythropoietin

Transfusion
Marrow
expansion
Inc
r
ab ease
so d
rp iro
tio n
Skeletal deformity n Iron loading
Increased metabolic rate
Wasting
Gout
Folate deficiency
Endocrine deficiencies
Cirrhosis
Fig. 1.8 The pathophysiology of β thalassemia. Cardiac failure
14 Molecular Hematology

therefore the degree of globin chain imbalance is reduced.


Table 1.2 Mechanisms for the phenotypic diversity of the β
Furthermore, the likelihood of γ chain production seems to thalassemias
be increased in a highly stimulated erythroid bone marrow.
It seems likely that these two factors combine to increase the Genetic modifiers
relative output of Hb F in this disorder. However, it has a Primary: alleles of varying severity
higher oxygen affinity than Hb A, and hence patients with Secondary: modifiers of globin chain imbalance
β thalassemia are not able to adapt to low hemoglobin levels α Thalassemia
as well as those who have adult hemoglobin. Increased α globin genes: ααα or αααα
The greatly expanded, ineffective erythron leads to an Genes involved in unusually high Hb F response
increased rate of iron absorption; this, combined with iron Tertiary: modifiers of complications
Iron absorption, bone disease, jaundice, infection
received by blood transfusion, leads to progressive iron
loading of the tissues, with subsequent liver, cardiac, and Adaptation to anemiaa
endocrine damage. Variation in oxygen affinity (P50 ) of hemoglobin
The constant bombardment of the spleen with abnormal Variation in erythropoietin response to anemia
red cells leads to its hypertrophy. Hence there is progressive Environmental
splenomegaly with an increased plasma volume and trapping Nutrition
of part of the circulating red cell mass in the spleen. This Infection
leads to worsening of the anemia. All these pathophysiolog- Others
ical mechanisms, except for iron loading, can be reversed by
a There may be genetic variation in the adaptive mechanisms.
regular blood transfusion which, in effect, shuts off the inef-
fective bone marrow and its consequences.
Thus it is possible to relate nearly all the important fea-
tures of the severe forms of β thalassemia to the primary of the β thalassemia phenotype into primary, secondary, and
defect in globin gene action. However, can we also explain tertiary classes (Table 1.2).
their remarkable clinical diversity? The primary modifiers are the different β thalassemia alle-
les that can interact together. For example, compound het-
erozygotes for a severe β0 thalassemia mutation and a milder
Phenotypic diversity
one may have an intermediate form of β thalassemia of
Although the bulk of patients who are homozygous for β tha- varying severity, depending on the degree of reduction in β
lassemia mutations or compound heterozygotes for two dif- globin synthesis under the action of the milder allele. This is
ferent mutations have a severe transfusion-dependent phe- undoubtedly one mechanism for the varying severity of Hb
notype, there are many exceptions. Some patients of this type E/β thalassemia; it simply reflects the variable action of the
have a milder course, requiring few or even no transfusions, β thalassemia mutation that is inherited together with Hb E.
a condition called β thalassemia intermedia. A particularly However, this explanation is not relevant in cases in which
important example of this condition is illustrated by the clin- patients with identical β thalassemia mutations have widely
ical findings in those who inherit β thalassemia from one disparate phenotypes.
parent and Hb E from the other, a disorder called Hb E/β The secondary modifiers are those which directly affect
thalassemia. Because the mutation that produces Hb E also the degree of globin chain imbalance. Patients with β tha-
opens up an alternative splice site in the first exon of the β lassemia who also inherit one or other form of α thalassemia
globin gene, it is synthesized at a reduced rate and therefore tend to have a milder phenotype because of the reduction
behaves like a mild form of β thalassemia. It is the common- in the excess of α globin genes caused by the coexistent α
est hemoglobin variant globally and Hb E/β thalassemia is thalassemia allele. Similarly, patients with severe forms of
the commonest form of severe thalassemia in many Asian thalassemia who inherit more α genes than normal because
countries. It has an extraordinarily variable phenotype, rang- their parents have triplicated or quadruplicated α gene
ing from a condition indistinguishable from β thalassemia arrangements tend to have more severe phenotypes. Other
major to one of such mildness that patients grow and develop patients with severe thalassemia alleles appear to run a
quite normally and never require transfusion. milder course because of a genetically determined ability to
Over recent years a great deal has been learned about some produce more γ globin chains and hence fetal hemoglobin,
of the mechanisms involved in this remarkable phenotypic a mechanism that also results in a reduced degree of globin
variability. In short, it reflects both the action of modifying chain imbalance. It is now clear that several gene loci are
genes and variability in adaptation to anemia and, almost cer- involved in this mechanism; the best characterized is a
tainly, the effects of the environment. Given the complexity of polymorphism in the promoter region of the G γ globin gene
these interactions, it is helpful to divide the genetic modifiers that appears to increase the output from this locus under
Beginnings: the molecular pathology of hemoglobin 15

conditions of hemopoietic stress. However, there are clearly Fetus Adult


other genes involved in increasing the output of Hb F. Recent γ β
genome-wide linkage studies have shown clear evidence that α
there are determinants on chromosomes 6 and 8 and a par-
ticularly strong association has been found with BCL11A, a
transcription factor known to be involved in hematopoiesis. α2γ2 α2β2
The exact mechanism for the associated increase in Hb F Excess Excess
in β thalassemia and in sickle cell anemia remains to be
determined.
The tertiary modifiers are those that have no effect on
hemoglobin synthesis, but modify the many different com- γ4 β4
Hb Bart's Hb H
plications of the β thalassemias, including osteoporosis, iron
absorption, jaundice, and susceptibility to infection. High oxygen affinity—hypoxia
Although neglected until recently, it is also becoming Instability of homotetramers
Inclusion bodies. Membrane damage
apparent that variation in adaptation to anemia and the envi-
Shortened red cell survival—hemolysis
ronment may also play a role in phenotypic modification Splenomegaly—hypersplenism
of the β thalassemias. For example, patients with Hb E/β
Fig. 1.9 The pathophysiology of α thalassemia.
thalassemia have relatively low levels of Hb F and hence
their oxygen dissociation curves are more right-shifted than
patients with other forms of β thalassemia intermedia with course, there is a reduction in normal hemoglobin synthe-
significantly higher levels of Hb F. Very recent studies also sis, which results in hypochromic, microcytic erythrocytes.
suggest that the erythropoietin response to severe anemia Another important factor in the pathophysiology of the α
for a given hemoglobin level varies considerably with age; thalassemias is the fact that Hb Bart’s and Hb H are useless
patients during the first years of life have significantly higher oxygen carriers, having an oxygen dissociation curve similar
responses to the same hemoglobin level than those who to that of myoglobin. Thus the circulating hemoglobin level
are older. This observation may go some way to explaining may give a false impression of the oxygen-delivering capacity
the variation in phenotype at different ages that has been of the blood, and patients may be symptomatic at relatively
observed in children with Hb E/β thalassemia. Finally, it is high hemoglobin levels.
clear that further studies are required to dissociate the effects The different clinical phenotypes of the α thalassemias are
on the phenotype of genetic modifiers and environmental an elegant example of the effects of gene dosage (Figure 1.10).
factors. The heterozygous state for α+ thalassemia is associated with
Thus, the phenotypic variability of the β thalassemias minimal hematological changes. That for α0 thalassemia (the
reflects several layers of complex interactions involving loss of two α globin genes) is characterized by moderate
genetic modifiers together with variation in adaptation and, hypochromia and microcytosis, similar to that of the β tha-
almost certainly, the environment. These complex interac- lassemia trait. It does not matter whether the α genes are lost
tions are summarized in Table 1.2. on the same chromosome or on opposite pairs of homolo-
gous chromosomes. Hence the homozygous state for α+ tha-
lassemia, −α/−α, has a similar phenotype to the heterozy-
The α thalassemias
gous state for α0 thalassemia (− −/αα).
The pathophysiology of the α thalassemias differs from that The loss of three α globin genes, which usually results
of the β thalassemias mainly because of the properties of the from the compound heterozygous states for α0 and α+ tha-
excess globin chains that are produced as a result of defective lassemia, is associated with a moderately severe anemia with
α chain synthesis. While the excess α chains produced in β the production of varying levels of Hb H. This condition,
thalassemia are unstable and precipitate, this is not the case hemoglobin H disease, is characterized by varying anemia
in the α thalassemias, in which excess γ chains or β chains and splenomegaly with a marked shortening of red cell
are able to form the soluble homotetramers γ4 (Hb Bart’s) survival.
and β4 (Hb H) (Figure 1.9). Although these variants, par- Finally, the homozygous state for α0 thalassemia (− −/
ticularly Hb H, are unstable and precipitate in older red cell − −) is characterized by death in utero or just after birth, with
populations, they remain soluble sufficiently long for the red the clinical picture of hydrops fetalis. These babies produce
cells to mature and develop relatively normally. Hence there no α chains and their hemoglobin consists mainly of Hb
is far less ineffective erythropoiesis in the α thalassemias and Bart’s with variable persistence of embryonic hemoglobin.
the main cause of the anemia is hemolysis associated with This is reflected in gross intrauterine hypoxia; although these
the precipitation of Hb H in older red cells. In addition, of babies may have hemoglobin values as high as 8–9 g/dL,
16 Molecular Hematology

α0 Thal. trait α0 Thal. trait different amino acid in the affected globin chain. Rarely,
these variants result from more subtle alterations in the
structure of the α/β globin chains. For example, shortened
X chains may result from internal deletions of their particular
genes, while elongated chains result from either duplications
within genes or frameshift mutations, which allow the
chain termination codon to be read through and in which
additional amino acids are added to the C-terminal end. The
majority of the 700 or more structural hemoglobin variants
are of no clinical significance but a few, because they interfere
with the stability or functions of the hemoglobin molecule,
are associated with a clinical phenotype of varying severity.
Normal α0 Thal. α0 Thal. Hb Bart's
trait trait hydrops

Genotype–phenotype relationships
α0 Thal. trait α+ Thal. trait The sickling disorders

The sickling disorders represent the homozygous state for the


X sickle cell gene, sickle cell anemia, and the compound het-
erozygous state for the sickle cell gene and various structural
hemoglobin variants, or β thalassemia. The chronic hemol-
ysis and episodes of vascular occlusion and red cell seques-
tration that characterize sickle cell anemia can all be related
to the replacement of the normal β6 glutamic acid by valine
in Hb S. This causes a hydrophobic interaction with another
hemoglobin molecule, triggering aggregation into large poly-
mers. It is this change that causes the sickling distortion of the
Normal α0 Thal. α+ Thal. Hb H
trait trait disease
red blood cell and hence a marked decrease in its deformabil-
ity. The resulting rigidity of the red cells is responsible for the
Fig. 1.10 The genetics of the common forms of α thalassemia.
vaso-occlusive changes that lead to many of the most serious
The open boxes represent normal α genes and the green boxes
deleted α genes. The mating shown at the top shows how two α0
aspects of all the sickling disorders.
thalassemia heterozygotes can produce a baby with the Hb Bart’s The different conformations of sickle cells (banana shaped
hydrops syndrome. In the mating at the bottom, between individuals or resembling a holly leaf) reflect different orientations of
with α0 and α+ thalassemia, one in four of the offspring will have Hb bundles of fibers along the long axis of the cell, the three-
H disease. dimensional structure of which is constituted by a rope-like
polymer composed of 14 strands. The rate and extent of poly-
mer formation depend on the degree of oxygenation, the cel-
most of it is unable to release its oxygen. This is reflected in
lular hemoglobin concentration, and the presence or absence
the hydropic changes, a massive outpouring of nucleated red
of Hb F. The latter inhibits polymerization and hence tends
cells, and hepatosplenomegaly with persistent hematopoiesis
to ameliorate sickling. Polymerization of Hb S causes dam-
in the liver and spleen.
age to the red cell membrane, the result of which is an irre-
versibly sickled cell. Probably the most important mechanism
Structural hemoglobin variants is cellular dehydration resulting from abnormalities of potas-
sium/chloride cotransport and Ca2+ -activated potassium
The structural hemoglobin variants are described in detail in efflux. This is sufficient to trigger the Ca2+ -dependent (Gar-
Chapter 14. Here, their molecular pathology and genotype– dos) potassium channel, providing a mechanism for the loss
phenotype relationships are briefly outlined. of potassium and water and leading to cellular dehydration.
However, the vascular pathology of the sickling disorders
is not entirely related to the rigidity of sickled red cells.
Molecular pathology
There is now a wealth of evidence that abnormal interactions
The molecular pathology of the structural hemoglobin between sickled cells and the vascular endothelium play a
variants is much less complex than that of the thalassemias. major role in the pathophysiology of the sickling disorders.
The majority result from missense mutations – base sub- Recently it has been demonstrated that nitric oxide may
stitutions that produce a codon change which encodes a also play a role in some of the vascular complications of
Beginnings: the molecular pathology of hemoglobin 17

this disease. It has been found that nitric oxide reacts much substitution of a tyrosine for either the proximal or distal
more rapidly with free hemoglobin than with hemoglobin histidine residue in the α or β chain.
in erythrocytes, and therefore it is possible that such decom-
partmentalization of hemoglobin into plasma, as occurs in
sickle cell disease and other hemolytic anemias, diverts nitric Molecular aspects of the high
oxide from its homeostatic vascular function. frequency of the hemoglobin variants
The sickling disorders are discussed in detail in Chap-
ter 14, Hemoglobinopathies due to Structural Mutations. The inherited disorders of hemoglobin are by far the com-
monest monogenic diseases. It is estimated that between
Unstable hemoglobin variants 300 000 and 400 000 babies are born with these conditions,
mainly across the tropical belt or in countries with large num-
There is a variety of different mechanisms underlying bers of immigrants from this region. Work over many years
hemoglobin stability resulting from amino acid substitu- has shown that the major reason for this high frequency is
tions in different parts of the molecule. The first is typi- heterozygote protection against malaria infection, particu-
fied by amino acid substitutions in the vicinity of the heme larly that due to Plasmodium falciparum. Heterozygote pro-
pocket, all of which lead to a decrease in stability of the tection raises the frequency of births of heterozygotes until an
binding of heme to globin. A second group of unstable vari- equilibrium is achieved by the early loss of the homozygotes
ants results from amino acids that simply disrupt the sec- for many of these diseases. In the case of α+ thalassemia, pre-
ondary structure of the globin chains. About 75% of globin sumably because homozygotes have an extremely mild phe-
is in the form of α helix, in which proline cannot partici- notype, the gene frequency is by far the commonest for any
pate except as part of one of the initial three residues. At least monogenic disorder, in some parts of the world almost reach-
11 unstable hemoglobin variants have been described that ing fixation.
result from the substitution of proline for leucine, five that are The mechanism for the protective effect of heterozygotes
caused by the substitution of alanine by proline, and three in against malaria is still not completely understood, although
which proline is substituted for histidine. Another group of there is extensive evidence that in most cases it may be
variants that causes disruption of the normal configuration multifactorial. In the case of the sickle cell trait, there is
of the hemoglobin molecule involves internal substitutions evidence for impairment of invasion and growth of the
that somehow interfere with its stabilization by hydropho- malarial parasites under conditions of low oxygen tension
bic interactions. Finally, there are two groups of unstable and enhanced removal of the parasite from infected red
hemoglobins that result from gross structural abnormalities cells. There is also evidence for a reduced display of the
of the globin subunits; many are due to deletions involving parasite-encoded protein P. falciparum Erythrocyte Protein-
regions at or near interhelical corners. A few of the elongated 1 (PfEMP1), which is involved in the sequestration of mature
globin chain variants are also unstable. parasites in small blood vessels and hence damage to tissues,
particularly the brain. There is also evidence for improved
Abnormal oxygen transport acquisition of malaria-specific immunity in heterozygotes.
There is a family of hemoglobin variants associated with high Rather similar findings have been observed in the case of
oxygen affinity and hereditary polycythemia. Most result carriers for HbC and for impairment of P. falciparum red
from amino acid substitutions that affect the equilibrium cell invasion and growth in heterozygotes for HbE. In the
between the R and T states (see Structure and Function). Thus, case of the α thalassemias, there appears to be reduced
many of them result from amino acid substitutions at the α1 – pathogenicity due to reduced cytoadherence or rosetting, a
β2 interface, the C-terminal end of the β chain, and at the process whereby uninfected red cells aggregate round cells
2,3-BPG binding sites. that are infected with P. falciparum. There is also evidence
of immunological priming, whereby babies are more prone
to infection by Plasmodium vivax early in life, which may
Congenital cyanosis due to hemoglobin variants
result in later protection because of cross immunity between
There is a family of structural hemoglobin variants that is P. falciparum and P. vivax.
designated Hb M, to indicate congenital methemoglobine- There are some remarkable epistatic interactions between
mia, and is further defined by their place of discovery. The the hemoglobin disorders in response to malaria. For
iron atom of heme is normally linked to the imidazole group example, while heterozygotes for HbS or α+ thalassemia are
of the proximal histidine residue of the α and β chains. protected in those who inherit the genes for both conditions,
There is another histidine residue on the opposite side, this protection is completely canceled out. Although the
near the sixth coordination position of the heme iron; this, exact mechanism is not understood, this interaction reduces
the so-called distal histidine residue, is the normal site of the amount of HbS in heterozygotes, suggesting that a
binding of oxygen. Several M hemoglobins result from the critical mass of this variant is required for protection against
18 Molecular Hematology

malaria. Several other epistatic interactions of this type have these are expressed as discrete clinical phenotypes. Perhaps
been described recently and they are providing invaluable more importantly, however, the globin field has taught us how
information about the variable distribution of these variants the interaction of a limited number of genes can produce a
in different populations. remarkably diverse series of clinical pictures, and something
of the basis for how monogenic diseases due to the same
mutation may vary widely in their clinical expression.
Molecular aspects of the prevention
and management of the hemoglobin
disorders Further reading
Over the years there have been increasingly sophisticated General background
approaches applied for screening and prenatal diagnosis of
the hemoglobin variants, particularly the thalassemias, in an Peltonen, L. and McKusick, V.A. (2001). Genomics and medicine. Dis-
attempt to reduce the numbers of births of homozygotes or secting human disease in the postgenomic era. Science 291: 1224–
1229.
compound heterozygotes. At first this was carried out by fetal
Weatherall, D.J. (2013). The role of the inherited disorders of
blood sampling in the mid-trimester of pregnancy, followed
hemoglobin, the first “molecular diseases,” in the future of human
by hemoglobin analysis by globin chain synthesis. When genetics. Annu. Rev. Genomics Hum. Genet. 14: 1–24.
DNA analysis became possible, this approach was replaced Weatherall, D.J. and Clegg, J.B. (2001). The Thalassaemia Syndromes, 4e.
by DNA studies of material obtained by chorion villous sam- Oxford: Blackwell Science.
pling. Both these approaches were invasive, of course, but Weatherall, D.J., Schechter, A.N., and Nathan, D.G. (eds.) (2013).
they have led to a dramatic reduction in the births of affected Hemoglobin and Its Diseases. Cold Spring Harbor, NY: Cold Spring
babies in many high-frequency populations. Very recently Harbor Laboratory Press.
a non-invasive approach has been established by attempt-
ing to detect homozygotes for monogenic diseases by next- Hemoglobin genetics and structural
generation sequencing of fetal blood in maternal plasma. variants
This approach is still in its early days, but some promising
results have already been obtained. Steinberg, M.H., Forget, B.G., Higgs, D.R., and Weatherall, D.J. (eds.)
Although for many years there has been a gradual (2009). Disorders of Hemoglobin, 2e. New York: Cambridge University
improvement in the symptomatic management of the Press.
hemoglobin disorders, the only approach for a cure has been
the application of bone marrow transplantation, which of Hemoglobin switching
course has the disadvantage of requiring matched donors.
Forget, B.G. (2011). Progress in understanding the hemoglobin switch.
More recently there has been a major effort directed at
N. Engl. J. Med. 365: 852–854.
the development of somatic-cell gene therapy, particularly
Sankaran, V.G. and Nathan, D.G. (2010). Reversing the hemoglobin
for the thalassemias. There has been considerable progress switch. N. Engl. J. Med. 363: 2258–2260.
toward this process using retroviral vectors and related Sankaran, V.G. and Orkin, S.H. (2013). The switch from fetal to
approaches. Although progress has been slow, successes have adult hemoglobin. Cold Spring Harb. Perspect. Med. https://doi.org/
been reported and by the use of new gene-editing techniques 10.1101/cshperspect.a011643.
further progress in the near future seems very likely. The Thein, S.L. and Menzel, S. (2009). Discovering the genetics underlying
other area of molecular therapy – that is, attempts to increase foetal haemoglobin production in adults. Br. J. Haematol. 145: 455–
the level of fetal hemoglobin in patients with different forms 467.
of β thalassemia or sickle cell anemia – was discussed earlier
in this chapter. The β thalassemias
Fucharoen, S. and Weatherall, D.J. (2013). The hemoglobin E tha-
Postscript lassemias. Cold Spring Harb. Perspect. Med. https://doi.org/10.1101/
cshperspect.a011734.
Lettre, G. (2013). The Search for Genetic Modifiers of Disease Sever-
In this short account of the molecular pathology of ity in the beta-Hemoglobinopathies. Cold Spring Harb. Perspect. Med.
hemoglobin, we have considered how mutations at or close https://doi.org/10.1101/cshperspect.a015032.
to the α or β globin genes result in a diverse family of clinical Nienhuis, A.W. and Nathan, D.G. (2013). Pathophysiology and clinical
disorders due to the defective synthesis of hemoglobin or its manifestations of the beta thalassemias. Cold Spring Harb. Perspect.
abnormal structure. Work in this field over the last 30 years Med. https://doi.org/10.1101/cshperspect.a011726.
has given us a fairly good idea of the repertoire of differ- Olivieri, N.F., Muraca, G.M., O’Donnell, A. et al. (2008). Studies in
ent mutations that underlie single-gene disorders and how haemoglobin E beta-thalassaemia. Br. J. Haematol. 141: 388–397.
Beginnings: the molecular pathology of hemoglobin 19

Quek, L. and Thein, S.L. (2007). Molecular therapies in beta- Weatherall, D.J., Williams, T.N., Allen, S.J., and O’Donnell, A. (2010).
thalassaemia. Br. J. Haematol. 136: 353–365. The population genetics and dynamics of the thalassemias. Hematol.
Thein, S.L. (2008). Genetic modifiers of the beta-haemoglobinopathies. Oncol. Clin. North Am. 24: 1021–1031.
Br. J. Haematol. 141: 357–366. Williams, T.N., Mwangi, T.W., Wambua, S. et al. (2005). Negative epis-
tasis between the malaria-protective effects of alpha+ − thalassemia
and the sickle cell trait. Nat. Genet. 37: 1253–1257.
The α thalassemias
Higgs, D.R. (2013). The molecular basis of alpha thalassemia. Cold Molecular basis of prevention and
Spring Harb. Perspect. Med. https://doi.org/10.1101/cshperspect.
management of hemoglobin disorders
a011718.
Higgs, D.R. and Gibbons, R.J. (2010). The molecular basis of alpha- Cao, A. and Kan, Y.W. (2013). The prevention of thalassemia.
thalassemia: a model for understanding human molecular genetics. Cold Spring Harb. Perspect. Med. https://doi.org/10.1101/cshperspect.
Hematol. Oncol. Clin. North Am. 24: 1033–1054. a011775.
Vichinsky, E. (2013). Natural history and clinical manifestations Lo, Y.M. and Chiu, R.W. (2010). Noninvasive approaches to prena-
of the alpha thalassemias. Cold Spring Harb. Perspect. Med. tal diagnosis of hemoglobinopathies using fetal DNA in maternal
https://doi.org/10.1101/cshperspect.a011742. plasma. Hematol. Oncol. Clin. North Am. 24: 1179–1186.
Nienhuis, A.W. and Persons, D.A. (2013). Development of gene ther-
Evolutionary background of hemoglobin apy for thalassemia. Cold Spring Harb. Perspect. Med. https://doi.org/
10.1101/cshperspect.a011833.
disorders
Orkin, S.H. and Reilly, P. (2016). MEDICINE. Paying for future success
Penman, B.S., Pybus, O.G., Weatherall, D.J., and Gupta, S. (2009). in gene therapy. Science 352: 1059–1061.
Epistatic interactions between genetic disorders of hemoglobin can
explain why the sickle-cell gene is uncommon in the Mediterranean.
Proc. Natl. Acad. Sci. U.S.A. 106: 21242–21246.
Chapter 2 Stem cells
David T Scadden1,2
1 Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Boston MA, USA
2
Center for Regenerative Medicine, Massachusetts General Hospital, Boston MA, USA

Introduction, 21 Trafficking of primitive hematopoietic cells, 29


Stem cell definitions and distinctions, 21 Manipulating HSCs for clinical use, 31
Hematopoietic stem cell concepts and their origin, 21 Summary, 34
Molecular regulation of hematopoiesis, 24 Further reading, 34

the placental tissues (Figure 2.1). Pluripotent stem cells may


Introduction give rise to any type of cell found in the body except those
of the extra-embryonic membranes. They can produce ecto-
The generation of sufficient numbers of blood cells to main-
derm, mesoderm, or endoderm cells. It has also become pos-
tain homeostasis requires the sustained production of mature
sible to create pluripotent cells by “reprogramming” mature
cells. This process, called hematopoiesis, yields approxi-
cells. This allows for pluripotent cells to be made from any
mately 1011 blood cells daily, with the capability for dra-
individual, a powerful tool for basic biology, disease mod-
matic increases in the number and subsets of cells in response
eling, and possible future cell therapies. These are called
to physiological stress. Hematopoiesis is therefore a highly
induced pluripotent stem cells (iPS). Pluripotent stem cells
dynamic process dependent upon numerous modulating fac-
not made by reprogramming include embryonic stem cells,
tors. Its prodigious production capability derives from the
isolated from the inner cell mass of the blastocyst; embry-
sustained presence of a cell type which is generally quiescent,
onic germ cells, isolated from embryonic gonad precursors;
but the descendants of which proliferate vigorously. This cell
and embryonic carcinoma cells, isolated from teratocarcino-
is the hematopoietic stem cell (HSC).
mas. Pluripotent stem cells may be maintained indefinitely in
culture under specialized conditions that prevent differenti-
ation. In particular, embryonic stem cells have been used to
Stem cell definitions and distinctions generate “knockout” mice, animals harboring targeted gene
disruptions via homologous recombination that permit the
Stem cells derive their name from their ability to produce in vivo study of individual gene function. Lastly, multipo-
daughter cells of different types. Stem cells are defined by a tent stem cells, such as the HSCs of the bone marrow, are
combination of the traits of self-maintenance and the ability capable of giving rise to multiple mature cell types, but only
to produce multiple, varied offspring. Putting this in more those of a particular tissue, such as blood. Multipotent stem
biological terms, stem cells have the unique and defining cells are found in adults, perhaps in all tissue, and function
characteristics of self-renewal and differentiation into multi- to replace dead or damaged tissue. Such stem cells are com-
ple cell types. Thus, with each cell division there is an inher- monly referred to as “adult” stem cells.
ent asymmetry in stem cells that is generally not found with
other cell types.
While their name implies that stem cells have specific Hematopoietic stem cell concepts and
intrinsic characteristics, there are multiple different types of their origin
stem cells, each defined by their production ability. Totipo-
tent stem cells are capable of generating any type of cell in the
The cellular compartment model
body, including those of the extra-embryonic tissues, such as
The short-lived nature of most blood cells was first deduced
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. in the 1960s using thymidine labeling of reinfused blood.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. These studies demonstrated that the maintenance of normal

21
22 Molecular Hematology

Inner cell mass

Fertilization Totipotent cells Blastocyst Fetal tissues Adult tissues

Pluripotent cells Multipotent cells

Embryonic stem “Tissue” or “adult”


(ES) cells stem cells

Fig. 2.1 Sources and types of stem cells.


Source: Adapted with gratitude from the National Institutes of Health Stem Cell Information website.

numbers of blood cells in the adult requires a process with arising from committed progenitors with little capacity
the capacity to briskly generate large numbers of mature for self-renewal, gives rise to colonies that peak in size by
cells along multiple blood lineages. The early history of day 8, while a second, arising from a more primitive cell
HSC research was largely shaped by cellular biology and that is capable of self-renewal, yields colonies that peak in
animal transplantation experiments. It was advanced by size at day 12. To further highlight the complexity of the
experiments in the early 1960s demonstrating that injection hematopoietic hierarchy, a rarer population of hematopoietic
of marrow cells could generate large hematopoietic colonies cells provides longer-term repopulation of an irradiated host
in the spleen of irradiated mice. Such colonies were the than CFU-S. These long-term repopulating cells have the
clonal progeny of single initiating cells, termed colony- capacity for sustained self-renewal and were considered the
forming units, spleen (CFU-S), and contained hematopoietic true adult stem cells. The presence of stromal cells in the
populations of multiple lineages. CFU-S were further cultures is important for the long-term culture of CFU-S
transplantable, demonstrating their self-renewing nature. and repopulating cells. Cells capable of long-term survival in
HSCs are a minor component of marrow cells, able both culture on stroma were termed long-term culture-initiating
to generate large numbers of progeny differentiated along cells (LTC-ICs) and cobblestone area-forming cells (CAFCs).
multiple lines and to renew themselves. These multipotential cell types were considered more
The field was further advanced by the use of in vitro cell primitive than lineage-committed progenitor cells, but more
culture techniques; in particular, solid-state cultures of mar- mature than long-term repopulating cells.
row and spleen cells furthered understanding of the colony- Thus, a more complex version of the compartmental
forming capacity of individual hematopoietic cells. The orig- model has emerged. This provides a model with two pop-
inal technique demonstrated clonal colonies of granulocytes ulations of stem cells, the most immature group consisting
and/or macrophages, termed in vitro colony-forming cells of long-term repopulating cells and a more mature group
(CFCs), which are now considered lineage-committed pro- of short-term repopulating cells. These groupings are simple
genitor cells. These cells could be separated from whole mar- models for what is likely to be a continuum of cells with self-
row cells and from CFU-S, were more numerous than CFU-S, renewal and multipotent differentiation capacity. The long-
and could be detected in splenic colonies as the progeny term repopulating cells also tend to be more quiescent, serv-
of CFU-S. These observations gave rise to the concept of ing as a deep reserve for blood cell production in times of
the three-compartment model of hematopoiesis, the compart- physiological stress. More mature progenitors (also called
ments being stem cells, progenitor cells, and dividing mature colony-forming cells or CFCs) have the capacity to give rise to
cells in increasing numbers; each compartment consists of colonies of clonal origin in semisolid media containing fully
the amplified progeny of cells in the preceding compartment. mature cells, permitting their analysis. A more mature set of
Subsequent analyses have added further complexity to precursor cells constitutes the bulk of bone marrow cells and
the compartment model of hematopoiesis. The term CFU-S has unique, identifiable features by light microscopy. Rapid
describes at least two groups of precursor cells. One group, division of precursor cells culminates in the production of
Stem cells 23

Hematopoiesis

Stem cells Progenitor cells Precursor cells Mature cells

Microenvironment
Long-term Short-term
LTC-IC/CAFC
Fig. 2.2 Schematic view of hematopoiesis. See text CFU-S
for definition of abbreviations.
Source: Modified from Figure 12.1 in Hematology: Basic HPP-CFC
CFU-C
Principles and Practice, 3rd edn (ed. R. Hoffman), 2000,
with permission from Elsevier. CFU-F/RF CFU-Mix

mature cells. Although hematopoiesis proceeds according to suggest that hematopoiesis is a random, stochastic process.
this orderly scheme (Figure 2.2), special consideration must The stochastic theory is based on in vitro observations that
be given to the development of T and B lymphocytes. These multilineage colonies develop variable combinations of
cells are generated in the thymus and bone marrow, respec- lineages, and that such lineage choices occur independently
tively, by a similar hierarchical process. Mature T and B of external influences.
lymphocytes enter peripheral lymphoid organs, where they Similar controversy exists regarding the role of cytokines
encounter relevant antigens, leading to the production of new in cell lineage determination. An instructive model suggests
cells from reactivated mature cells. This process amplifies the that cytokine signaling forces the commitment of prim-
de novo bone marrow formation of T and B lymphocytes. In itive cells along a particular lineage. Ectopic expression
addition, some members of this type of cell, memory T or B of the granulocyte macrophage colony-stimulating factor
lymphocytes, are capable of sustained self-renewal, serving, (GM-CSF) receptor in a common lymphoid progenitor
in effect, as unipotent stem cells. Their inability to produce (CLP) population was capable of converting the cells from a
multiple different types of daughter cells distinguishes them lymphoid to a myeloid lineage. The influence of the GM-CSF
from multipotent HSCs. receptor was sufficiently dominant to change the entire
In summary, the compartment model has given rise to differentiation program of cells, but only the CLP stage of
terms that are generally applied to cells of hematopoietic ori- development. A permissive model postulates that decisions
gin. HSCs are those that are multipotent and self-renewing. about cell fate occur independently of extracellular signals.
Progenitor cells have limited ability to self-renew and are This model suggests that cytokines serve only to allow certain
likely to be unipotential or of very limited multipotential. lineages to survive and proliferate. Evidence supporting this
Precursor cells are restricted to a single lineage, such as neu- model is provided by the ectopic expression of growth recep-
trophil precursors, and are the immediate precursors of the tors in progenitor cells. Expression of the erythropoietin
mature cells found in the blood. The mature cells are generally receptor in a macrophage progenitor results in macrophage
short-lived and preprogrammed to be highly responsive to colony formation, whereas expression of the macrophage
cytokines, while the stem cells are long-lived, cytokine resis- colony-stimulating factor (M-CSF) receptor in an erythroid
tant, and generally quiescent. progenitor results in erythroid rather than macrophage
colony formation. Replacing the thrombopoietin receptor
(c-mpl) with a chimeric receptor consisting of the extra-
Models of lineage commitment
cellular domain of c-mpl with the cytoplasmic domain of
Several theories have emerged to describe the manner by the granulocyte colony-stimulating factor (G-CSF) receptor
which HSCs undergo lineage commitment and differentiate. results in normal platelet counts in homozygous “knock-in”
Some studies support a deterministic theory whereby the mice. Therefore, the instructive and permissive models may
stem cell compartment encompasses a series of closely both be correct, but at different stages of hematopoietic
related cells maturing in a stepwise process. Other studies differentiation. Cells at earlier points in the differentiation
24 Molecular Hematology

cascade may be more susceptible to fate-altering stimuli, to lose the function of specific genes have been used to
while more committed cells may be irreversibly deter- define the impact of those genes on hematopoiesis. Loss-
mined, with only proliferation, cell death, or the rate of of-function studies involving the transcription factors
differentiation susceptible to influence by external signals. c-Myb, AML1 (CBF2), SCL (tal-1), LMO2 (Rbtn2), GATA-2,
and TEL/ETV6 have demonstrated global effects on all
hematopoietic lineages. Stem cells in animals deficient in
Molecular regulation of these molecules fail to establish definitive hematopoiesis.
hematopoiesis Similar methods have indicated that some regulators have
different roles at different times in development. For example,
The molecular nature of stem cell regulatory pathways has the gene product of transcription factor SCL is absolutely
been determined using a variety of genetic approaches, required for establishing HSCs. Unexpectedly, there is not
including genetic loss-of-function and gain-of-function stu- a requirement for SCL once the stem cell pool is present
dies. These have provided several important concepts regard- in the adult. Rather, SCL is required only for erythroid
ing the molecular control of hematopoiesis. First, some genes and megakaryocytic homeostasis. Therefore, transcription
have binary functions and exert an effect by being above a factor regulation of the stem cell compartment can be highly
threshold of expression, rendering their targets “on.” Other dependent on the stage of development of the organism.
genes function in a continuum and have different effects at Loss-of-function studies have also proved useful in identi-
different levels. Secondly, while perturbations in single genes fying lineage-specific transcription factors. Mice genetically
may have dramatic cellular effects, gene products often func- deficient in the transcription factor Ikaros lack T and B lym-
tion in complexes and, in the case of transcription regulators, phocytes and natural killer cells, but maintain erythropoiesis
are only active if they have access to sites of open chromatin. and myelopoiesis. Notch is required for T lineage induction
Finally, signal integration often depends on the assembly and shifts differentiation away from B cell development.
of large signaling complexes and the spatial proximity of Further, losing expression of some genes can enable cells to
molecules to facilitate interaction is therefore important. revert in differentiation. For example the Pax-5 transcription
factor is essential for B cell maturation, and loss of it results
Cell-intrinsic regulators of hematopoiesis in cells that have already rearranged their B cell receptor
or immunoglobulin locus showing up in the T and NK cell
Cell cycle control compartment. The critical roles for specific transcription
The quiescent nature of HSCs is supported by their low level factors in establishing and maintaining cell fate are shown in
of staining with DNA and RNA nucleic acid dyes, which is Figure 2.3. The ability of transcription factors to affect gene
consistent with low metabolic activity. These studies have expression is also highly dependent upon epigenetic features
indicated a heterogeneity among stem cells with a subgroup of a particular gene locus. The histone marks and chromatin
that is deeply quiescent. Various studies have sought to deter- accessibility regulators that participate in the epigenome are
mine the cell-intrinsic regulators of hematopoiesis involved beyond the scope of this chapter but should be recognized
in HSC cycle control. as critical aspects of gene expression and therefore, cell
RNA analysis has been used to profile pertinent tran- state.
scription factors and other molecules in HSCs induced
to differentiate along various lineages by the application Cell-extrinsic regulators
of cytokines. Elevated levels of cyclin-dependent kinase Ultimately, hematopoietic stem and progenitor cell decisions
inhibitors (CDKIs) have been observed, suggesting that are regulated by the coordinated action of transcription fac-
CDKIs present in HSCs function to exert a dominant tors as modified by extracellular signals. Extracellular sig-
inhibitory tone on HSC cell cycling. The bone marrow nals in the form of hematopoietic growth factors are medi-
of some mouse strains deficient in CDKI p57(CDKN1C), ated via cell surface hematopoietic growth factor receptors.
p21(CDKN1A), or p18(CDKN2C) have increased HSC cell Hematopoietic growth factors exert specific effects when act-
cycling, suggesting that these CDKIs function as domi- ing alone and may have different effects when combined
nant negative regulators of HSC proliferation. Other CDKIs, with other cytokines. There are at least six receptor super-
such as p27(CDKN1B), may serve as negative regulators of families, and most growth factors are members of the type
hematopoietic progenitor cells. I cytokine receptor family. The effects of various cytokines
during myelopoiesis are illustrated in Figure 2.4.
Self-renewal, commitment, and lineage determination
Type I cytokine receptors
Experimental results involving transcription factors have
demonstrated cell-intrinsic roles in both global and lineage- Type I receptors do not possess intrinsic kinase activity, but
specific hematopoietic development. Mice engineered lead to phosphorylation of cellular substrates by serving
Stem cells 25

NK cell
Pro-T

T cell
Lymphoid pathway IL-7R+
CLP SCL (–) C/EBPα (–)
GATA-2 (–) PU.1 (–)
NF-E2 (–) Aiolos (++)
IL-7R+ GATA-1 (–) GATA-3 (++)
c-mpl–
Pro-B
SCL (–) C/EBPα (–)
GATA-2 (–) PU.1 (+)
HSC B cell
NF-E2 (–) Aiolos (+)
IL-7R+
GATA-1 (–) GATA-3 (+)
SCL (–) C/EBPα (–)
GATA-2 (–) PU.1 (+)
NF-E2 (–) Aiolos (++)
LT-HSC ST-HSC GATA-1 (–) GATA-3 (–)
SCL (++) C/EBPα (±) GMP
Myeloid pathway
GATA-2 (++) PU.1 (±) Monocyte
NF-E2 (–) Aiolos (±) CMP
IL-7R–
GATA-1 (±) GATA-3 (±) Epo-R– Granulocyte
c-mpl+
SCL (+) C/EBPα (++)
GATA-2 (–) PU.1 (±)
SCL (++) C/EBPα (±) NF-E2 (–) Aiolos (–)
GATA-2 (+) PU.1 (±) GATA-1 (–) GATA-3 (–)
NF-E2 (+) Aiolos (±) MEP
GATA-1 (+) GATA-3 (–) Megakaryocyte
Epo-R+
Erythrocyte
SCL (++) C/EBPα (–)
GATA-2 (++) PU.1 (±)
NF-E2 (++) Aiolos (–)
GATA-1 (++) GATA-3 (–)

Fig. 2.3 Transcription factors active at various stages of hematopoiesis. CLP, common lymphoid progenitor; CMP, common myeloid
progenitor; GMP, granulocyte monocyte progenitor; MEP, megakaryocyte erythrocyte progenitor; NK, natural killer.
Source: Redrawn from Akashi, K., Traver, D., Miyamoto, T., Weissman, I.L. (2000). A clonogenic common myeloid progenitor that gives rise to all
myeloid lineages. Nature 404: 193–197, with permission of Spring Nature.

as docking sites for adapter molecules with kinase activity. Receptor tyrosine kinases
Examples of receptors in this family include leukemia
Receptors with intrinsic kinase activity are of considerable
inhibitory factor (LIF), interleukin (IL)-1, IL-2, IL-3, IL-4,
relevance to hematology because their ligands are growth fac-
IL-5, IL-6, IL-7, IL-9, IL-13, IL-18, GM-CSF, G-CSF, erythro-
tors, but also because abnormalities of them can result in
poietin, prolactin, growth hormone, ciliary neurotrophic
unregulated activation. For example, internal tandem dupli-
factor, and c-mpl. These receptors share several features,
cations of the Flt3 receptor are associated with acute myeloid
including enhanced binding and/or signal transduction
leukemia (AML) and activating mutations of c-kit play a
when expressed as heterodimers or homodimers, four cys-
key role in systemic mastocytosis and core binding factor
teine residues and fibronectin type III domains in the extra-
AMLs. Loss of c-fms, the receptor for M-CSF, has been asso-
cellular domain, WSXWS ligand-binding sequence in the
ciated with myelodysplasia and a predisposition to AML. The
extracellular cytokine receptor domains, and lack of a known
availability of agents targeting tyrosine kinases also makes
catalytic domain in the cytoplasmic portion. Another shared
these receptors of particular interest in hematologic disease.
feature of receptors in this family is the ability to transduce
Details of receptor–ligand pairs are provided in Table 2.1.
signals that prevent programmed cell death (apoptosis).
Protein serine–threonine kinase receptors
Type II cytokine receptors
This family includes the 30 members of the transforming
This class includes the receptors for tissue factor, IL-10, and growth factor (TGF)-β superfamily, which bind to their
interferon (IFN)-γ. This family contains a type III fibronectin receptors as homodimers. Members of this family include
domain in the extracellular domain, like the type I family. the three TGF-β receptors: type I (TbRI, 53 kDa), type II
26 Molecular Hematology

G-CSF, IL-1, IL-6, IL-10, IL-11, IL-12, IL-13


HPP-CFC
b-FGF, HGF, LIF, SCF/KL, FLT3 Ligand, TPO
Mixed progenitor Pluripotent
CFU-GEMM stem cell
cell
FLT3, SCF, IL-3, IL-6, GM-CSF, G-CSF SCF IL-4
Myelomonocytic IL-3 IL-5
progenitor G-CSF
CFU-GM GM-CSF
SCF, IL-3, IL-6, G-CSF
GM-CSF, CSF-1 IL-5

CFU-M CFU-G CFU-Eos CFU-Baso


NGF

CSF-1, IL-3 G-CSF IL-5 IL-10, IL-9,


GM-CSF GM-CSF GM-CSF CSF
IL-4
Monoblast Myeloblast

CSF-1 G-CSF
GM-CSF GM-CSF
Promonocyte Myelocyte

IL-5, IL-3, IL-4


GM-CSF
CSF-1 G-CSF
Basophil,
Monocyte Neutrophil Eosinophil mast cell
NGF

Chemokines
MCP-1-3, IP-10, Rantes, MIP-1α IL-8, NAP-2, Gro-α Eotaxin, MCP-4 Rantes IL-8, MCP-1, 3
Fig. 2.4 Cytokines active at various stages of hematopoiesis. See text for definition of abbreviations.
Source: Modified from Figure 16.3 in Hematology: Basic Principles and Practice, 3rd edn (ed. R. Hoffman), 2000, with permission from Elsevier.

(TbRII, 75 kDa), and type III (TbRIII, 200 kDa). Members downstream mediators act as braking factors for a number of
of this family have a profound inhibitory effect on the cell types and are frequently inactivated by somatic mutation
growth and differentiation of hematopoietic cells and on in a number of cancers.
auxiliary hematopoietic cells. Binding of TFG-β requires
TbRII. After binding, signal transduction occurs via acti-
Chemokine receptors
vation of serine–threonine kinase cytoplasmic domains of
the receptor chains, which results in the phosphorylation of This family comprises seven transmembrane-spanning
Smad molecules on serines. Phosphorylated Smad complexes G-protein-coupled receptors that influence both cell cycle
translocate to the nucleus, where they induce or repress and cellular movement, or chemotaxis. These receptors are
gene transcription. TGF-β is the best-characterized negative divided into three families, α or CXC, β or CC, and γ or C, on
regulator of hematopoiesis. It inhibits mitosis by inducing the basis of variability in cysteine residues. The best charac-
cell cycle inhibitors such as p21(CDKN1A), p27(CDKN1C), terized is CXCR4, which mediates homing and engraftment
and p16(CDKN2A), inhibiting the cyclin-dependent kinases of HSCs in bone marrow and is critical to hematopoietic
Cdk4 and Cdk6, and inducing phosphorylation of the development. IL-8 and macrophage inflammatory protein
retinoblastoma protein. The TGF-β receptor family and its (MIP)-1α act as inhibitors of progenitor cell proliferation.
Stem cells 27

Table 2.1 Factors affecting hematopoietic control

Growth
factor
Growth factor receptor Produced by Bioactivity Deficient states

Erythropoiesis
EPO (erythropoietin) EPO-R Adult kidney Stimulates clonal growth of CFU-E and BFU-E subsets Anemia
Liver during Suppresses erythroid progenitor cell apoptosis
development Induces bone marrow release of reticulocytes
Induces erythroid globin synthesis
SF (steel factor), kit c-kit (CD117) Bone marrow Promotes proliferation and differentiation of pre-CFC cells Anemia
ligand, mast cell perivascular Acts synergistically with IL-3, GM-CSF, and TPO to support Mast cell deficiency
growth factor mesenchymal growth of CFU-GEMM, BFU-E, and CFU-Mk
cells Expansion of committed progenitor cells in vivo
Endothelial cells Stimulates mast cell hyperplasia, degranulation, and
IgE-dependent mediator release
IGF-1 (insulin-like IGF-1R Liver Induces DNA synthesis and has anti-apoptotic effects in Growth retardation,
growth factor, erythroid progenitors neurological
somatomedin C) Simulates erythroid colony growth in the absence of EPO defects,
at high doses homozygous
deficiency lethal
Granulopoiesis
G-CSF (granulocyte G-CSFR Monocytes, Stimulates growth of progenitors committed to neutrophil Neutropenia, failure to
colony-stimulating macrophages, differentiation develop neutrophilic
factor) endothelial cells, Activates neutrophil phagocytosis leukocytosis in
fibroblasts Stimulates quiescent HPCs to enter G1 /S response to
Stimulates mobilization of HSCs and HPCs from bone infection
marrow to periphery
GM-CSF GM-CSFR Mast cells, T Stimulates multilineage hematopoietic progenitor cells Susceptibility to
(granulocyte lymphocytes, Stimulates BFU-E and granulocyte, macrophage, and infections caused by
macrophage endothelial cells, eosinophil colony growth obligate
colony-stimulating fibroblasts, thymic intracellular
factor) epithelial cells organisms
M-CSF (macrophage c-fms Monocytes, Induces monocyte/macrophage growth and differentiation Macrophage and
colony-stimulating macrophages, and activation osteoclast
factor) fibroblasts, deficiency,
epithelial cells, hematopoietic
vascular failure
endothelium,
osteoblasts
Thrombopoietin c-mpl Bone marrow Stimulates in vitro growth of CFU-Mk, megakaryocytes, Thrombocytopenia
stroma, spleen, and platelets
renal tubule, liver, Stimulates clonal growth of individual CD34+ CD38− cells
muscle, brain Synergizes with SF, IL-3, and FL
Primes response to platelet activators ADP, epinephrine,
and thrombin, but no effect on aggregation
IL-5 IL-5R T lymphocytes Stimulates eosinophil production and activation Inability to mount
Activates cytotoxic T cells eosinophilic
Induces immunoglobulin secretion response
IL-11 IL-11R Fibroblasts, bone Acts synergistically with IL-3 or SF to stimulate the clonal No hematological
marrow stroma growth of erythroid (BFU-E and CFU-E) and primitive defect
megakaryocytic (BFU-Mk) progenitors
Shortens duration of G0 of HPCs
Quickens hematopoietic recovery after chemotherapy and
radiation

(continued)
28 Molecular Hematology

Table 2.1 (Continued)

Growth
factor
Growth factor receptor Produced by Bioactivity Deficient states

Lymphopoiesis
IL-7 IL-7R Bone marrow Induces clonal growth of pre-B cells B- and T-cell
stroma, spleen, Induces growth of pre-T cells lymphopenia
thymus
IL-2 IL-2R T lymphocytes Induces proliferation and activation of T cells, B cells, and Fatal
NK cells immunoproliferative
disorder, loss of
self-tolerance
IL-15 IL-15R Monocytes, Induces proliferation and activation of T cells, B cells, and
macrophages, NK cells
epithelial cells,
skeletal muscle
cells, bone
marrow and
thymic stroma
IL-4 IL-4R T lymphocytes Induces proliferation of activated B cells Defective T helper cell
Inhibits IL-2-stimulated proliferation of B cells responses
Induces T-cell proliferation
IL-10 Inhibits monocyte/macrophage-dependent synthesis of
Th1- and Th2-derived cytokines
Early-acting factors
IL-3 IL-3R T lymphocytes, mast Stimulates multilineage colony growth and growth of No hematopoietic
cells primitive cell lines with multilineage potential defect in steady
Stimulates BFU-E proliferation state, deficient
delayed-type
hypersensitivity
FLT3-ligand (FL) FLT-3R, flk2 Most tissues, Weak colony-stimulating activity alone, but synergizes Reduction in pro-B
including spleen, with IL-3, GM-CSF, SF, IL-11, IL-6, G-CSF, IL-7, and others cells, pre-B cells,
lung, stromal Augments retroviral transduction of HSCs when added to B-cell
cells, peripheral cytokine cocktails colony-forming
blood Mobilizes HSCs to periphery weakly alone, but adds potential, reduced
mononuclear cells greatly to G-CSF repopulating
capacity of stem
cells
IL-9 (T-cell growth IL-9R T lymphocytes Stimulates growth of BFU-E when combined with EPO
factor) Stimulates clonal growth of fetal CFU-Mix and CFU-GM
IL-6 IL-6R Macrophages, Synergistic with IL-3 for CFU-GEMM colony growth Reduced HSC and
endothelial cells, Synergistic with IL-4 in inducing T-cell proliferation and progenitor cell
fibroblasts, T colony growth survival, reduced
lymphocytes Synergistic with M-CSF in macrophage colony growth T-cell numbers,
Synergistic with GM-CSF in granulocyte colony growth reduced
Co-induces differentiation of B cells proliferation and
maturation of
erythroid and
myeloid cells

BFU-E, burst-forming unit, erythroid; CFU-mix, colony-forming unit, mix; CFU-Mk, colony-forming unit, megakaryocyte; CFU-GM, colony-forming
unit, granulocyte/macrophage; CFU-GEMM, colony-forming unit, granulocyte, erythroid, monocyte, megakaryocyte.
Stem cells 29

Members of this receptor family have also been implicated of cells to arrive at a particular environment, while retention
in cancer metastasis and the entry of HIV-1 into cells. is their ability to remain in such an environment after arrival.
Lastly, engraftment reflects the ability of cells to divide and
Tumor necrosis factor receptor family form functional progeny in a given microenvironment.
Much has been learned about trafficking from the ontogeny
Members of the tumor necrosis factor receptor (TNFR) fam- of mouse and human HSCs.
ily have varied effects, some having the ability to induce pro-
grammed cell death and others stimulating mesenchymal
cells to secrete hematopoietic growth factors. These recep- Hematopoietic ontogeny
tors contain Cys-rich extracellular domains and 80-amino In both humans and mice, hematopoiesis occurs sequen-
acid cytoplasmic “death domains,” which are required for tially in distinct anatomical locations during development.
transducing the apoptotic signal and inducing NF-κB acti- These shifts in location are accompanied by changes in the
vation. Members of this family include TNFR1, TNFR2, fas, functional status of the stem cells and reflect the changing
CD40, nerve growth factor (NGF) receptor, CD27, CD30, needs of the developing organism. These are relevant for
and OX40, each with at least one distinct biological effect. adult hematopoiesis, since they offer insight into how the
blood production process can be located in different places
Components of the hematopoietic with distinct regulation.
microenvironmental niche There are essentially five sites of blood cell forma-
While soluble factors influence stem cell fate, these factors tion recognized in mammalian development, and these
are seen by the cell in the context of the cell–cell contact are best defined in the mouse. At about embryonic day
among heterologous cell types and cell–matrix contact that 7.5 (E7.5), blood and endothelial progenitors emerge in
comprise the three-dimensional setting of the bone marrow. the extra-embryonic yolk sac blood islands. The yolk sac
What actually constitutes the critical microenvironment for supports the generation of primitive hematopoietic cells,
hematopoiesis is surprisingly poorly defined. The ability of which are primarily composed of nucleated erythrocytes and
primitive cells to mature in vitro in complex stromal cul- macrophages. The macrophages generated in the yolk sac are
tures suggests that at least some elements of the regulatory generally tissue-resident cells that durably populate tissues
milieu of the bone marrow can be recapitulated ex vivo. Stud- throughout adult life. Examples are the microglia of the brain
ies based solely on ex vivo systems are suspect, however, as no and the Kupffer cells of the liver. More sustained or definitive
fully satisfactory re-creation of stem cell expansion or self- hematopoiesis derives from the aorta–gonad–mesonephros
renewal has been defined. Recognizing this limitation, it has (AGM) region, as has been defined in both the mouse (E8.5)
been determined that mesodermal cells of multiple types are and the human. Current data suggest that yolk sac cells do
needed to enable hematopoietic support. These include mul- not seed the AGM region, but rather hematopoietic cells arise
tiple mesenchymal stromal cells, adipocytes, hematopoietic there de novo. The placenta may also be a site of de novo HSC
cells, and endothelium. HSCs tend to be perivascular in loca- generation. By E10 in the mouse, the fetal liver assumes the
tion endogenously and in periendosteal regions when trans- primary role of cell production. By E14 in the mouse and
planted. The most immature hematopoietic stem/progenitor the second trimester of human gestation, the bone marrow
cells (HSPCs) have been reported to be adjacent to periar- becomes populated with HSCs and it takes over blood cell
teriolar mesenchymal cells, while more mature cells of lym- production, along with the spleen and thymus. The spleen
phoid lineage are nearby to more mature osteolineage cells. remains a more active hematopoietic organ in the mouse than
There appears to be spatial organization of the bone marrow in the human. Stem cell proliferation is very active in the
with multiple niches for particular subsets of HSPC, though AGM region and fetal liver. While it is also initially robust
the precise details of the marrow microenvironment are still in the bone marrow, there is a dramatic shift to quiescence
being defined. shortly after residence in the bone marrow, experimentally
defined as after four weeks in the mouse.

Trafficking of primitive
Homing and engraftment of HSCs
hematopoietic cells
following infusion
The migratory behavior characteristic of primitive Despite the use of HSC transplantation for over three
hematopoietic cells is an area of intense research because of decades, the exact mechanisms whereby bone marrow cells
its relationship to bone marrow transplantation. Trafficking home to the bone marrow are not fully understood. Other
of HSCs can be divided into the components of homing, than lectins, no adhesion receptors have been identified that
retention, and engraftment. Homing describes the tendency are exclusively present on HSCs. Furthermore, no adhesion
30 Molecular Hematology

ligands, other than hemonectin, have been identified that are However, a population of CD34+ cells capable of forming
exclusively present in the bone marrow microenvironment. CFCs and LTC-ICs and of long-term repopulation may be
When first infused, HSCs lodge in the microvasculature found circulating in the peripheral blood, and these may
of the lung and liver; they then colonize the bone marrow, increase after physiological stressors such as exercise, stress,
first passing through marrow sinusoids, migrating through and infection. Animal studies have suggested that a rela-
the extracellular space of the bone marrow, and ultimately tively large number of bone marrow–derived stem cells cir-
settling in stem cell niches. Passage through the endothelial culates during the course of a day, and that these cells peri-
barriers at first requires tethering, through endothelium- odically transit back into an engraftable niche to establish
expressed addressins that bind hematopoietic cell selectins, hematopoiesis. Defining the processes involved is important
and this is followed by firm attachment mediated by in guiding new approaches to peripheral blood stem cell
integrins. mobilization for transplantation.
Selectins are receptors expressed on hematopoietic cells Examining mice in which specific adhesion molecules
(L- and P-selectins) and endothelium (E- and P-selectins). have been deleted has revealed several key molecular deter-
They have long extracellular domains containing an minants of stem cell localization in the bone marrow. Among
aminoterminal Ca2+ -binding domain, an epidermal growth these, the chemokine receptor CXCR4 has perhaps the most
factor domain, and a series of consensus repeats similar to striking phenotype. In the absence of this receptor, stem
those present in complement regulatory molecules. Ligands cells fail to traffic from the fetal liver to the bone marrow
for selectins are sialylated fucosyl glucoconjugates present on in development. Partly because of these studies, others have
endothelium. L-selectin is present on CD34+ hematopoietic defined that CXCR4 is relevant for the engraftment of trans-
progenitors, while L-selectin and P-selectin are present on planted stem cells, and that the modulation of CXCR4 sig-
more mature myeloid and lymphoid cells. Tethering by naling can affect adult stem cell localization in the bone
selectins allows integrin-mediated adhesion to the endothe- marrow versus peripheral blood. As described, the integrin
lium. Integrins, a family of glycoproteins composed of α and selectin families are also important molecular partici-
and β chains responsible for cell–extracellular matrix and pants in stem cell location. For example, HSCs from animals
cell–cell adhesion, not only provide firm attachment, but also that are heterozygous deficient for β1 integrin cannot com-
allow migration of hematopoietic cells through the endothe- pete with wild-type cells for the colonization of hematopoi-
lium and bone marrow extracellular space. The functional etic organs. Pre-incubation of HSCs with α4 integrin anti-
state of integrins is only loosely tied to their expression level bodies prior to transplantation results in decreased bone
and depends on ligand affinity modulation, regulated by the marrow and increased peripheral recovery of cells, while
β subunit in response to cytokines and other stimuli. the continued presence of α4 antibodies prevents engraft-
The process of migration depends on the establishment ment. Evidence for selectin involvement has been demon-
of adhesion at the leading edge of the cell and simultaneous strated in animals deficient of single selectins or combi-
release at the trailing edge. The rate of migration depends nations of selectins. Endothelial P-selectin mediates leuko-
on dynamic changes in the strength of the cell–ligand inter- cyte rolling in the absence of inflammation, while L-, P-,
actions, which is dictated by the number of receptors and and E-selectins contribute to leukocyte rolling in the set-
their affinity state and the strength of the adhesion receptor– ting of inflammation. L-selectin is important in lymphocyte
cytoskeleton interactions. Cell–ligand interaction strength homing. Transplantation studies performed in animals defi-
may also be modulated by chemokines that provide direc- cient in P- and E-selectins demonstrate severely decreased
tional cues for cell migration based on ligand gradients. Thus, engraftment due to impaired homing, an effect that is further
successful engraftment relies not only on the presence of sev- compromised by blocking vascular cell adhesion molecule
eral different adhesion receptors, but also on the presence of (VCAM)-1.
chemokine gradients that encourage directional movement Mature hematopoietic cells are thought to migrate from
of the cells. Chemokines such as CXCL12, the ligand for the the marrow to the blood by similar mechanisms, though
receptor CXCR4, are localized discontinuously in the bone these are not well defined. One purported mechanism is a
marrow microvasculature and provide site-specific guidance shift in expression from molecules thought to interact with
for HSPC migration and localization. stromal proteins to those that interact with endothelium.
For example, myeloid progenitors express functional α4β1
and α5β1 integrins that act to ensure that these progenitors
Egress of HSCs from bone marrow under
are retained in the bone marrow through interactions
physiological conditions
with VCAM and fibronectin. Mature neutrophils, in con-
The majority of primitive HSCs are resident within the bone trast, express β2 integrins that permit interaction with
marrow space under steady-state physiological conditions. ligands, such as intercellular adhesion molecule, expressed
Stem cells 31

by endothelial cells. Mature neutrophils also express β1 Isolating stem cells for manipulation
integrins that permit interaction with collagen and laminin
Characteristics of HSCs used for isolation
present in basal membranes, perhaps regulating a progres-
sive shift in cell affinities for specific microenvironmental Physical Early attempts to isolate HSCs were based on cell
determinants that ultimately results in cell egress into the size and density. In order to clarify whether the heterogene-
blood. Mobilization of murine HSCs induced by cyclophos- ity of CFU-S was due to differences in the input cells used,
phamide or G-CSF is accompanied by changes in integrin velocity sedimentation was performed to separate cells by
expression levels and functional changes in homing, thus size, demonstrating that smaller cells were more likely to
linking cellular localization with adhesion molecule receptor produce secondary CFU-S than larger cells. HSCs are similar
expression. in size to mature lymphocytes and, when flow cytometry
is performed, overlap the lymphocyte region on plots of
forward and side scatter.

Manipulating HSCs for clinical use Using cell-cycle-active drugs Because HSCs are largely in a
quiescent portion of the cell cycle (G0 or G1 ), investigators
have used cell-cycle-active drugs to deplete bone marrow
Mobilization of HSCs
populations of cycling cells and thereby enrich for primitive
Mobilization of HSCs in response to chemotherapy or HSCs. Treatment of mice with nitrogen mustard resulted in
cytokines was first documented in the 1970s and 1980s. a 30-fold enrichment in CFU-S. HSCs may be isolated by in
This process may be induced by a variety of molecules, vitro treatment with 5-fluorouracil, and this remains the most
including cytokines such as G-CSF, GM-CSF, IL-7, IL-3, commonly used agent. In addition, HSC populations may be
IL-12, SCF, and flt-3 ligand; and chemokines such as IL-8, further enriched by first stimulating cells to enter the cell
MIP-1α, Gro-β, and SDF-1. The one that is most often cycle with the early-acting cytokines c-kit ligand and IL-3,
used clinically is G-CSF, which may be combined with before forcing them to metabolic death. This strategy is use-
chemotherapeutic agents for added benefit. This mobilizing ful for human cells but not murine cells, probably because of
capability has resulted in a dramatic change in the manner different cycling characteristics. It should be noted that these
by which HSCs are harvested for transplantation. Up to 25% techniques may affect the quality of HSCs obtained.
of candidates for autologous transplantation are unable to
mobilize sufficient cells to enable the procedure to be safely Markers of primitive HSCs A variety of strategies have been
performed. The study of mobilization and its counterpart, used to identify HSC surface markers. In general, these have
engraftment, has implications of great significance for depended on excluding cells that express antigens known to
patient care. The ability of G-CSF to mobilize bone marrow be on more mature cells. For example, B cell, T cell, mono-
HSCs has several apparent mechanisms. The first is reported cyte, granulocyte, and erythroid markers are combined in
to be the activation of neutrophils, causing the release of a so-called lineage cocktail to remove cells that bind any
neutrophil elastases capable of cleaving CXCR4 on HSCs, of these antibodies. The remainder of the cells have been
thus reducing HSC–bone marrow interaction. Other recep- sequentially tested for binding of particular antibodies and
tors that undergo cleavage are VCAM-1 and c-kit. A second then transplanted to test the bound cells’ function as stem
mechanism of G-CSF-induced mobilization is via CD26, cells. This iterative process has enabled the definition of cells
an extracellular dipeptidase present on primitive HSCs that enriched for reconstituting function in irradiated mice. In the
is able to cleave SDF-1 to an inactive form. Other options case of human cells, the mice are multi-immune deficient,
for improving mobilization include co-administration of so they are tolerant of human hematopoietic cell engraft-
G-CSF and inhibitors directed against the CXCR4–CXCL12 ment. Using such strategies, antibody combinations are now
interaction. The inhibition of the CXCR4 receptor by a defined that enrich for long-term reconstituting HSCs. The
small molecule has been shown to effectively mobilize panels of markers used are provided in Table 2.2.
HSCs into the blood of patients, including those with poor
G-CSF-induced mobilization. Adding this compound to Supravital stains Since HSCs are inherently quiescent, spend
G-CSF enhances HSPC mobilization and leukopheresis most of their time in inactive portions of the cell cycle, and
yields. G-CSF does invoke an inflammatory response and are resistant to toxins, exclusion of dyes has been used as a
alternatives for its use are actively sought. Recent pre-clinical method of isolation. The DNA dye Hoechst 33342 was first
data suggest that CXCR4 antagonism plus CXCR2 agonism used to separate quiescent cells from the bone marrow. Cells
with agents such as Gro-b or CXCR4 plus VLA-4 antagonists with low-intensity staining were enriched for high prolifer-
may provide efficient mobilization alternatives. ative potential (HPP)-CFC and day-12 CFU-S. The red and
32 Molecular Hematology

antibodies are typically coupled to a hapten. A second-step


Table 2.2 Proposed surface markers of primitive hematopoietic
stem cells
incubation is then performed using a magnetic microbead
conjugated to a hapten that is able to bind the first-step
Mouse Human hapten. The effect is to label HSCs with a magnetic bead.
Cells are then passed through a column mounted adjacent to
CD34low/− CD34+ a magnet. Labeled cells are retained within the column and
Sca-1+ CD49f+ unbound cells can be washed through. Then, the column is
C-kit+ Thy1+ (CD90)+ removed from the magnet and the desired cells may be eluted.
CD150+ CD38low/−
Alternatively, negative selection may be performed by cap-
CD48− CD45RA−/low
turing only the cells that pass through the column. For exam-
lin− lin−
ple, a sample may be depleted of mature cells by labeling
with antibodies directed against mature blood cell antigens
blue emissions from this dye have been used to define a small (Linpos ). Cells can then be passed over a column in which
subset of bone marrow cells known as the side population the mature cells adhere and immature cells pass through and
(SP). SP cells have extremely low fluorescence emission in may be isolated.
these channels, resulting from efflux of Hoechst 33342 by Systems of these types permit rapid isolation of large num-
multidrug resistance pumps that are highly expressed on bers of primitive cells of relatively high purity. Bead-based
HSCs. SP cells constitute approximately 0.1% of the bone methods sacrifice cell purity for higher cell yields compared
marrow and are highly enriched in reconstitution potential. with FACS.
The mitochondrial dye rhodamine-123 (Rh-123) has also
been used to subdivide primitive stem cells. Mitochondria in Ex Vivo expansion
quiescent cells bind low levels of Rh-123 and fluorescence-
activated cell sorting (FACS) can be used to separate Rh- Given the possible clinical applications of HSCs for such uses
123low cells. These cells were enriched for day-13 CFU-S and as bone marrow transplantation, there is increasing interest
multilineage reconstituting potential. in strategies that both result in an increase in the quantity of
While supravital stains have been useful, the simplicity of HSCs and the ability to manipulate HSCs ex vivo. Thus, ex
analysis with fluorescent antibodies against cell surface mark- vivo expansion of HSCs represents a highly prioritized goal
ers has generally supplanted them. Using a combination of of clinically oriented HSC research.
antibody staining features, it is possible to enrich for HSCs The first benefit of expanding HSCs is to provide sufficient
such that fewer than 10 cells are required to reconstitute cells for transplantation when insufficient numbers exist. For
hematopoiesis. example, cord blood represents a rich source of primitive
CD34+ cells that are less immunocompetent and are there-
fore transplantable across partial human leukocyte antigen
Methods of isolation of HSCs (HLA) disparity barriers. However, the absolute quantity of
Fluorescence-activated cell sorting
HSCs within a single cord blood is low and transplantation
is followed by periods of aplasia. Ex vivo expansion would
While the flow cytometer may be used for the analysis of cells, thereby facilitate cord blood transplantation. Similarly, selec-
the apparatus may also physically sort cells of desired fluo- tive expansion of HSC subsets would permit the extension
rescence or fluorescence pattern, size, and granularity char- of tumor-free cells from patients with limited quantities
acteristics. Sorting is both expensive and labor intensive, as it of normal bone marrow due to bone marrow–infiltrating
requires costly machines, a high degree of expertise, and time diseases, such as leukemia, for the purpose of autologous
to sort samples consisting of single-cell suspensions. How- transplantation.
ever, FACS is the most commonly used method to isolate The second benefit of ex vivo manipulation is that HSCs
highly purified HSCs using both positive and negative selec- have a relative growth advantage over other cell types, such
tion strategies, with fluorescence-labeled antibodies directed as tumor cells. Therefore, ex vivo growth provides a purging
against primitive hematopoietic cell antigens, as described effect. Furthermore, specific tumor cell purging may be
earlier. achieved via the application of certain cytokines (IL-2, IFN-
γ), antitumor agents such as 5-fluorouracil or cyclophos-
Magnetic bead columns Large-volume isolation of HSC phamide, tumor-specific antibodies combined with
subsets has been facilitated by the use of magnetic bead complement-mediated lysis, and oncogene-specific tyrosine
columns. Using this system, cells are incubated with anti- kinase inhibitors, in addition to other targeted therapies,
bodies directed against primitive hematopoietic cells. These such as antisense oligonucleotides, prior to use of the graft.
Stem cells 33

The third benefit is the support of gene transfer into HSCs to proliferate. These are more primitive progenitors, but they
for the purpose of gene therapy or gene editing. A variety cannot be equated with HSCs.
of gene transfer and gene editing mechanisms are improved The LTC-IC assay correlates more closely to HSCs. Here,
with cell cycling. Further, low efficiency events in gene mod- hematopoietic cells are plated on top of stromal cell lines or
ification may be made clinically relevant if HSC expansion irradiated primary bone marrow stroma. Primitive HSCs are
can be achieved. able to initiate growth and generate progeny in vitro for up
Strategies to expand HSCs ex vivo have used a wide variety to 12 weeks. Progenitor cells and mature myeloid cells are
of cytokine cocktails. These have often resulted in more cells, removed weekly to prevent overgrowth. Ultimately, HSCs,
but not more stem cells. Progress has been made with small characterized by high proliferative and self-renewal capabil-
molecules added to cytokine combinations. The cytokines ities, are able to sustain long-term culture and may be enu-
generally involve kit ligand, thrombopoietin, IL-6, and flt-3 merated at the conclusion of the assay.
ligand. Adding aryl hydrocarbon receptor antagonist led to The CAFC assay represents a type of LTC-IC that similarly
improved outcome in umbilical cord blood transplant in one measures the ability of cells to initiate growth and generate
clinical study. Also, early results look encouraging for nicoti- progeny in vitro for up to 12 weeks. However, the readout
naminde or the small molecule UM171 currently in clini- is slightly different. Hematopoietic cells are plated at limit-
cal testing. Other results indicate that culture of cells with ing dilution on top of a monolayer consisting of irradiated
Notch ligands can reduce intervals of myeloid cell cytopenia bone marrow stroma or a stromal cell line. The growth of
and brief exposure to prostaglandinE2 analogues may affect colonies consisting of at least five small non-refractile cells
cord blood transplant. This area of research has historically reminiscent of cobblestones, found underneath the stromal
been one of frustration, but these developments indicate that layer, is counted. Such cultures are maintained using weekly
progress is being made. (Note to reader: the chapter author half-media changes until up to 5 weeks after seeding. In this
does have involvement in companies focused on this area, so assay, more primitive cells appear later, and day-35 CAFCs
interpret with caution.) represent a close correlate of a cell with in vivo long-term
multilineage repopulating potential. LTC-ICs may be enu-
merated after day 35 by completely removing the CAFC
Functional analysis of HSCs
medium, overlaying methylcellulose, and counting the num-
Functional assays for HSCs are critical, as immunopheno- ber of colonies produced after 8–10 days.
typic and other isolation methods depend on characteris-
tics that are not necessarily consistent with stem cell func- In Vivo assays The CFU-S assay, first developed by Till and
tion. This is particularly true when assessing cells after in McCulloch in 1961, is described earlier in this chapter (see
vitro manipulation or in older subjects. Immunophenotypes Hematopoietic stem cell concepts and their origin). Bone mar-
have been defined by transplantation, generally using young row or spleen cells are transplanted to irradiated recipients
donors. Only functional validation should be considered a and animals are killed after 8 or 12 days for analysis of spleen
bona fide measure of stem cells. However, even in the case colonies, termed CFU-S8 and CFU-S12 , respectively. Cells
of most assays, the setting is transplantation, which is an that give rise to CFU-S8 are predominantly unipotential and
extreme state and does not necessarily reflect stem cells under produce erythroid colonies. CFU-S12 colonies consist of sev-
homeostasis. eral types of myeloid cells, including erythrocytes, megakary-
ocytes, macrophages, and granulocytes. Cells giving rise to
In Vitro assays The CFU-C measures hematopoietic CFU-S12 represent a more primitive population of multipo-
progenitor function and is performed by plating cells in tent cells than those that result in CFU-S8 . However, these
semisolid media containing methylcellulose and one or more assays measure primitive progenitors, not HSCs.
cytokines. After 5–14 days, colonies comprising mature cell The long-term repopulation assay is a more accurate
populations committed to either myeloid or lymphoid measure of HSC activity. Whole collections of hematopoi-
lineages may be observed. While most colonies obtained etic cells or fractionated subpopulations are transplanted to
using this assay are composed of cells of a single lineage, lethally irradiated syngeneic mice, typically by tail vein injec-
less frequently multipotent progenitors can yield colonies tion. Recipients are screened for ongoing hematopoiesis 8–
containing multiple lineages. Another type of primitive cell, 10 weeks after transplantation. By this time, hematopoiesis
known as the HPP-CFC, which possesses a high degree of is firmly established and donor-derived blood is produced
proliferative and multilineage potential, may be detected by transplanted HSCs. This assay requires that cells ful-
in this culture system. Formation of HPP-CFC colonies, fill the two central features of HSCs: multilineage recon-
characterized by size greater than 0.5 mm and multilineage stitution, consistent with multipotentiality, and indefinite
composition, requires the use of multiple cytokines in order hematopoiesis, indicative of self-renewal.
34 Molecular Hematology

Assess percentage of
multilineage bone
marrow engraftment
Lethal irradiation
using flow cytometry
with 9-10Gy
with antibodies directed
against Ly5.1 and Ly5.2
Ly5.2 test cells 104
>16 weeks

103

Ly5.1 PE
Ly5.1 mouse 102

101

Ly5.1 whole bone 100 101 102 103 104


marrow competitor Ly5.2 FITC
cells Fig. 2.5 Competitive repopulation assay.

Tracking of transplanted cells was originally conducted termed competitive repopulation units. The competitive
using radiation-induced chromosomal abnormalities or by repopulation assay using congenic mouse strains is depicted
retrovirally marking donor cells. However, a major advance in Figure 2.5.
in the ability to track transplanted cells has been the develop-
ment of congenic mice with minor allelic differences in the
leukocyte common antigen Ly5, which is expressed on all
Summary
nucleated blood cells. The C57/BL6 (“black-6”) strain con-
tains the Ly5.2 antigen, while the BL6/SJL strain contains a
Investigation of HSCs has been facilitated by the develop-
separate allele, Ly5.1. However, these syngeneic strains may
ment of in vitro and in vivo assays of hematopoietic cell func-
be transplanted interchangeably. Both antibodies are avail-
tion, followed by the identification of molecular cell surface
able with distinct fluorescent labels. FACS analysis using
markers that permit the isolation of purified subsets of cells
these antibodies permits measurement of donor-derived
with defined characteristics. Studies in this field have con-
reconstitution of the nucleated blood lineages. However, ery-
tributed greatly to the understanding of both general stem
throcytes and platelets do not express the Ly5 antigen and
cell biology and hematopoiesis. Further investigation of cell-
cannot be tracked using this technique. Instead, investigators
intrinsic and cell-extrinsic regulators of hematopoiesis will
use congenic strains with allelic variants of hemoglobin and
enable rational manipulation of HSCs and thereby extend the
glucose phosphate isomerase to track erythroid and platelet
current uses of stem cells in clinical practice.
engraftment, respectively.
A modification of this assay permits quantitation of HSCs
within the graft. Here, HSCs are quantified by transplanting
limiting-dilution numbers of bone marrow into lethally Further reading
irradiated recipients. Each recipient also receives 1 × 105
cells of the host’s marrow to ensure survival during the Regulation of hematopoiesis
period of pancytopenia immediately after irradiation. At
Chai-Ho, W. and Chute, J.P. (2017). Paracrine regulation of normal and
10–12 weeks, host peripheral blood is assessed to determine
malignant hematopoiesis. Curr. Opin. Hematol. 24 (4): 329–335.
whether donor-derived reconstitution has occurred. Donor Gottgens, B. (2015). Regulatory network control of blood stem cells.
cells must constitute at least 1% of the peripheral blood to Blood 125 (17): 2614–2620.
contend that at least one HSC was present in the donor Hu, D. and Shilatifard, A. (2016). Epigenetics of hematopoiesis and
population. Also, both lymphoid and myeloid lineages must hematological malignancies. Genes Dev. 30 (18): 2021–2041.
demonstrate at least 1% donor derivations. The percentage
of reconstituted animals in each group may be plotted
against the number of input cells to determine a limiting- Hematopoietic stem cell niche
dilution estimate of the frequency of HSCs within the donor Gao, X., Xu, C., Asada, N., and Frenette, P.S. (2018). The hematopoi-
population. This assay is termed a competitive repopulation etic stem cell niche: from embryo to adult. Development 145 (2),
assay, as transplanted HSCs compete with the host’s HSCs dev139691.
that survive irradiation-induced death, in addition to host Hoggatt, J., Kfoury, Y., and Scadden, D.T. (2016). Hematopoietic stem
cells transplanted with the graft. The HSCs detected are cell niche in health and disease. Annu. Rev. Pathol. 11: 555–581.
Stem cells 35

Hematopoietic stem cell quiescence Hematopoietic stem/progenitor cell


expansion
Foudi, A., Hochedlinger, K., Van Buren, D. et al. (2009). Analysis of
histone 2B-GFP retention reveals slowly cycling hematopoietic stem Boitano, A.E., Wang, J., Romeo, R. et al. (2010). Aryl hydrocarbon recep-
cells. Nat. Biotechnol. 27 (1): 84–90. tor antagonists promote the expansion of human hematopoietic stem
Wilson, A., Laurenti, E., and Trumpp, A. (2009). Balancing dormant and cells. Science 329 (5997): 1345–1348.
self-renewing hematopoietic stem cells. Curr. Opin. Genet. Dev. 19 Lund, T.C., Boitano, A.E., Delaney, C.S. et al. (2015). Advances in umbil-
(5): 461–468. ical cord blood manipulation – from niche to bedside. Nat. Rev. Clin.
Oncol. 12 (3): 163–174.
Isolation of hematopoietic stem cells
Hematopoietic stem cell engraftment
Majeti, R., Park, C.Y., and Weissman, I.L. (2007). Identification of a
hierarchy of multipotent hematopoietic progenitors in human cord Broxmeyer, H.E. (2016). Enhancing the efficacy of engraftment of cord
blood. Cell Stem Cell 1 (6): 635–645. blood for hematopoietic cell transplantation. Transfus. Apher. Sci. 54
Oguro, H., Ding, L., and Morrison, S.J. (2013). SLAM family mark- (3): 364–372.
ers resolve functionally distinct subpopulations of hematopoietic Hoggatt, J., Speth, J.M., and Pelus, L.M. (2013). Concise review: sowing
stem cells and multipotent progenitors. Cell Stem Cell 13 (1): 102– the seeds of a fruitful harvest: hematopoietic stem cell mobilization.
116. Stem Cells 31 (12): 2599–2606.
Chapter 3 The genetics of acute
myeloid leukemias
Amy M. Trottier & Carolyn J. Owen
Division of Hematology and Hematological Malignancies, University of Calgary, Foothills Medical Centre, Calgary, Canada

Introduction, 37 AML therapies and MRD monitoring targeted by genetics, 45


AML with recurrent cytogenetic abnormalities, 37 Summary, 46
Molecular genetic aberrations not detectable by conventional Further reading, 46
cytogenetics, 40

biallelic mutations of CEBPA, as well as the addition of a


Introduction new provisional entity, AML with mutated RUNX1. Molecu-
lar analysis for mutations of CEBPA, NPM1, and FLT3 is now
Acute myeloid leukemia (AML) is a heterogeneous dis-
standard of care in newly diagnosed AML and the impor-
ease with respect to clinical and morphological features
tance of many other gene mutations is being increasingly
as well as acquired genetic aberrations. Traditionally AML
recognized. These insights have led to the incorporation of
patients were divided into three broad risk groups based
molecular aberrations into validated risk stratification mod-
on cytogenetic abnormalities: favorable, intermediate, and
els, such as that of the European LeukemiaNet (ELN), and
adverse, with each having different cure rates. However,
recommendations for risk-adapted treatment strategies, aim-
approximately 50% of patients have a normal karyotype
ing to improve treatment outcomes and minimize toxicity.
(normal-karyotype AML). These patients have varied clini-
Unfortunately, the overall outcomes in AML remain poor,
cal outcomes, with some attaining a good response to con-
with most patients succumbing to their disease. Outcomes
ventional chemotherapy and others having high rates of
are particularly poor for older adults (>65 years).
relapse and disease-related mortality. This led to the search
This chapter reviews the prognostically important recur-
for recurrent genetic aberrations that are not detectable
rent genetic aberrations observed in AML. As most cur-
by conventional cytogenetics. With the advent and wider
rently available data on the impact of genetic abnormalities in
availability of next-generation sequencing methods such as
AML are derived from large trials enrolling younger patients
whole-genome sequencing and gene expression profiling,
(<65 years), the implications for older adults, who constitute
significant progress has been made in the identification of
the majority of incident AML cases (median age at diagnosis
recurrent genetic mutations at the molecular level. These
is between 65 and 70 years), remain unclear.
“molecular markers” include mutations in specific genes as
well as altered gene expression.
The importance of these new molecular markers was man-
ifest in the 2008 update of the World Health Organization AML with recurrent cytogenetic
(WHO) classification with the addition of the provisional abnormalities
entities AML with mutated NPM1 and AML with mutated
CEBPA. Verification of the prognostic significance of these Cytogenetic abnormalities in AML have long been known to
molecular aberrations and the discovery of numerous other have prognostic relevance. Several recurrent translocations
leukemogenic driver mutations via whole-genome sequenc- are associated with a favorable prognosis when treated with
ing are reflected in the recent 2016 revision to the WHO appropriate therapeutic agents, while particular numerical
classification with the formal incorporation (no longer pro- chromosomal abnormalities, such as monosomies of chro-
visional entities) of AML with mutated NPM1 and AML with mosomes 5 and/or 7, are associated with a poor prognosis.
Advances in molecular genetic research have enabled a better
understanding of the mechanisms by which these transloca-
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. tions cause leukemia. Importantly, these studies have demon-
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. strated that the pathogenesis of AML is one of a sequential

37
38 Molecular Hematology

acquisition of genetic aberrations, with a single aberration and ∼30% of inv(16)/t(16;16) – negatively impact prognosis
being insufficient alone to cause overt leukemia. by leading to a higher risk of relapse. Also, a recent study
using high-throughput sequencing in CBF AML found
that cooperative mutations in chromatin modifiers and
Core-binding factor leukemias
cohesin were common in t(8,21) patients and associated
The core-binding factor (CBF) is a key regulator of with a particularly high risk of relapse when combined
hematopoiesis and the most frequent target of chromo- with mutations in the tyrosine kinase signaling pathway.
somal translocations associated with leukemia. This tran- Similar mutations in chromatin modifiers and cohesin were
scription factor is composed of two heterodimeric com- uncommon in inv(16)/t(16;16) AML patients.
ponents, the α-subunit encoded by RUNX1 (also called
AML1 or CBFA) and the β subunit encoded by CBFB.
Acute promyelocytic leukemia
Homozygous loss of function of either RUNX1 or CBFB
in genetically engineered mice results in a complete lack As with the CBF genes, there are also multiple chromoso-
of definitive hematopoiesis and embryonic death, indicat- mal translocations that involve the retinoic acid receptor
ing that both components of CBF are necessary for nor- RAR𝛼 locus on chromosome 17. The subgrouping of patients
mal hematopoietic development. CBF AML is a relatively with RAR𝛼 translocations is particularly important because
frequent subtype of adult AML, classified in the favorable of the distinctly better prognosis that most of these translo-
risk category, accounting for ∼15% of adult AML. CBF cations confer. In t(15;17)(q22;q12), the RAR𝛼 gene fuses
AML includes the balanced translocations t(8;21)(q22;q22), with a nuclear regulatory factor called the promyelocytic
in which the RUNX1 gene is fused to RUNX1T1 (ETO), and leukemia gene (PML), giving rise to the PML–RARα gene
inv(16)(p13.1q22) or t(16;16)(p13.1q22), in which CBFB is fusion product. Rarely, the PML–RARα fusion can result
fused to MYH11. The CBF partner gene in each case has from cryptic or complex cytogenetic rearrangements and
constitutive activity leading to persistent expression of the lack t(15;17)(q22;q12) on routine cytogenetic testing. Sev-
RUNX1–RUNX1T1 (AML-ETO) or CBFβ–MYH11 fusion eral infrequent variant translocations with RAR𝛼 also occur
protein, causing recruitment of the corepressor complex and involving the ZBTB16 (previously known as PLZF), NPM1,
underexpression of genes regulated by the CBF complex. NuMA, STAT5B, or PRKAR1A genes. Each of these is asso-
Data from in vitro studies and transgenic animal models ciated with an acute promyelocytic leukemia (APL) char-
suggest a dominant-negative role for these fusion genes. acterized by a block in differentiation at the promyelocyte
RUNX1–RUNX1T1 acts as a dominant-negative inhibitor stage of hematopoietic development. The t(15;17)(q22;q12)
of the wild-type CBF, with the fusion transcript retaining is the most frequent translocation causing APL and has
RUNX1’s DNA binding and heterodimerization domains, been extensively studied. PML–RARα expression is associ-
but lacking the C-terminal transcription activation domain. ated with a block in differentiation due to aberrant recruit-
The mechanism of dominant-negative activity is less clear ment of the nuclear co-repressor complex, similar to obser-
for CBFβ–MYH11, but both prevent transactivation of CBF vations in the context of the RUNX1–RUNX1T1 and CBFβ–
targets. Despite this understanding, the precise mechanism MYH11 fusions.
by which these fusion proteins exert their leukemogenic APL with PML-RARα is considered to have an excel-
effect remains to be elucidated. However, it is clear that lent prognosis (with a complete remission rate greater than
the translocations alone are not sufficient to result in overt 90%). It has a particular sensitivity to all-trans-retinoic acid
leukemia. For example, conditional alleles of RUNX1– (ATRA), a ligand for RARα, which has proven to be very
RUNX1T1 expressed in adult hematopoietic progenitors are effective therapy for this subtype of AML. The efficacy of
not sufficient to cause AML, but result instead in a myelo- ATRA in the treatment of APL is related to the ability of
proliferative phenotype. Experience from mouse models has ATRA to bind to the fusion protein, with resultant disso-
shown that expression of these fusion proteins resulted in ciation of the nuclear co-repressor complex. Promyelocytes
aberrant self-renewal but, by themselves, were insufficient are then able to engage normal hematopoietic differentia-
to result in leukemia, with exposure to chemical mutagens tion programs that ultimately result in apoptotic cell death.
being necessary to induce overt AML. However, some variant RAR𝛼 translocations, including cases
Although CBF AML is classified in the favorable risk with the ZBTB16-RARα and STAT5B-RARα fusion prod-
category, there is heterogeneity within this group and ∼40% ucts, are resistant to ATRA, suggesting they have different
of patients will relapse after standard treatment. Recent functional effects than the PML–RARα translocation. To
studies have found additional genetic aberrations in ∼90% stress the importance of the PML-RARα fusion, including
of CBF AML. Several of these cooperating mutations may cryptic rearrangements, the 2016 revision to the WHO clas-
contribute to leukemogenesis as well as impact survival. sification has renamed APL with t(15;17)(q22;q12); PML-
For example, c-KIT mutations – found in 20–25% of t(8;21) RARα to APL with PML-RARα.
The genetics of acute myeloid leukemias 39

Similar to the CBF leukemias, expression of PML–RARα t(6;9)(p23;q34.1) results in the formation of a chimeric
is not sufficient to induce AML. There are several lines fusion gene DEK-NUP214. The DEK-NUP214 protein alters
of evidence supporting this assertion, including transgenic nuclear transport and acts as an aberrant transcription factor.
murine models of PML–RARα-induced AML. In this model, t(6;9)(p23;q34.1) is a rare adverse risk cytogenetic abnormal-
although the fusion gene is present in the germline and is ity, found in 0.7–1.8% of AML. It typically occurs in children
expressed during embryonic and adult development, the ani- and young adults, is often associated with basophilia and
mals do not develop AML until three to six months after multilineage dysplasia, is usually the sole karyotypic abnor-
birth, and even then with a modest penetrance of only 15– mality, is commonly associated with FLT3-ITD mutations,
30%, and often with the acquisition of secondary cytogenetic and is associated with a poor prognosis.
abnormalities such as FLT3 mutations, which occur in 34– Inv(3)(q21.3q26.2) or t(3;3)(q21.3;q26.2) is the most com-
45% of APL. mon abnormality of chromosome 3q in AML, results in
MECOM activation by repositioning of a GATA2 enhancer,
and leads to GATA2 haploinsufficiency. Inv(3)/t(3;3) is found
KMT2A gene rearrangements (11q23)
in 1–2% of AML, is seldom seen in pediatric AML, and is
Abnormalities of 11q23 containing the KMT2A gene (for- an adverse cytogenetic risk factor conferring a short over-
merly known as the mixed lineage leukemia, MLL, gene) all survival (OS) and low complete remission rate. AML
are found in 3–7% of AMLs. These abnormalities can occur (megakaryoblastic) with t(1;22)(p13.3;q13.3); RBM15-MKL1
at any age, but are particularly common in children and occurs in <1% of AML, is found in infants and young chil-
in therapy-related AML after treatment with DNA topoiso- dren (≤3 years) with acute megakaryoblastic leukemia with-
merase II inhibitors. The KMT2A gene is the most promiscu- out Down syndrome, and is suggested to have a poor progno-
ous of genes, involved in over 120 different translocations in sis, though this is uncertain due to the low number of cases
AML with more than 70 different partner genes now identi- reported.
fied. These translocations lead to fusion genes containing the
N-terminus of KMT2A fused in-frame to the partner gene,
Numerical chromosomal abnormalities
most of which have subsequently been noted to have roles
in normal hematopoiesis. Molecular studies have also shown In addition to translocations, recurrent numeric chromo-
that KMT2A is frequently rearranged by cryptic transloca- somal abnormalities are noted frequently in AML. These
tions that are not detectable by conventional cytogenetics. numerical aberrations are non-random, with particular chro-
KMT2A encodes for a histone methyltransferase involved mosomes being more likely to exhibit losses or gains.
in the regulation of gene transcription via chromatin remod- Acquired trisomy of chromosome 8 or 21 is frequently seen
eling. It is important for embryonic development and regula- and is associated with an intermediate prognosis. Partial or
tion of hematopoietic differentiation. Similar to RUNX1 and complete loss of chromosome 5 and/or 7 confers a very poor
CBFB, homozygous disruption of KMT2A in mice is lethal to prognosis. Abnormalities of chromosome 7, usually in the
the embryos because of disordered hematopoiesis. KMT2A is form of monosomy or partial deletion of the long arm, con-
thought to function as both a transcriptional activator and stitute the second most frequent numerical chromosomal
a transcriptional repressor, using different domains within abnormality in de novo AML, after trisomy 8.
the protein. Importantly, the repression domain is more Many investigators have attempted to narrow the region
N-terminal to the activation domain, such that oncogenic of interest on chromosomes 5 and 7 in order to determine
KMT2A fusion proteins are thought to retain their repres- the deleted genes that result in leukemia, but no single
sion, while losing their transcriptional activation capability. culprit gene has been conclusively identified. However,
The prognostic impact of KMT2A translocations is vari- investigations in myelodysplastic syndromes (MDS) involv-
able and depends on the particular fusion partner gene. The ing deletion of the long arm of chromosome 5 (5q–) were
most frequent KMT2A translocation detectable by cytoge- more successful. A novel investigational technique using
netics in AML is t(9;11)(p22;q23), involving MLLT3 (AF9), RNA interference identified loss of the ribosomal subunit
which has an intermediate prognosis. In general, 11q23 encoding gene RPS14 as the probable cause of the erythroid
abnormalities confer an intermediate to poor prognosis. defect in the 5q– syndrome. In models using genetically
engineered mice, haploinsufficiency of ERG1 and APC genes
(both located on 5q) in combination with loss of TP53
Other rare chromosomal translocations
activity increased the rate of development of AML, but did
Other rare recurrent chromosomal translocations, including not necessarily cause overt leukemia. These results suggest
t(6;9)(p23;q34.1), inv(3)(q21.3q26.2) or t(3;3)(q21.3;q26.2), that deletion of a number of different genes on chromosome
and t(1;22)(p13.3;q13.3), are found in AML and are 5 along with cooperating mutations in tumor suppressor
listed as distinct entities in the WHO 2016 classification. genes may be required to result in leukemia.
40 Molecular Hematology

Monosomy 7 is also interesting, because it is the most com- mutation alone is not sufficient to transform a normal cell
monly acquired abnormality in children with syndromes that into a leukemic blast.
predispose to AML, such as Fanconi anemia and other bone Using whole-genome and whole-exome sequencing of 200
marrow failure syndromes. This suggests that monosomy 7 is cases of de novo AML, a landmark study by the Cancer
a secondary genetic aberration and likely requires the pres- Genome Atlas Research Network published in 2013 gener-
ence of other mutations to cause AML. ated a genomic database and found that nearly all cases had at
least one driver mutation in one of eight functional categories
relevant in the pathogenesis of AML, including the nucle-
Molecular genetic aberrations not ophosmin gene (NPM1) (27% of cases), activated signaling
detectable by conventional (59%), myeloid transcription factors (22%), tumor suppres-
cytogenetics sor genes (16%), spliceosome genes (14%), cohesin-complex
genes (13%), DNA methylation-related genes (44%), and
The development of next-generation sequencing technology, chromatin-modifying genes (30%). The relative frequency of
also known as high-throughput sequencing, has allowed for mutations in these functional categories in de novo AML is
rapid and relatively inexpensive DNA and RNA sequencing, shown in Figure 3.1.
including whole-genome sequencing, and has revolutionized A summary of the now well-established molecular mark-
the understanding of the complex genetic landscape in AML. ers that have been incorporated into routine care of AML
These technologies have uncovered hundreds of novel recur- (e.g. NPM1, FLT3, and CEBPA) as well as recently discov-
rent genetic mutations at the molecular level; however, only a ered mutations in AML is presented in what follows, focusing
small subset of these have been found to contribute to leuke- on driver gene mutations associated with possible prognostic
mogenesis and/or to clearly impact prognosis. impact in patients with AML.

Driver gene mutations NPM1

The subset of recurrent gene mutations that have been impli- NPM1 mutations were first reported in 2005 by Falini and
cated in leukemogenesis are known as driver mutations. colleagues as the most frequent genetic alteration in patients
The majority of mutations found in AML are not involved with normal-karyotype AML. NPM1 is a nucleolar phospho-
directly in leukemogenesis, but rather are believed to occur protein that continuously shuttles between the nucleus and
in unstable leukemic cells or in hematopoietic stem cells the cytoplasm, but with predominant nucleolar localization.
prior to an AML-initiating event. These are called passenger NPM1 acts as a molecular chaperone in the nucleolus and is
mutations and do not impact the course of disease. Some also involved in cell cycle progression and regulation of the
driver mutations are thought to initiate the leukemogenic alternate reading frame (ARF)/TP53 tumor suppressor path-
process (the initiating or founding clone), whereas others way. Mutations in NPM1 occur in exon 12, with more than
act in a cooperating fashion, accelerating or encouraging the 95% consisting of a 4-bp insertion at position 960. This 4-bp
leukemogenic process, and in some cases contributing to insertion results in a frameshift within the C-terminal region
chemotherapy resistance and/or risk of relapse. Various pat- of the NPM1 protein, leading to loss of nucleolar localization
terns of cooperativity as well as mutual exclusivity between and gain of a nuclear export signal. The mutant NPM1
different genes have also been found. It is thus clear that one protein (called NPM1c+) therefore acquires aberrant

NPM1 (27%) Activated signaling (59%)


Myeloid transcription factors (22%) Tumor suppressor genes (16%) Fig. 3.1 Frequency of driver mutations within eight distinct
functional categories in de novo acute myeloid leukemia, as
Spliceosome genes (14%) Cohesin-complex genes (13%)
found by the Cancer Genome Atlas Research Network using
DNA methylation-related genes (44%) Chromatin-modifying genes (30%) whole-genome sequencing.
The genetics of acute myeloid leukemias 41

cytoplasmic localization, which is readily detected by and expression is lost as hematopoietic cells differentiate. In
immunohistochemistry. This cytoplasmic localization is comparison, ITDs of FLT3 result in constitutive activation of
thought to be necessary for the leukemogenic activity of the tyrosine kinase. This results in subsequent activation of
NPM1c+, although details of the exact mechanism are still downstream signaling targets, including the phosphatidyli-
not fully elucidated. nositol 3-kinase (PI3K)/AKT, Ras/MAPK, and JAK2/STAT
NPM1 mutations occur predominantly in the normal- pathways. Mutations in FLT3 also occur in the activation
karyotype AML subgroup, with about 50% of this group loop of the tyrosine kinase domain (TKD) in about 5–10%
affected. A recent landmark study from the cancer genome of AML cases. These also result in constitutive kinase acti-
project used cytogenetics and gene sequencing to evaluate vation, but there is increasing evidence that the downstream
driver mutations in 1540 AML patients. They examined pat- signaling events are different in the setting of TKD versus
terns of co-mutation and mutually exclusive mutations. In ITD, such that their clinical implications are not equivalent.
this study NPM1 mutations were almost never found in AML with FLT3-ITD constitutes an important subset of
isolation. Commonly co-occurring mutations in DNMT3A, AML cases that has a poor prognosis in most studies of chil-
FLT3, NRAS, and TET2 were found. Of these patterns of co- dren and adults. The clinical implications of TKD muta-
mutation, the most extensively studied has been AML with tions remain controversial and larger studies are required
mutated NPM1 also carrying the FLT3-ITD mutation, which to define if these also confer a poor prognosis, though they
coexists in ∼40% of NPM1-mutated cases. are probably not independently prognostic. The frequency
The prognostic relevance of NPM1 mutations has been of FLT3 mutations varies considerably in different cytoge-
clarified in several large studies, with clear evidence that netic subgroups of AML, with a particularly high frequency
patients with mutated NPM1/wild-type FLT3 have an of 30–35% in normal-karyotype and t(15;17) AML cases. The
improved prognosis over other normal-karyotype AML poor prognosis of FLT3-ITD in normal-karyotype AML is
patients. With conventional chemotherapy these individuals conferred via increased relapse rates (RRs), with subsequent
have an event-free survival (EFS) and OS similar to those reduced EFS and OS. The complete remission (CR) rate is
with CBF leukemias. In most studies, the favorable out- not affected, but several studies have shown that complete
come predicted by NPM1 mutations is restricted to patients remission duration (CRD) is significantly reduced in these
who lack FLT3-ITD. Interestingly, initial studies suggest patients. While the significance of FLT3-ITD in t(15;17) cases
that NPM1 mutations are very stable between diagnosis is unclear, it does not appear to be an independent prognostic
and relapse, and thus are likely to constitute an initiating factor in APL patients.
rather than cooperating event in leukemogenesis. Subse- The dosage of FLT3-ITD is also important. Most FLT3-
quent acquisition of FLT3 or other cooperating mutations is ITDs are found in the heterozygous state, although some
thought to be necessary to trigger overt AML development. cases are noted to have homozygous FLT3-ITD with com-
Given the stability of NPM1 mutations and their high fre- plete loss of the wild-type allele. The homozygous FLT3
quency, quantitative reverse transcription polymerase chain mutation is thought to arise as a consequence of mitotic
reaction (RT-qPCR) assays for NPM1 mutations have been recombination resulting in segmental uniparental disomy.
developed for minimal residual disease (MRD) monitoring Prognosis appears to be worse for these patients and for other
and found, in research studies, to be predictive for relapse; patients with high expression ratios of the mutated to wild-
however, they are not yet used in routine clinical practice and type FLT3. Finally, the size of the duplicated segment may
require validation. also affect EFS, with a suggestion that longer ITDs result in
worsened outcomes.
The relatively high frequency of FLT3-ITD initially stimu-
Activated signaling lated interest for its use as a marker of MRD. However, studies
have shown that the mutational status of FLT3 may change
FLT3
between diagnosis and relapse, with about 9% of patients
FLT3 (FMS-like tyrosine kinase 3) encodes a membrane- losing their FLT3-ITD status. The length of the FLT3-ITD
bound receptor tyrosine kinase with important roles in has also been noted to vary between diagnosis and relapse.
hematopoietic stem/progenitor cell survival, differentiation, This discordance between diagnosis and relapse suggests
and proliferation. FLT3 is constitutively activated by acquired that FLT3 mutations are not sufficient for the development
mutation in approximately 20–40% of cases of AML. In 20– of overt AML, but may be secondary, cooperating events
25% of cases, internal tandem duplications (ITDs) are noted in leukemogenesis. This is further demonstrated by murine
in the juxtamembrane domain, ranging in size from 3 to bone marrow transplantation and “knock-in” models, in
400 nucleotides, with the resultant transcripts always being which FLT3-ITD induces a myeloproliferative neoplasm
in-frame and adding to the length of the protein. FLT3 is nor- (MPN), but does not cause AML. The FLT3-ITD phenotype
mally expressed in myeloid and lymphoid progenitor cells is similar to that reported in the murine bone marrow
42 Molecular Hematology

transplantation assay for other constitutively activated tyro- (FPD/AML syndrome) is an autosomal dominant disorder
sine kinases associated with myeloproliferative phenotypes caused by germline mutations in the RUNX1 gene. Individ-
in humans, including BCR-ABL, TEL-PDGFβR, TEL-ABL, uals with FPD/AML have a variable risk of developing overt
and TEL-JAK2. Taken together, these data indicate that MDS/AML (20–60% in different pedigrees) and a large range
constitutive activation of tyrosine kinases is sufficient to in age of presentation of MDS/AML (age 6–75 years). This
induce a myeloproliferative phenotype, but not AML. latency suggests that other cooperating driver mutations are
necessary to trigger overt MDS/AML. RUNX1 mutations are
c-KIT frequently associated with trisomy 21 (Down syndrome) with
an increased incidence of AML, and AML with trisomy 13
The c-KIT gene, on chromosome 4q, encodes c-KIT, a and FLT-ITD. RUNX1 mutations are mutually exclusive of
member of the type III receptor tyrosine kinase family, NPM1 and CEBPA mutations.
the same family as FLT3. c-KIT’s ligand, stem cell factor Patients with RUNX1 mutations have significantly lower
(SCF), promotes c-KIT dimerization and phosphorylation, CR rates, shorter EFS, and shorter OS than patients with
activating downstream signaling pathways important for AML with wild-type RUNX1. The adverse prognostic signifi-
proliferation, differentiation, and migration of hematopoi- cance of RUNX1 mutations has been recognized by the WHO
etic stem cells. Gain-of-function mutations in c-KIT cause and in the 2016 revision to the WHO classification a provi-
ligand-independent constitutive kinase activation and are sional category of AML with mutated RUNX1 (for cases of
found in 5–10% of all AML. However, a much higher de novo AML not associated with MDS-related cytogenetic
frequency of 12–45% is observed in the CBF leukemias. abnormalities) has been added.
Interestingly, c-KIT mutations are not seen in APL. Thus, a
significant functional redundancy is noted between c-KIT
and FLT3 mutations, with c-KIT mutations arising in CBF CEBPA
leukemias in which FLT3 aberrations are absent, and FLT3 CEBPA (CCAAT/enhancer-binding protein α) is a transcrip-
mutations arising in APL in which c-KIT mutations are tion factor that regulates genes involved in myeloid differenti-
absent. The occurrence of c-KIT and FLT3 mutations within ation, particularly by inducing granulocytic development of
the same patient is also very rare, suggesting mutual exclu- bipotential myeloid progenitors. The protein consists of N-
sivity. Like FLT3-ITD in normal-karyotype AML, c-KIT terminal transactivating domains, a DNA-binding domain,
mutations, particularly in exons 8 and 17, confer a higher and a C-terminal leucine-zipper region (bZIP), necessary for
risk of relapse and possibly inferior OS in CBF leukemia. dimerization. The gene contains two translational start sites,
The National Comprehensive Cancer Network guidelines yielding a 42-kDa and a smaller 30-kDa isoform. Somatic
now incorporate c-KIT, with c-KIT mutations moving CBF CEBPA mutations are noted in 5–14% of AML cases and
leukemia from the favorable to intermediate risk category. 70% of these mutations are detected in patients with normal-
Due to its recognized adverse prognostic impact, molecular karyotype AML. Two classes of mutations commonly occur.
testing for c-KIT mutations in CBF leukemia has become One involves N-terminal mutations, which prematurely stop
standard of care in many major centers. translation and abolish the production of the full-length
42-kDa isoform, while translation of the 30-kDa isoform
Myeloid transcription factors remains intact. The second class of mutations involves in-
frame mutations in the bZIP region and may impair DNA
RUNX1
binding, homodimerization, and heterodimerization. Most,
RUNX1 is commonly dysregulated in AML, by translocations but not all, AML cases with mutated CEBPA are biallelic,
in t(8;21) CBF leukemia, copy number variations, and point with a combination of an N-terminal and a bZIP mutation.
mutations. Although point mutations occur in only 6–10% of Others involve a homozygous CEPBA mutation and fewer
de novo AMLs, they are much more frequent in specific sub- still are monoallelic, with a single heterozygous mutation and
groups, arising in about 20% of AML with minimal differ- retained expression of a wild-type allele.
entiation (FAB M0) and about 25% of AML associated with Normal-karyotype AML with biallelic CEBPA mutations
MDS, including refractory anemia with excess blasts, AML represents a distinct molecular and clinical subtype with
with multilineage dysplasia, and AML following MDS. These favorable prognosis. CR rates are significantly higher and
point mutations are yet more frequent in therapy-related EFS and OS are significantly longer for biallelic-mutated
MDS/AML and in radiation-associated MDS/AML (tested in CEBPA compared to wide-type CEBPA or monoallelic
survivors of the atomic bomb in Hiroshima). CEBPA mutations. FLT3 and NPM1 mutations are common
Further evidence of RUNX1’s involvement in the patho- in both wide-type and monoallelic CEBPA-mutated AML,
genesis of AML has come from the analysis of pedigrees with but are rare in biallelic variants.
an inherited predisposition to MDS/AML. Familial platelet Similar to RUNX1, CEBPA mutations have been observed
disorder with a propensity to develop myeloid malignancy in very rare pedigrees with familial AML. The reported
The genetics of acute myeloid leukemias 43

pedigrees were all noted to exhibit germline mutations in as the frequent co-occurrence of cytogenetic aberrations,
the N-terminal region of CEBPA. The nature and timing of supports the necessity of additional cooperating mutations
CEBPA mutations in familial AML provide further insight for the development of AML.
into the sequence of molecular events in the development of Mutation of TP53, most commonly a missense mutation
leukemia. This insight comes particularly from second muta- in the DNA-binding domain, is the most frequent muta-
tions targeting the bZIP region of the remaining wild-type tion observed in AML with complex cytogenetics. It is
allele, which are commonly observed in familial and sporadic also often associated with specific copy number variants,
CEBPA-associated AML. These appear necessary to cause including −5/−5q, −7/−7q, and −17p. TP53 mutations are
overt leukemia in familial CEBPA-associated AML. As with mutually exclusive of FLT3, NPM1, and RUNX1 mutations.
FPD/AML, the age of presentation of disease is variable in TP53 mutations are independently associated with resistance
familial CEBPA-associated AML, ranging from 4 to 39 years. to chemotherapy, lower CR rates, and shorter OS. Their
However, the disease appears to have near-complete pene- association with complex cytogenetics is additive, with co-
trance, suggesting that the combining two CEBPA mutations occurrence having a particularly poor prognosis.
are initiating rather than cooperating leukemogenic events.
Interestingly, genome sequencing has found that mutations
in GATA2 (another myeloid transcription factor) are com- WT1
mon in cases of AML with biallelic CEBPA mutations (20– WT1 (Wilms tumor 1) gene, on chromosome 11p, encodes
40% of cases), but rare in monoallelic or wild-type CEBPA, a zinc-finger transcription factor. The precise role of WT1 in
suggesting that GATA2 may act as a cooperating mutation normal and malignant hematopoiesis remains controversial,
and contribute to leukemogenesis in a significant number but it has been implicated in the regulation of cell survival,
of AML with biallelic-mutated CEBPA cases. The prognostic proliferation, and differentiation. Its role as a tumor suppres-
impact of this cooperating mutation is unclear and requires sor gene has been implicated as mutations in WT1, primarily
further study. in exons 7 and 9, are observed in 10% of normal-karyotype
AMLs. However, WT1 is also overexpressed in various can-
cers, including the majority of AMLs, suggesting a role as
Tumor suppressors an oncogene. High expression of WT1 in AML made it an
TP53 attractive potential marker for MRD monitoring; however,
unlike NPM1, which shows stability during disease progres-
TP53 (Tumor protein 53), located on chromosome 17p, sion, WT1 mutational status has been found to change often
encodes p53, a tumor suppressor protein that contains during disease progression and at relapse, decreasing its util-
DNA binding, transcriptional activation, and oligomeriza- ity for MRD monitoring. Mutations in WT1 act as an inde-
tion domains. p53 acts as a tumor suppressor through several pendent negative prognostic indicator in AML by reducing
mechanisms including regulation of DNA repair, cell cycle rates of CR, increasing rates of relapse, and shortening OS.
arrest, and initiation of apoptosis. TP53 mutations are asso- Mutations are noted in heterozygous and homozygous states
ciated with a variety of human malignancies, are commonly and show some association with FLT3-ITD, though larger
found in therapy-related AML (t-AML; 30–40% of cases) and studies are required to confirm this.
AML with complex cytogenetics (53–78%), and are associ-
ated with poor prognosis.
Until recently, it was believed that TP53 mutations in Spliceosome genes
t-AML were the result of DNA damage from cytotoxic
SRSF2
chemotherapy. However, recent studies using genome
sequencing of t-AML patients and chemotherapy-naı̈ve SRSF2 (Serine/Arginine-Rich Splicing Factor 2) encodes a
adults have shown that TP53 mutations increase with pre-mRNA splicing factor protein that is part of the spliceo-
age, can occur several years (three to six years) before some. SRSF2 is commonly mutated in MDS and CMML
the development of t-AML, and have been found prior and has been implicated in driving dysplasia and ineffec-
to the exposure of chemotherapy in patients who later tive hematopoiesis. Co-mutations in ASXL1, RUNX1, and
developed t-AML. This led to an alternative hypothesis for IDH1/2 have been commonly described in SRSF2-mutated
the pathogenesis of t-AML, one in which chemotherapy MDS. Mutations in SRSF2 are most commonly heterozygous
does not directly induce TP53 mutations, but instead rare, missense mutations resulting in a gain of function. SRSF2
age-related TP53 mutations occurring in hematopoietic mutations are associated with older age at diagnosis and a
stem cells are relatively chemotherapy resistant and are worse prognosis in MDS, with shorter OS and increased fre-
preferentially selected for after exposure to chemotherapy. quency of progression to AML.
This hypothesis was supported by murine models. The SRSF2 mutations in AML are activity being studied
latency time between TP53 mutation and overt AML, as well and have been found in 6–7% of cases. They have been
44 Molecular Hematology

established to be highly specific for secondary AML (AML (DNMT3A) was the most common, seen in 26% of all AML
arising from MDS or CMML). These mutations have been cases examined.
found to occur early in leukemogenesis, are not sufficient DNMT3A encodes a DNA methyltransferase enzyme that
to induce overt leukemia as the sole mutation, and show functions independently of replication. Whole-genome and
stability with disease evolution, often persisting in clonal whole-exome sequencing have found that DNMT3A is one of
remissions. Studies have shown that SRSF2 driver mutations the most frequently mutated genes in AML, occurring in 12–
in AML are associated with older age and inferior OS; how- 35% of all cases, and is most often seen in normal-karyotype
ever, data for their role as an independent prognostic marker AML. The most common mutations of DNMT3A involve
in AML have been conflicting and larger studies are needed. missense mutations at residue R882 near the carboxyl termi-
nus of the encoded protein. The precise biological and func-
tional effect of these mutations is not yet fully understood.
Cohesin complex genes However, a recent study examining clonal relationships in
Cohesin is a large multiprotein complex composed of AML found that mutations in DNMT3A, along with muta-
SMC1A, SMC3, RAD21, STAG1, and STAG2. The cohe- tions in other genes that encode epigenetic modifiers, were
sion complex’s main role is to maintain the polarity of sister acquired early on in leukemogenesis, were usually present
chromatids during mitosis, but it is also involved in double- in the founding clone, and were rarely the sole mutation.
stranded DNA damage repair and transcriptional regula- DNMT3A is frequently co-mutated along with NPM1, FLT3,
tion. Recurrent mutations, typically loss-of-function muta- and IDH1/2 (particularly IDH2). These data suggest that iso-
tions, in the cohesin complex have been found in 6–13% of lated mutations in DNMT3A are not sufficient to result in
de novo and 20% of secondary AML cases, respectively. In overt AML.
one large cohort study, 48% of cohesion-mutated AML cases DNMT3A mutations have been found to confer a worse
had a normal karyotype. Cohesin mutations have also been prognosis, with reduced OS and relapse-free survival (RFS)
reported in MDS and are especially prevalent in high-risk compared to wide-type DNMT3A in several studies, includ-
MDS. Mutations of STAG2, located on chromosome X, were ing two meta-analyses with over 4000 patients. However,
found to be associated with shorter OS in MDS and were other studies, stratified by ELN risk group, have found that
more common in high-risk disease. Mutations of the cohesin mutated DNMT3A is associated with worse OS and RFS only
complex are mutually exclusive of one another and have for patients in the ELN Intermediate-1 risk group.
been found to co-occur with NPM1, RUNX1, and ASXL1
mutations. IDH1/2
Murine and in vitro models have shed light on the role of
cohesin mutations in leukemogenesis. In these studies, muta- IDH1 and IDH2 encode for isocitrate dehydrogenases
tion of cohesin in hematopoietic stem cells led to a differ- that catalyze the oxidative decarboxylation of isocitrate
entiation block with increased CD34+ progenitor cells and to α-ketoglutarate. IDH1 is cytoplasmic, whereas IDH2 is
a shift toward myeloid lineages, with cohesin knockdown mitochondrial in location. IDH1/2 are mutated in several
mice later developing features of MPNs. Cohesin mutations different cancers, including AML, and share similar fea-
appear to occur early in leukemogenesis and are insufficient tures in these cases. IDH1/2 mutations are predominantly
to cause overt leukemia as the sole mutation. The prognos- somatic, are heterozygous, and occur at an early stage of
tic significance of cohesin complex mutations in AML is tumourigenesis. Mutations in IDH1 and IDH2 have been
currently unknown, with contradictory reports that require found in 6–16% and 8–19% of AML, respectively. They
more investigation. are most commonly found in normal-karyotype AML, are
mutually exclusive of one another, and are associated with
mutated NPM1. Epigenetic studies have found that IDH1/2
DNA methylation mutations result in global DNA hypermethylation and
impair hematopoietic differentiation.
DNMT3A
The prognostic impact of IDH1 and IDH2 mutations is less
DNA methylation is an epigenetic process (a process that clear. Several studies have found no impact of IDH mutations
alters gene expression without changing DNA sequences) on OS, CR rate, or RFS for normal-karyotype AML; how-
that is frequently altered in malignancies, including AML. ever, results from the literature are conflicting. Some studies
Cancer genomes often exhibit global DNA hypomethylation, have found an adverse impact of particular IDH1 mutations,
but have also been found to display DNA hypermethyla- whereas others found no difference. The findings for IDH2
tion of the promotor regions of tumor suppressor genes. The mutations have ranged from a favorable impact with longer
Cancer Genome Atlas Research Network found recurrent OS for R140Q IDH2 mutation to an adverse prognosis with
mutations in genes involved in DNA methylation in 44% of worse OS and reduced CR rates for R172 IDH2 mutations,
AML cases. Of these, mutation in DNA methyltransferase 3A and no significant impact in other cases.
The genetics of acute myeloid leukemias 45

TET2 mutated ASXL1 and SRSF2 was additive, with co-occurrence


having a worse prognosis than either mutation alone.
TET2 (Tet Methylcytosine Dioxygenase 2) encodes a methyl-
cytosine dioxygenase, which is involved in myelopoiesis. In
2009, somatic mutations in TET2 were described for the first KMT2A (formerly MLL)
time in various myeloid disorders, including MDS, MPNs,
As with RUNX1, the KMT2A gene was first noted to be
and AML. Since that time the frequency of TET2 mutations
aberrantly regulated in AML through translocations. Subse-
and its role in leukemogenesis have been examined. Mouse
quently, KMT2A partial tandem duplications (PTDs) were
models and in vitro studies have found that TET2 mutations,
reported in 3–11% of normal-karyotype AML patients.
which lead to loss of function, result in widespread DNA
These mutations are more frequent in AML patients with
hypermethylation of enhancer elements of tumor suppressor
normal karyotype or with trisomy 11, and are often associ-
genes and consequent decreased expression of these protec-
ated with FLT3 mutation. KMT2A-PTD was the first gene
tive genes.
mutation shown to negatively affect prognosis in normal-
Mutated TET2 occurs in 12–17% and 24–32% of de novo
karyotype AML patients, with most large studies confirming
and secondary AML, respectively. Like the other genes
these initial findings. Similar to the other genes involved in
involved in DNA methylation (IDH1/2 and DNMT3A),
DNA methylation and chromatin modification previously
TET2 mutations occur early in leukemogenesis. TET2-
discussed, KMT2A-PTDs are interesting because the pres-
mutated AML is most frequently associated with a normal
ence of a heterozygous KMT2A-PTD has been associated
karyotype and is often seen with co-mutations in NPM1 and
with silencing of the wild-type allele in AML blasts. The
ASXL1. TET2 mutations and IDH1/2 mutations are mutually
mechanism for this silencing appears to involve epigenetic
exclusive.
modifications rather than direct mutational effects. Activity
As with IDH mutations, the independent prognostic sig-
of DOT1L, a histone methyltransferase protein, has recently
nificance of TET2 mutations remains unclear, with conflict-
been found to cause aberrant hypermethylation in KMT2A-
ing evidence. Some groups have reported inferior OS ver-
rearranged AML, and appears to be vital in the pathogenesis
sus wide-type TET2 for young (age <60) patients with AML,
and maintenance of KMT2A-rearranged leukemia. This
whereas others have reported no significant difference. Other
suggests that epigenetic events may be sufficient to serve
studies have found that mutated TET2 confers worse CR rates
as cooperating events in leukemogenesis. This mechanistic
and shorter EFS and OS only for normal-karyotype patients
understanding is also important in identifying a new avenue
in the ELN favorable risk category.
for molecularly targeted therapy in AML through the use
of targeted therapy, hypomethylating agents, and histone
Chromatin modifiers deacetylase inhibitors.
ASXL1

ASXL1 (Additional Sex Combs Like-1), located on chro- AML therapies and MRD monitoring
mosome 20q, encodes a protein member of the Polycomb targeted by genetics
group that is thought to disrupt chromatin in particular areas,
resulting in both epigenetic activation of certain genes and Advances in molecular technologies have led to the discov-
repression of others. ASXL1 mutations have been found in ery of numerous driver mutations in AML and have pro-
MPNs, MDS, CMML, and AML. The majority of mutations vided abundant new targets for directed therapies in AML
are frameshift and nonsense mutations in exon 12. ASXL1 as well as for MRD monitoring. A comprehensive review of
mutations are relatively specific for secondary AML, occur- targeted therapies and MRD monitoring is beyond the scope
ring at a frequency of 6.5% of de novo AML but 30% of of this chapter and readers are referred to recent review arti-
secondary AML. ASXL1 mutations are also associated with cles for further details. The best example of targeted therapy
RUNX1, SRSF2, and IDH2 mutations as well as older age in AML is the use of ATRA in APL, in which ATRA over-
at diagnosis, whereas they are almost mutually exclusive of comes the PML–RARα-induced differentiation block, lead-
NPM1, FLT3-ITD, and DNMT3A mutations. Like other epi- ing to maturation of the malignant myeloblasts and promye-
genetic modifiers, mutations in ASXL1 are believed to occur locytes. Mutations in genes involved in DNA methylation,
early in the process of leukemogenesis and are not sufficient including DNMT3A and TET2, are attractive targets for
alone to result in overt AML. hypomethylating agents such as azacitidine and decitabine.
Mutated ASXL1 has been associated with a poor prognosis Several other molecular targeted therapies are also being
in AML, with lower CR rates and shorter OS and EFS com- investigated and are in various stages of development, rang-
pared to wide-type ASXL1. A recent study on the genomic ing from preclinical studies to phase III trials, and some with
classification and prognosis of AML that enrolled over recent FDA approval. These include first-generation broad-
1500 patients found that the negative prognostic impact of spectrum kinase inhibitors (e.g. midostaurin and sorafenib),
46 Molecular Hematology

second-generation FLT3-specific inhibitors (Quizartinib, implications and may help risk stratify patients to make treat-
Crenolanib, and Gilteritinib), IDH1 (Ivosidenib) and IDH2 ment decisions clearer. The recently published 2016 revision
inhibitors (Enasidenib), DOT1L inhibitors for KMT2A- to the WHO classification now incorporates several molecu-
mutated AML (EPZ-5676), polyvalent WT1 peptide vaccine, lar categories, including AML with mutated NPM1 and AML
and several others. There is much hope that these targeted with biallelic mutations of CEBPA, and has a new provisional
therapies will improve survival for AML patients. entity of AML with mutated RUNX1. Molecular analysis for
Assessment of treatment response currently relies on eval- mutations of CEBPA, NPM1, and FLT3 is now standard of
uation of bone marrow morphology and MRD detection by care in newly diagnosed AML and has been incorporated
flow cytometry. However, morphologic assessment is sub- in various risk stratification models. New sequencing tech-
ject to limited sensitivity and interobserver variability, and nologies and the discovery of numerous driver mutations
even the additional use of flow cytometry can be affected by have spurred research toward novel targeted therapies, use
hemodilution. Though the presence of MRD by flow cytom- of molecular markers for MRD monitoring, and develop-
etry is prognostically informative, studies have found that ment of a fully genomic classification of AML. In the future,
about one-third of younger AML patients with no detectable it may be possible to use combinations of conventional and
MRD by flow cytometry will still relapse. Due to these flaws, molecularly targeted therapies, in selected clinical contexts,
there has been a push toward finding more sensitive mark- to improve outcomes and/or to reduce toxicity. Given the
ers of MRD. RT-qPCR is used for MRD monitoring in APL dismal prognosis for most patients with AML, these discov-
and an undetectable PML-RARα transcript is a prerequi- eries have brought new excitement to the field of leukemia
site for molecular remission. Other recurrent genetic aberra- research and have identified new avenues for the prognosti-
tions such as mutated NPM1, FLT3-ITD, and WT1 have been cation, treatment, and monitoring of patients.
investigated for RT-qPCR MRD monitoring. Of these, NPM1
seems the most promising given its high frequency and stabil-
ity in AML. RT-qPCR assays for MRD monitoring have also
been evaluated for CBF leukemia and found to be predictive Further reading
of relapse.
Genetics of AML
Bullinger, L. and Valk, P.J. (2005). Gene expression profiling in acute
myeloid leukemia. J. Clin. Oncol. 23: 6296–6305.
Summary Cancer Genome Atlas Research Network (2013). Genomic and epige-
nomic landscapes of adult de novo acute myeloid leukemia. N. Engl.
Recurrent cytogenetic abnormalities are frequent in AML J. Med. 368: 2059–2074.
and many new molecular aberrations have also been discov- Dohner, H., Estey, E.H., Amadori, S. et al. (2010). Diagnosis and
ered that are undetectable by conventional karyotyping. The management of acute myeloid leukemia in adults: recommenda-
advent of next-generation sequencing technologies has shed tions from an international expert panel, on behalf of the European
much light on leukemogenesis and has uncovered numerous LeukemiaNet. Blood 115: 453–474.
Jamani, K. and Owen, C. (2015). Update on recurrent genetic aber-
driver mutations commonly found in AML. Some of these
rations in acute myeloid leukemia. Int. J. Hematol. Oncol. 4: 179–
driver mutations are initiating events, others are cooperating 190.
mutations, and several have prognostic impact. Studies Lindsley, R.C., Mar, B.G., Mazzola, E. et al. (2015). Acute myeloid
suggest that at least two, and often more, driver mutations leukemia ontogeny is defined by distinct somatic mutations. Blood
are required for the development of overt AML. Various 125: 1367–1376.
patterns of mutual cooperativity as well as exclusivity have Mrozek, K., Marcucci, G., Paschka, P. et al. (2007). Clinical relevance
been found among these mutations. The vast majority of of mutations and gene-expression changes in adult acute myeloid
identified driver mutations fall within one of eight functional leukemia with normal cytogenetics: are we ready for a prognostically
categories, including NPM1, activated signaling, myeloid prioritized molecular classification? Blood 109: 431–448.
transcription factors, tumor suppressor genes, spliceosome Papaemmanuil, E., Gerstung, M., Bullinger, L. et al. Genomic classifica-
genes, cohesion-complex genes, DNA methylation-related tion and prognosis in acute myeloid leukemia. N. Engl. J. Med. 374:
2209–2221.
genes, and chromatin-modifying genes.
Raghavan, M., Lillington, D.M., Skoulakis, S. et al. (2005). Genome-
Currently, AML patients who are at high risk of relapse wide single nucleotide polymorphism analysis reveals frequent par-
are recommended for allogeneic stem cell transplantation, tial uniparental disomy due to somatic recombination in acute
a treatment with a relatively high morbidity and mortality. myeloid leukemias. Cancer Res. 65: 375–378.
The decision of whether to proceed to transplantation is a Renneville, A., Roumier, C., Biggio, V. et al. (2008). Cooperating gene
difficult one for both the patient and the physician. Many mutations in acute myeloid leukemia: a review of the literature.
of the newly discovered driver mutations have prognostic Leukemia 22: 915–931.
The genetics of acute myeloid leukemias 47

Core-binding factor leukemias Wu, S.J., Kuo, Y.Y., Hou, H.A. et al. (2012). The clinical implication of
SRSF2 mutation in patients with myelodysplastic syndrome and its
Duployez, N., Marceau-Renaut, A., Boissel, N. et al. (2016). Compre- stability during disease evolution. Blood 120: 3106–3111.
hensive mutational profiling of core binding factor acute myeloid
leukemia. Blood 127: 2451–2459.
Speck, N.A. and Gilliland, D.G. (2002). Core-binding factors in Cohesin complex genes
haematopoiesis and leukemia. Nat. Rev. Cancer 2: 502–513. Thota, S., Viny, A.D., Makishima, H. et al. (2014). Genetic alterations
of the cohesin complex genes in myeloid malignancies. Blood 124:
NPM1 1790–1798.

Falini, B., Mecucci, C., Tiacci, E. et al. (2005). Cytoplasmic nucleophos-


min in acute myelogenous leukemia with normal karyotype. N. Engl. DNA methylation
J. Med. 352: 254–266. Figueroa, M.E., Abdel-Wahab, O., Lu, C. et al. (2010). Leukemic IDH1
Schnittger, S., Schoch, C., Kern, W. et al. (2005). Nucleophosmin gene and IDH2 mutations result in a hypermethylation phenotype, disrupt
mutations are predictors of favorable prognosis in acute myelogenous TET2 function, and impair hematopoietic differentiation. Cancer Cell
leukemia with a normal karyotype. Blood 106: 3733–3739. 18: 553–567.
Im, A.P., Sehgal, A.R., Carroll, M.P. et al. (2014). DNMT3A and IDH
Activated signaling mutations in acute myeloid leukemia and other myeloid malignan-
cies: associations with prognosis and potential treatment strategies.
Chen, W., Xie, H., Wang, H. et al. (2016). Prognostic significance of KIT Leukemia 28: 1774–1783.
mutations in core-binding factor acute myeloid leukemia: a system-
atic review and meta-analysis. PLoS One 11: e0146614.
Gale, R.E., Green, C., Allen, C. et al. (2008). The impact of FLT3 internal Chromatin modifiers
tandem duplication mutant level, number, size, and interaction with Schnittger, S., Eder, C., Jeromin, S. et al. (2013). ASXL1 exon 12 muta-
NPM1 mutations in a large cohort of young adult patients with acute tions are frequent in AML with intermediate risk karyotype and are
myeloid leukemia. Blood 111: 2776–2784. independently associated with an adverse outcome. Leukemia 27:
Thiede, C., Steudel, C., Mohr, B. et al. (2002). Analysis of FLT3- 82–91.
activating mutations in 979 patients with acute myelogenous
leukemia: association with FAB subtypes and identification of sub-
groups with poor prognosis. Blood 99: 4326–4335. AML therapies and MRD monitoring
targeted by genetics
Myeloid transcription factors Grimwade, D. and Freeman, S.D. (2014). Defining minimal residual
disease in acute myeloid leukemia: which platforms are ready for
Dufour, A., Schneider, F., Metzeler, K.H. et al. (2010). Acute myeloid
“prime time”? Hematol. Am. Soc. Hematol. Educ. Program 2014: 222–
leukemia with biallelic CEBPA gene mutations and normal karyotype
233.
represents a distinct genetic entity associated with a favorable clinical
Perl, A. and Carroll, M. (2007). Exploiting signal transduction pathways
outcome. J. Clin. Oncol. 28: 570–577.
in acute myelogenous leukemia. Curr. Treat. Options Oncol. 8: 265–
Osato, M. (2004). Point mutations in the RUNX1/AML1 gene: another
276.
actor in RUNX leukemia. Oncogene 23: 4284–4296.
Stein, E.M. and Tallman, M.S. (2016). Emerging therapeutic drugs for
Wouters, B.J., Lowenberg, B., Erpelinck-Verschueren, C.A. et al. (2009).
AML. Blood 127: 71–78.
Double CEBPA mutations, but not single CEBPA mutations, define a
subgroup of acute myeloid leukemia with a distinctive gene expres-
sion profile that is uniquely associated with a favorable outcome. Familial AML
Blood 113: 3088–3091.
Michaud, J., Wu, F., Osato, M. et al. (2002). In vitro analyses of
known and novel RUNX1/AML1 mutations in dominant familial
Tumor suppressors platelet disorder with predisposition to acute myelogenous leukemia:
implications for mechanisms of pathogenesis. Blood 99: 1364–
Wong, T.N., Ramsingh, G., Young, A.L. et al. (2015). Role of TP53 muta-
1372.
tions in the origin and evolution of therapy-related acute myeloid
Owen, C., Barnett, M., and Fitzgibbon, J. (2008). Familial myelodyspla-
leukaemia. Nature 518: 552–555.
sia and acute myeloid leukemia: a review. Br. J. Haematol. 140: 123–
132.
Spliceosome genes Song, W.-J., Sullivan, M.G., Legare, R.D. et al. (1999). Haploinsufficiency
of CBFA2 (AML1) causes familial thrombocytopenia with propensity
Thol, F., Kade, S., Schlarmann, C. et al. (2012). Frequency and prognos-
to develop acute myelogenous leukemia (FPD/AML). Nat. Genet. 23:
tic impact of mutations in SRSF2, U2AF1, and ZRSR2 in patients with
166–175.
myelodysplastic syndromes. Blood 119: 3578–3584.
Chapter 4 Molecular diagnostics and risk
assessment in myeloid malignancies
Christian Scharenberg1,2 & Torsten Haferlach3
1
Department of Hematology, Skaraborgs Hospital, Skövde, Sweden
2
Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
3 MLL Munich Leukemia Laboratory, Munich, Germany

Introduction, 49 Clinical implications, 52


General methodology of cytogenetic analysis in hematological Treatment selection, 52
malignancies, 50 Therapy-related chromosomal aberrations in secondary AML, 54
Methods of detection, 50 Recurrently mutated genes in MDS, 54
Genetic and clinical consequences of chromosomal translocation, 51 Recurrently mutated genes in AML, 56
Karyotypic evolution, 52 Further reading, 57

20% in patients over the age of 65 and 90, respectively. To


Introduction describe this phenomenon, the acronym CHIP for clonal
hematopoiesis of indeterminate potential has been adopted.
The recent progress in high-throughput genome sequencing
Individuals in whom mutations were detected had usually
technologies has enabled the discovery of genetic lesions that
only a single mutation. Remarkably, mutations in only three
drive the pathogenesis of the majority of human cancers. This
genes, DNMT3A, TET2, and ASXL1, explained the majority
progress allowed for the identification of recurring chromo-
of driver mutations sustaining clonal hematopoiesis.
somal abnormalities and gene alterations that has tremen-
The initial hit occurs in a single stem cell, and it is believed
dously advanced our understanding of the pathobiology of
that the subsequent expansion of the formed clone is quite
myeloid neoplasms.
substantial (median clone size measured in the peripheral
While previously considered related albeit distinct enti-
blood was approximately 18%). In contrast to previous stud-
ties, acute myeloid leukemia (AML) and myelodysplastic
ies that could detect certain chromosomal translocations
syndromes (MDS) are now thought of as part of a dis-
only transiently in healthy individuals, CHIP appears to per-
ease continuum with a recognizable pre-malignant state. In
sist for years with no indication of spontaneous resolution.
addition to the usefulness of molecular analyses in diag-
Clonal hematopoiesis seems to be a strong predictor for
nosis and prognosis, the discovery of such mutations has
the development of hematological malignancy (hazard ratio
offered genetic tools to study clonal diversity and disease
of ∼12), confirming that the initial mutation constitutes a
evolution.
pre-malignant state. Direct evidence for the evolution from
clonal hematopoiesis to hematologic malignancy was possi-
Clonal hematopoiesis ble in a small number of patients who developed AML, as the
leukemic samples were found to contain the somatic muta-
While cancer is now known to result from the stepwise accu- tions at high allele fractions. Therefore, clonal hematopoiesis
mulation of somatic mutations, it has remained difficult to is now viewed as part of the natural evolution from a pre-
define the initial stages preceding the development of overt malignant state to overt leukemia.
hematological malignancies.
Using existing datasets of exome sequencing on periph-
eral blood samples from more than 30 000 patients with-
Myelodysplastic syndromes
out known hematological cancers, three studies found evi-
dence for somatic mutations in blood DNA. Despite being The MDS comprise a series of hematologic conditions of
rare (less than 1%) in people under 40 years of age, somatic abnormal cellular maturation leading to chronic cytopenias
mutations increase with age, with frequencies up to 10% and (i.e. anemia, neutropenia, thrombocytopenia). As a result,
patients with MDS are at risk for symptomatic anemia, infec-
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. tion, and bleeding, as well as progression to AML, which is
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. often refractory to standard treatment.

49
50 Molecular Hematology

The complexities of the pathobiology of MDS are begin- understanding of the pathobiology of AML. In addition
ning to be elucidated. The development of MDS occurs via a to having considerable prognostic significance for patients
series of genetic changes in a hematopoietic stem cell (HSC). with AML, certain cytogenetic abnormalities identified by
These changes alter normal hematopoietic growth and differ- karyotype analysis affect the planning of treatment. Abnor-
entiation, resulting in an accumulation of abnormal, imma- malities in certain genes (e.g. mutations in FLT3, NPM1,
ture myeloid cells in the bone marrow, leading to impairment KIT) as well as gene expression profiles confer prognostic
of normal hematopoiesis. Advances in the identification of significance in adult patients with AML.
recurring chromosomal abnormalities and gene alterations
have provided insight into the pathobiology of MDS.
It is well established that specific cytogenetic abnormal-
General methodology of cytogenetic
ities identified by karyotype analysis or fluorescence in situ
analysis in hematological
hybridization (FISH) analysis bear prognostic significance
malignancies
for patients with primary MDS and affect the planning
of treatment. While certain gene mutations also confer
The malignant cells in many patients with leukemia, lym-
independent prognostic significance in adult patients with
phoma, or another malignant hematologic disease have
MDS, it is not yet clear how to incorporate these changes into
acquired clonal chromosomal abnormalities. Some spe-
treatment planning. Even those patients without obvious
cific cytogenetic abnormalities are closely, and sometimes
abnormalities detected by karyotypic analysis, FISH, or
uniquely, associated with morphologically and clinically
gene mutation analyses likely have acquired copy number
distinct subsets of leukemia or lymphoma, as well as with
alterations or abnormalities in gene expression profiles,
their prognosis.
which may help to identify genes with important roles for
Compared to other malignancies and given the ease of
the pathogenesis of MDS.
obtaining and processing bone marrow or peripheral blood
The development of secondary, therapy-related MDS or
samples, significantly more information is available about
AML is associated with characteristic chromosomal abnor-
the relationship between cytogenetic abnormalities and the
malities in patients who developed MDS or AML after radi-
pathogenesis and natural history of the leukemias.
ation therapy and/or chemotherapy for an earlier disease, for
The detection of clonal cytogenetic abnormalities allows
example lymphoma or a solid tumor, or even non-malignant
the differentiation between benign reactive lymphoid or
disorders, such as rheumatoid arthritis, or following organ
myeloid hyperplasia and a monoclonal malignant prolifer-
transplantation.
ation, and in some cases the establishment of specific diag-
noses, for example the Philadelphia chromosome in chronic
Acute myeloid leukemia myeloid leukemia (CML). In addition, the study of cytoge-
netic abnormalities has provided insight into disease patho-
At first glance the development of AML resembles that of
genesis (by identifying genes controlling cell growth and
MDS. However, after a series of genetic changes generate
leukemogenesis), prognosis (clonal evolution can signify a
pre-AML stem cells, the final transformative event leading to
more aggressive disease course), and treatment planning
overt development of AML occurs in a hematopoietic pre-
(certain chromosomal changes predict for response to spe-
cursor rather than in HSCs. By altering normal hematopoi-
cific therapies, or for the choice of targeted therapy).
etic growth and differentiation, large numbers of abnormal,
immature myeloid cells accumulate in the bone marrow and
peripheral blood. While capable of dividing and proliferat-
ing, these cells cannot differentiate into mature hematopoi- Methods of detection
etic cells.
Not unlike other malignant diseases, the genetic alter- The following methods are used for the detection of cytoge-
ations in AML include mutation of oncogenes as well as the netic aberrations: conventional metaphase cytogenetic anal-
loss of tumor suppressor genes. Compared to solid tumors, ysis, FISH analysis, reverse transcription-polymerase chain
however, many hematologic malignancies are associated reaction (RT-PCR), microarray-based genomic copy number
with a single characteristic cytogenetic abnormality, for analysis, and DNA or RNA sequencing.
instance the Philadelphia chromosome t(9;22) in chronic Though technically difficult, the entire tumor genome can
myeloid leukemia and t(15;17) in acute promyelocytic be examined by conventional metaphase cytogenetic analy-
leukemia (APL). sis. This method is therefore of particular importance in the
The identification of recurring chromosomal abnor- evaluation of hematologic malignancies with many potential
malities and translocations has largely advanced our genetic abnormalities (e.g. AML). Conventional metaphase
Molecular diagnostics and risk assessment in myeloid malignancies 51

cytogenetic analysis is performed by culturing the tumor entities (e.g. multiple tyrosine kinase gene fusions in Ph-like
sample in vitro to stimulate cellular division. Once the cells B-ALL).
divide readily, mitotic division is arrested in metaphase by While the detection of specific chromosome translocations
adding a chemical (e.g. colchicin). Subsequently, chromoso- and gene fusions allows a precise diagnosis of leukemia, anal-
mal banding is performed using various enzymes and histo- ysis of recurrent somatic mutations in AML and MDS adds
logical stains. important genetic information, offering better risk stratifica-
FISH analysis relies on the ability of single-stranded DNA tion and selection of targeted therapies.
to anneal to complementary DNA. The technique employs Importantly, each technique has its advantages and dis-
fluorescently labeled DNA probes that are hybridized to advantages – while molecular methods, such as RT-PCR or
either metaphase chromosomes or interphase nuclei affixed NGS techniques, assess the entire cell population, FISH pro-
to a glass slide. This method can be employed with bone vides analysis at the single-cell level. A comprehensive picture
marrow or peripheral blood smears, cytospin slides, as well of the genetic landscape of leukemia or lymphoma emerges
as fixed and sectioned tissue. It is both rapid and sensitive, from the combination of conventional cytogenetic analysis,
allowing the detection of recurring numerical and structural FISH tests, microarray, and NGS studies, which enables per-
abnormalities. FISH is most useful when certain abnormali- sonalized genetic-based treatment.
ties are suspected or seem likely based on other investigations
(e.g. based on distinctive morphology). A significant short-
coming is that additional abnormalities or multiple clones Genetic and clinical consequences of
will not be detected by this method alone. Hence, FISH serves chromosomal translocation
as a complement to conventional cytogenetic analysis in the
majority of clinical cases. An important role in the process of malignant transforma-
PCR-based methods markedly increase the sensitivity with tion is that of alterations in the expression of genes that lie at
which chromosomal aberrations can be detected. Transloca- the breakpoints of the chromosomal translocations or within
tions resulting in fusion genes are ideally suited for detection functional domains of the encoded proteins. The genes with
by RT-PCR, a technique where messenger RNA (mRNA) is a role in transformation can be grouped into the follow-
first copied into complementary DNA (cDNA) and subse- ing functional classes: (i) tyrosine or serine protein kinases;
quently the fusion gene is amplified by PCR using specific (ii) cell surface receptors and growth factors; (iii) proteins
primers from each gene. In certain cases, DNA sequencing that regulate transcription; and (iv) regulators of apoptosis.
is then used to confirm the identity of the amplified prod- Among these, genes that encode proteins regulating tran-
uct. Given the high sensitivity of RT-PCR (down to detection scription are the most commonly involved. Gene transcrip-
of one abnormal in a million normal cells), this method is tion is the process by which the information in genomic DNA
ideally suitable for the detection of minimal residual disease is transcribed to RNA. Transcription factors are central to
(MRD) in patients who are in clinical remission. this process, recognizing and binding to target sequences in
Genomic microarray technology using single-nucleotide the regulatory elements of genes or to other DNA-binding
polymorphisms (SNPs) allows for the identification of dis- proteins in a tissue-specific fashion. Chromosomal translo-
ease susceptibility loci in genome-wide association studies cations can alter gene function by aberrant regulation of gene
and the identification of other acquired abnormalities, such expression or via expression of a novel fusion protein.
as genomic imbalances (e.g. duplications or cryptic deletions) The latter result from the juxtaposition of coding
and loss of heterozygosity (LOH), which occurs without sequences from two genes that are normally located on
concomitant changes in the gene copy number via somatic different chromosomes. This produces a fusion mRNA and a
mitotic recombinations known as uniparental disomy. chimeric protein with altered gene function. There are three
Next-generation sequencing (NGS) techniques are increas- well-characterized fusion proteins: BCR-ABL1, PML-RARA,
ingly being incorporated into the routine diagnostic eval- and RUNX-RUNXT1.
uation of leukemia and lymphoma. Highly sensitive at the As a result of the t(9;22) in CML, a shortened chromo-
single-nucleotide level, they allow the detection of recur- some 22 (i.e. Philadelphia chromosome) leads to the BCR-
ring gene fusion transcripts and gene mutations. The anal- ABL1 fusion protein. BRC-ABL1 contains the entire catalytic
ysis of DNA using gene panels of targeted exon mutations domain from the tyrosine kinase ABL1, leading to constitu-
allows the concurrent detection of multiple genes in a panel tively increased tyrosine kinase activity and explaining the
reaction (targeted sequencing). RNA sequencing (performed abnormal cellular proliferation observed in CML. Imatinib
using either targeted panels or whole RNA) enables detection and other tyrosine kinase inhibitors specifically target this
of recurring leukemia-specific gene fusion transcripts, and tyrosine kinase activity and exhibit remarkable activity in all
can identify novel gene fusions that define specific leukemia phases of CML.
52 Molecular Hematology

In APL, another fusion protein is formed by the t(15;17)


producing a fusion protein involving the promyelo-
Treatment selection
cytic leukemia and retinoic acid receptor-alpha proteins
The optimal timing of hematopoietic cell transplantation
(PML-RARA). The APL fusion protein is responsible for
(HCT) in the treatment of AML and ALL is still unclear.
arresting the growth of promyelocytes, presumably by
Cytogenetic analysis before commencement of treatment can
aberrant repression of retinoic acid receptor-mediated
guide the choice among post-remission therapies that differ
gene transcription through histone deacetylase-dependent
in effectiveness, acute and chronic morbidity, as well as cost.
chromatin remodeling. Treatment with one of two com-
The majority of AML patients with favorable cytogenetic fea-
pounds, all-trans-retinoic acid (ATRA) or arsenic trioxide,
tures – such as t(8;21) and inv(16) or t(16;16) – can be cured
can bypass the transcriptional repression and activate genes
solely with intensive consolidation chemotherapy. Patients
leading to the terminal differentiation of promyelocytes.
who relapse can be salvaged with HCT in early relapse or
AML with t(8;21) constitutes a clinically distinct sub-
second remission. On the other hand, patients with a chro-
set of AML with a characteristic morphology, portend-
mosome abnormality that portends a bad prognosis – for
ing a favorable prognosis. Molecularly, the t(8;21) forms
instance complex karyotype (≥3 abnormalities) in AML –
a chimeric protein RUNX1-RUNX1T1. The gene RUNX1
are rarely cured with standard therapy and can have drug-
encodes one subunit of a heterodimeric transcription fac-
resistant disease. These patients may benefit from transplan-
tor termed core-binding factor (CBF), shown to be essential
tation in first remission or investigational drugs.
for hematopoiesis. The oncogenic potential of the RUNX1-
RUNX1T1 fusion protein is believed to be due to aber-
rant recruitment of nuclear transcriptional co-repressor
Recurrent cytogenetic abnormalities
complexes, leading to transcriptional repression of normal
in MDS
RUNX1 target genes.
All of the fusion proteins mentioned are specific for the A diagnosis of MDS is usually made by evaluation of the
disease. Thus, they can be used for the initial diagnosis as well bone marrow and peripheral smear in the appropriate clin-
as for detecting MRD. ical context. In patients with otherwise unexplained refrac-
tory cytopenia and no morphological evidence of dysplasia,
certain cytogenetic abnormalities are sufficient for the diag-
nosis of MDS.
Karyotypic evolution
At the time of diagnosis, approximately half of patients
with MDS have clonal chromosomal abnormalities. These
Additional cytogenetic abnormalities may help to detect
chromosomal changes can manifest as structural abnormal-
transformation to a more advanced disease stage or to a more
ities involving one chromosome (e.g. inversion, interstitial
aggressive malignancy. As evidenced by subclones on cyto-
deletion), numerical changes (e.g. monosomy or trisomy), or
genetic analysis, clonal heterogeneity and evolution are fre-
a balanced translocation between two chromosomes. About
quent in AML and constitute an independent adverse prog-
15% of patients have multiple abnormalities and thus exhibit
nostic marker in cytogenetically adverse-risk AML.
complex karyotypes (≥3 or more abnormalities). During the
course of the disease, additional chromosomal aberrations
may develop on top of previous ones or emerge in patients
Clinical implications with no previous karyotypic abnormality. These changes are
invariably associated with poor prognosis and progression to
Both the clinical course as well as the likelihood of respond- acute leukemia. In contrast to AML, none of the chromoso-
ing to particular treatments can be predicted by specific mal abnormalities is specific for MDS and they are also found
genetic abnormalities (e.g. retinoic acid or arsenic trioxide in other myeloid diseases.
in APL). The prognostic information derived from cyto- The karyotypic abnormalities that are most commonly
genetic analysis is often independent of other clinical fea- found in MDS are del(5q), del(7q) or monosomy 7 (−7),
tures. Patients who have less favorable clinical or cytogenetic trisomy 8 (+8), and del(20q), as well as loss of the Y chromo-
characteristics may be better treated with more intensive or some. Although the same cytogenetic lesions also occur in
investigational therapies, while those with more favorable de novo AML, the balanced translocations typically found in
prognostic features may be treated with standard regimens. AML are very uncommon in MDS. Rather, the chromosomal
Moreover, the disappearance of a chromosomal abnormality abnormalities in MDS are usually unbalanced, indicating
present at diagnosis signifies complete remission following gain or loss of chromosomal material. Cytogenetic abnormal-
treatment, while its reappearance invariably indicates relapse ities are even more prevalent in therapy-related MDS/AML,
of the disease. as up to 70% of cases have chromosomal abnormalities,
Molecular diagnostics and risk assessment in myeloid malignancies 53

particularly deletions of chromosomes 5 and 7 or chromo- About 50–60% of patients with de novo AML have
some 17p. Deletion of the long arm of chromosome 5 with or abnormal karyotypes if standard banding techniques are
without additional karyotypic abnormalities is not only the used. The most common abnormalities seen on karyotype
most frequent chromosomal aberration in MDS, it is the only are t(15;17)(q24.1;q21.1), trisomy 8, t(8;21)(q22;q22.1),
one constituting its own entity in the WHO classification. rearrangements of 11q, and inv(16)(p13.1q22)/t(16;16)
The term “5q– syndrome” refers to a distinctive type (p13.1;q22). Regardless of blast count, AML is diagnosed if
of MDS that is defined by isolated del(5q) or del(5q) plus any of the following cytogenetic abnormalities are present:
one other abnormality, with the exception of −7/del(7q). A r AML with t(8;21)(q22;q22.1); RUNX1-RUNX1T1 (previ-
characteristic hallmark on morphology examination is the ously AML1-ETO). Found in approximately 7% of adults
finding of megakaryocytes with hypolobated nuclei. The with newly diagnosed AML, it is the most frequent abnor-
deletions on chromosome 5 are large interstitial deletions mality in children with AML. This cytogenetic abnormal-
resulting in the retention of only one normal allele of all genes ity has a distinct morphological phenotype and heralds a
contained within the deleted segment – this is referred to as favorable prognosis in adults, but a poor prognosis in chil-
a haploinsufficient state. Numerous genes on the long arm of dren.
chromosome 5 have been implicated in MDS pathogenesis r APL with t(15;17)(q24.1;q21.2); PML-RARA. Highly spe-
and its response to specific therapies. cific for APL, the translocation results in a novel PML-
MDS cases with del(5q) as the sole chromosomal abnor- RARA fusion protein that is thought to arrest differenti-
mality carry a relatively good prognosis and frequently ation at the promyelocyte stage. APL constitutes a medi-
respond to treatment with lenalidomide. In contrast, mono- cal emergency carrying a high rate of early mortality, often
somy 5 or del(5q) with more than one additional chromoso- due to hemorrhage from disseminated intravascular coag-
mal change is associated with advanced MDS and portends a ulation. Treatment should be started as soon as the diag-
worse prognosis. nosis is suggested by cytological criteria. Cytogenetic and
With the notable exception of MDS with isolated del(5q), molecular genetic analysis can then confirm the diagno-
other chromosomal aberrations in MDS have not correlated sis. If prompt treatment with ATRA or arsenic trioxide is
with specific morphological or clinical entities. However, started, these patients have an excellent prognosis.
they do play a substantial role in pathogenesis and progno- r AML with inv(16)(p13.1q22) or t(16;16)(p13.1;q22);
sis and have a major impact in the clinical management of CBFB-MYH11. Representing approximately 5% of newly
patients. diagnosed AML, its classical bone marrow morphology
Certain cytogenetic abnormalities in MDS are used in the typically shows monocytic and granulocytic differenti-
prediction of survival and progression to AML. The presence ation with abnormal eosinophils. Patients with inv(16)
or absence of specific chromosomal aberrations is incorpo- generally respond well to intensive chemotherapy.
rated into several prognostic scoring systems for MDS, such
as the original and revised International Prognostic Scoring While these cytogenetic aberrations constitute a diagno-
Systems (IPSS and IPSS-R), the World Health Organization sis of AML irrespective of blast count, there are additional
(WHO) Prognostic Scoring System, and the MD Anderson subtypes of AML where certain genetic abnormalities bear
Cancer Center MDS model. The most widely used system, prognostic significance. However, these require bone marrow
IPSS-R, is shown in Figure 4.1. blast counts of greater than 20% for diagnosis of AML:
Importantly, cytogenetic analysis cannot predict individ- r AML with t(9;11)(p21.3;q23.3); KMT2A-MLLT3. By far
ual MDS patient outcome due to the fact that many patients the most promiscuous translocation partner in AML
succumb to persistent pancytopenia, irrespective of progres- involves 11q23.3. More than 100 different recurring rear-
sion to AML. rangements involving 11q23.3 have been described in
AML, especially among the monoblastic and myelomono-
cytic types. Of these, t(9;11) is the most common abnor-
Recurrent cytogenetic aberrations in AML
mality and involves the KMT2A gene and MLLT3 (AF9).
A key component in the evaluation of all patients with newly Patients generally have an intermediate response to stan-
diagnosed or suspected AML is the cytogenetic analysis of dard therapy.
metaphase cells. In some cases, specific cytogenetic abnor- r AML with t(6;9)(p23;q34.1); DEK-NUP214. Seen in about
malities correspond to morphologically and clinically dis- 1% of patients with newly diagnosed AML, this is mor-
tinct subsets of the disease. Because of this, the WHO classi- phologically associated with basophilia, pancytopenia, and
fication of tumors of the hematopoietic and lymphoid tissues dysplasia and frequently associated with FLT3-ITD muta-
incorporates genetic findings in addition to morphological, tions. Patients generally have a poor response to standard
immunophenotypic, and clinical features to define distinct therapy, but it is currently unclear whether the association
subtypes of AML. with FLT3-ITD or the lesion itself is responsible for this.
54 Molecular Hematology

Calculation of risk score

Cytogenetic risk group

Very good 0 del(11q), -Y


Good 1 normal, del(5q) alone or with one other anomaly, del(12p), del(20)
Intermediate 2 +8, del(7q), i(17q), +19, +21, other abnormalities in two or more independent clones
Poor 3 der(3q), –7, double abnormality with –7 or del(7q), complex with 3 abnormalities
Very poor 4 complex with >3 abnormalities

Bone marrow blast percentage

≤2% 0
2–5% 1
5–10% 2 Assigning IPSS-R risk group
>10% 3
Time until
Median 25%
%of survival develop
Total score IPSS-R risk group patients (years) AML (years)
Hemoglobin (g/dL)
0–1.5 Very low 19 8.8 Not reached
≥10 0 1.5–3 Low 38 5.3 10.8
8–10 1 3–4.5 Intermediate 20 3 3.2
<8 1.5 4.5–6 High 13 1.6 1.4
>6 Very high 10 0.8 0.7
Platelet count (G/L)

≥100 0
50–100 0.5
<50 1

Absolute neutrophil count (G/L)

≥0.8 0
<0.8 0.5

Fig. 4.1 International Prognostic Scoring System Revised (IPSS-R). Karyotypic abnormalities, bone marrow blast percentage, and severity of
peripheral blood cytopenias are scored and used to assign myelodysplastic syndrome (MDS) patients to one of five risk groups exhibiting significant
differences in median survival and probability of developing acute myeloid leukemia (AML).
Source: Based on Greenberg, P.L., Tuechler, H., Schanz, J. et al. (2012). Revised international prognostic scoring system for myelodysplastic
syndromes. Blood 120(12): 2454–2465.

r AML with inv(3)(q21.3q26.2) or t(3;3)(q21.3;q26.2); The most common subtype of therapy-related MDS/AML
MECOM/EVI1. Accounting for approximately 1% of AML constitutes patients who were being treated with alkylating
cases, inv(3) aberrations are associated with increased agents. These patients often have abnormalities in chromo-
atypical megakaryocytes in the bone marrow and a poor somes 5 and/or 7.
response to therapy. As a consequence of the cytogenetic Patients being treated with DNA topoisomerase II
abnormality, the MECOM gene is overexpressed, likely inhibitors (e.g. etoposide, doxorubicin, mitoxantrone,
explaining the peripheral blood thrombocytosis and bone epirubicin) account for the second most frequent subtype
marrow morphology. of therapy-related AML. Frequently, balanced translocations
involving the KMT2A gene at 11q23.3, or the RUNX1 gene
at 21q22.1, are found in these AML cases.
Therapy-related chromosomal
aberrations in secondary AML Recurrently mutated genes in MDS

Patients who develop therapy-related secondary MDS or The recent advances in less expensive high-throughput
AML after chemotherapy and/or radiation therapy for an genome sequencing technologies have enabled the discovery
earlier disease have also been found to carry characteristic of genetic lesions that drive the pathogenesis of the majority
chromosomal abnormalities. of human cancers, including the MDS.
Molecular diagnostics and risk assessment in myeloid malignancies 55

Based on panels of genes that were previously found to Epigenetic regulators


be mutated in other myeloid diseases, a number of studies
Another set of genes found to be recurrently mutated in MDS
have described the mutational landscape of somatic point
are genes that are involved in epigenetic regulation. Epige-
mutations in MDS. According to current data, up to 90% of
netic regulation is the result of post-translational modifica-
patients bear mutations in at least one recurrently mutated
tion of either histones or the DNA itself.
gene, and it is believed that the remaining 10% of patients
have mutations in genes yet to be identified.
MDS is the result of the sequential accrual of somatic Histone modification
mutations in HSCs. Based on calculations, these stem cells
amass approximately 1.3 mutations in exons every decade, Enzymes can covalently modify the tails of histones, leading
but the majority of these are innocent bystanders with no to changes in the structure of chromatin and affecting the
functional alteration. However, this calculation presumes binding of protein involved in gene regulation. Among the
that the rate of mutation is constant – in reality, the process of central regulators of transcriptional silencing in development
mutational accrual could be dynamic, as one mutation might are the Polycomb repressive complexes (PRCs), composed
affect the acquisition of subsequent mutations. of two separate protein complexes, PRC1 and PRC2. Their
The spectrum of recurrent somatic mutations in MDS and action on histones H2A and H3 results in the compaction
other myeloid diseases can be grouped according to their bio- of chromatin. Genes that are part of both protein complexes
logical pathways, as in the following. have been found to be mutated in MDS. Mutation in two
components of PRC1 – that is, BCOR and BCORL1 – has been
shown to be recurrently mutated in 5% of MDS cases and to
Mutations in splicing factors portend an unfavorable prognosis. EZH2 is mutated in about
A hallmark of eukaryotes, the alternative splicing of pre- 5% of MDS patients; the resulting protein is part of the cat-
mRNA enables the generation of different proteins from the alytic subunit of PRC2 and studies in transgenic mice have
same gene. Interestingly, one of the hallmarks of cancer is confirmed that loss of Ezh2 results in MDS.
aberrant alternative splicing. Recurrent mutations in genes While in itself ASXL1 is not part of the PRC, it has been
that are part of the spliceosome have been found in up to 60% shown to indirectly affect PRC2, leading to deubiquitylation
of cases of MDS. of histone H2A. Approximately 20% of MDS patients carry
The mutations in SF3B1, U2AF1, and SRSF2 are invariably recurrent mutations in ASXL1. Mutations in BCOR, EZH2,
found within defined hotspots of these genes, leading to and ASXL1 have been shown to disrupt normal hematopoi-
loss of function and altering the operation of the splicing etic differentiation, likely explaining their putative role in
machinery. Mutations within the splicing componentry dysplasia and cytopenia.
most commonly affect SF3B1, occurring in up to 90% of
MDS patients with ring sideroblasts (RARS). In this context,
DNA methylation
SF3B1 mutations are associated with a good prognosis. It
is believed that the ring sideroblast phenotype is the result The regulatory regions of the DNA can either be accessible
of aberrant splicing of genes central to iron homeostasis. or inaccessible. Which state is favored is largely controlled
The second most frequent splicing factor mutation in MDS by the methylation of cytosines in repetitive CpG elements.
affects the SRSF2 gene; this gene is commonly mutated in the DNA methyltransferases (DNMTs) add methyl groups,
overlap syndromes of MDS/myeloproliferative neoplasms while the TET family of proteins demethylate in a multistep
(MPN). Mutations in SRSF2 were demonstrated to alter process. Mutations in DNMT3A leading to loss of function
binding of the protein to splice enhancers, with resulting of the protein are found in approximately 15% of MDS cases.
mis-splicing of several important downstream genes, one TET2 is one of the most frequently mutated genes, found in
of which is EZH2. In 10–15% of MDS cases mutations are up to 30% of MDS cases. The state of hypermethylation of
found in U2AF1. Resulting loss of function is reported to cytosines at enhancer sites is believed to repress several genes
lead to increased exon skipping. SRSF2 and U2AF1 have with important roles in myeloid differentiation. Of note is
not been associated with a similar favorable prognosis to that DNMT3A and TET2 are both expressed in HSCs – in
SF3B1. Despite these findings, no single mis-spliced isoform a complementary, interconnected, yin-and-yang kind of
has hitherto been implicated in the pathogenesis of MDS. fashion – having essential roles in self-renewal and myeloid
Moreover, studies using mouse models of splicing genes differentiation.
have not yielded similar patterns of altered splicing, either An initially puzzling observation was how mutations
between mice and humans or between individual mice. The in enzymes involved in the citric acid cycle play a role
role that these mutations play in MDS pathobiology is per- in MDS and leukemia pathogenesis. It was found that
haps unrelated to their known function within the splicing cells with mutations in IDH1 and IDH2 accumulated a
machinery. novel oncometabolite, 2-hydroxyglutarate (2-HG), instead
56 Molecular Hematology

of alpha-ketoglutarate. 2-HG was shown to diffuse to the (PV/ET), the frequencies with which these genes are mutated
nucleus leading to inhibition of, among others, TET2, and in MDS is quite low. Mutations in components of the MAPK
thereby also affecting DNA methylation. Mutations in IDH1 pathway, for example NRAS, KRAS, NF1, and PTPN11, are
or IDH2 occur in ∼5% of patients with MDS and portend an the most prevalent in MDS, and together account for approx-
unfavorable clinical outcome. While the oncometabolite was imately 10% of MDS cases. In general, most mutations in sig-
shown to be sufficient to promote leukemogenesis, the effects naling pathway components lead to the constitutive activa-
were found to be reversible. This has led to the development tion of the protein product. Of note is that these mutations
of 2-HG inhibitors, which are currently being evaluated in usually occur late in the course of the disease in more aggres-
clinical trials. sive subclones that signify the transition to secondary AML.

Cohesin complex TP53


The formation of a specific protein complex that holds The single most frequently mutated tumor suppressor gene
together sister chromatids preventing the collapse of the among all human cancers is tumor protein 53 (TP53). In the
replication fork is an important part of DNA repair. This pro- clinical condition known as Li–Fraumeni syndrome, humans
tein complex is made up of the so-called cohesins; that is, are born with a single mutant allele of TP53 and these indi-
STAG2, SMC3, SMC1A, and RAD21. In aggregate, inactivat- viduals have a drastically increased risk for many types of
ing mutations in cohesin genes are found in 15% of MDS cancer, including MDS and AML. In patients with MDS,
cases. Perhaps somewhat counterintuitive given their func- somatic mutations of TP53 are usually associated with high
tion, cohesin mutations have not been associated with chro- blast counts, low platelet counts, a complex karyotype, and
mosomal abnormalities. Rather, cohesin mutations are now previous exposure to chemotherapy. Dubbed “guardian of
believed to lead to altered gene expression due to a failure to the genome,” p53 functions as a mediator of cellular stress.
stabilize DNA loops that normally facilitate the interaction of In the appropriate context, p53 increases pro-apoptotic genes
promoters and distant enhancers. and mediates cell cycle arrest. Accumulation of additional
genomic aberrations and chromosomal instability together
with chemotherapy resistance observed in patients with inac-
Transcription factors
tivation of the p53 pathway are likely due to inadequate acti-
A small group of hematopoietic transcription factors have vation of the DNA damage response and loss of cell cycle
been found to be recurrently mutated in MDS. Since arrest measures, respectively.
germline mutations in RUNX1, GATA2, and ETV6 had been The negative prognostic impact of TP53 mutations had
linked to the development of MDS and AML in inher- previously been demonstrated in a number of hematolog-
ited bone marrow failure disorders, these genes were also ical malignancies, including MDS. Mainly seen in high-
included in targeted sequencing panels for the study of MDS. risk and secondary (therapy-related) MDS, TP53 mutations
Found in approximately 10% of MDS cases, mutations in occurred frequently in the context of complex karyotypes,
RUNX1 are often associated with severe thrombocytopenia. and have also been associated with disease progression and
The protein forms the subunit of the CBF and is responsi- poor response to therapy. In contrast, TP53 mutations were
ble for DNA binding. RUNX1 has several target genes with rarely observed in lower-risk MDS. However, these studies
important roles in hematopoiesis. suffered from small sample size and the inherent low sensi-
GATA2 is a zinc finger protein highly expressed in tivity associated with the use of conventional Sanger sequenc-
HSCs and essential for normal hematopoietic differentiation. ing techniques. When using sensitive deep sequencing tech-
Somatic mutations in GATA2 account for only 1% of MDS niques, TP53 mutations could be found in approximately
cases. The transcription factor Wilms tumor protein WT1 20% of low-risk del(5q) MDS patients. These mutations can
has been shown to recruit TET2 to specific loci and is mutated be detected years before disease progression and confer an
in fewer than 5% of cases of MDS. There are additional tran- increased risk of evolution to overt leukemia.
scription factors with mutation frequencies below 1%.

Recurrently mutated genes in AML


Cell signaling genes
Genes for components of cell signaling networks were found The observation that the mutational landscapes of MDS and
to be mutated in a host of other myeloid malignancies AML show a significant overlap underlines the notion that
and were thus obvious candidates to study in MDS. How- MDS and AML represent stages of a disease continuum. As
ever, compared to AML, chronic myelomonocytic leukemia with MDS, factors that contribute to splicing, transcription,
(CMML), or polycythemia vera/essential thrombocythemia epigenetic regulation, cohesion, and cell signaling are
Molecular diagnostics and risk assessment in myeloid malignancies 57

frequently found mutated in AML, as is the gene TP53. DNMT3A and FLT3-ITD mutations confers an adverse out-
However, there are some mutations representing late events come. Most interestingly, a strong prognostic impact was
in AML pathogenesis which are mostly not reported in MDS only detectable when all three genes were mutated. In con-
cases, notably mutations in NPM1, CEBPA, and FLT3. trast, a NPM1 mutation in the presence of mutated DNMT3A
Nucleophosmin 1 (NPM 1) participates in many different and NRAS-G12 or NRAS-G13 as well as FLT3 wild type is a
cellular processes. It acts as a histone chaperonin and inter- positive prognostic indicator.
acts with BRCA2 to regulate centromere duplication. It is also Three recurrently mutated genes define AML disease cate-
implicated in proliferation and cell survival, due to its regula- gories among the group “AML with recurrent genetic abnor-
tion of the tumor suppressor factors alternate reading frame malities.” Aside from the respective mutation, blast counts
(ARF) and TP53, as well as the proapoptotic protein kinase greater than 20% are required to meet the diagnostic criteria
R. Mutations usually occur in exon 12 of NPM1. for AML. There are no specific morphological features asso-
CEBPA is a myeloid transcription factor. Mutations in ciated with either of the three molecularly defined AML sub-
CEBPA result in inhibition of granulocytic maturation. Since types:
the introduction of “AML with mutated CEBPA” as a provi- r AML with mutated NPM1. This is found in 27–35% of all
sional entity in the 2008 edition of the WHO classification, adult AML cases and in 45–64% of adult cases with a nor-
it has become evident that cases with monoallelic and cases mal karyotype. Multilineage dysplasia may be present, but
with biallelic CEBPA mutation differ in terms of gene expres- does not take precedence over the NPM1 mutation. While
sion and prognosis. The overall favorable outcome was only the individual prognosis also depends on the genomic con-
observed when both CEBPA alleles were mutated, hence the text, as already discussed, patients with this subtype gener-
disease category was adapted in the current WHO classifica- ally correspond well to induction therapy and have a good
tion in 2017. prognosis.
Although FLT3 mutations do not define an AML disease r AML with biallelic mutation of CEBPA. As is the case for
category, it is strongly recommended to investigate the AML with mutated NPM1, this subtype is also associated
FLT3 mutation status at diagnosis. FLT3 encodes a receptor with a normal karyotype and a favorable outcome. The
tyrosine kinase, which acts as a receptor for the cytokine presence of multilineage dysplasia does not influence prog-
FLT3LG. Upon ligand activation, FLT3 regulates factors of nosis. FLT3-ITD and GATA2 zinc finger 1 are recurrently
both JAK-STAT and RAS signaling pathways. Mutations of found co-mutated in this AML subtype; however, their
FLT3 can occur within the tyrosine kinase domain (TKD) prognostic impact is still unclear.
and internal tandem duplications (ITDs) of FLT3. Both r AML with mutated RUNX1. This subtype was included
render FLT3 constitutively active, which promotes prolif- as a provisional entity in the revised 2016 edition of
eration and cell survival. The presence of a FLT3 mutation the WHO classification and should only be diagnosed
is generally associated with an adverse outcome. Aside to if AML with other genetic abnormalities, therapy-related
its prognostic value, early detection of a FLT3 mutation has myeloid neoplasms, or AML with myelodysplasia-related
clinical implications, as patients may benefit from FLT3 changes can be ruled out. AML with mutated RUNX1 may
inhibitors, such as midostaurin. display karyotypic aberrations and co-mutations are fre-
Mutations in AML are more prevalent than in MDS. In quently found. Among the frequently co-mutated genes
almost every case (96% to >99%) at least one mutation is are ASXL1, KMT2A-PTD, FLT3-ITD, IDH1-R132, and
readily detected. The vast majority of cases bear two or more IDH2-R140 or IDH2-R172. Some studies report an adverse
mutations, so that gene–gene interactions have to be taken outcome for this AML subtype. Additionally, a concomi-
into account, especially for prognostic evaluation. tant ASXL1 mutation worsens the prognosis.
The mutational landscape in AML displays a highly spe-
cific co-mutation pattern: while some mutations are fre-
quently found co-mutated, others are mutually exclusive. A
good example is the recurrently mutated gene NPM1. Co- Further reading
mutations of NRAS are frequently detected in amino acid
residues G12 and G13, but never for Q61. The same holds Abelson, S., Collord, G., Ng, S.W.K. et al. (2018). Prediction of acute
myeloid leukaemia risk in healthy individuals. Nature 559: 400–404.
true for the co-mutation pattern of NPM1 and IDH2: co-
Arber, D.A., Orazi, A., Hasserjian, R. et al. (2016). The 2016 revision
mutations are restricted to IDH2-R140 and not observed for to the World Health Organization (WHO) classification of myeloid
IDH2-R172. neoplasms and acute leukemia. Blood 127: 2391–2405.
The importance of the genetic context for prognosis Bacher, U., Schnittger, S., Macijewski, K. et al. (2012). Multilineage dys-
is increasingly recognized. Depending on occurring co- plasia does not influence prognosis in CEBPA-mutated AML, sup-
mutations, a NPM1 mutation can be associated with a favor- porting the WHO proposal to classify these patients as a unique
able or an inferior prognosis. The presence of concomitant entity. Blood 119: 4719–4722.
58 Molecular Hematology

Brandts, C.H., Sargin, B., Rode, M. et al. (2005). Constitutive activation Kunchala, P., Kuravi, S., Jensen, R. et al. (2018). When the good go
of Akt by Flt3 internal tandem duplications is necessary for increased bad: mutant NPM1 in acute myeloid leukemia. Blood Rev. 32: 167–
survival, proliferation, and myeloid transformation. Cancer Res. 65: 183.
9643–9650. Pabst, T., Mueller, B.U., Zhang, P. et al. (2001). Dominant-negative
Cancer Genome Atlas Research Network (2013). Genomic and epige- mutations of CEBPA, encoding CCAAT/enhancer binding protein-
nomic landscapes of adult de novo acute myeloid leukemia. N. Engl. alpha (C/EBPalpha), in acute myeloid leukemia. Nat. Genet. 27: 263–
J. Med. 368: 2059–2074. 270.
Chen, W., Drakos, E., Grammatikakis, I. et al. (2010). mTOR signaling Pang, Q., Christianson, T.A., Koretsky, T. et al. (2003). Nucleophosmin
is activated by FLT3 kinase and promotes survival of FLT3-mutated interacts with and inhibits the catalytic function of eukaryotic initia-
acute myeloid leukemia cells. Mol. Cancer 9: 292. tion factor 2 kinase PKR. J. Biol. Chem. 278: 41709–41717.
Choudhary, C., Schwable, J., Brandts, C. et al. (2005). AML-associated Papaemmanuil, E., Gerstung, M., Bullinger, L. et al. (2016). Genomic
Flt3 kinase domain mutations show signal transduction differences classification and prognosis in acute myeloid leukemia. N. Engl. J.
compared with Flt3 ITD mutations. Blood 106: 265–273. Med. 374: 2209–2221.
Dohner, H., Estey, E., Grimwade, D. et al. (2017). Diagnosis and man- Paschka, P., Schlenk, R.F., Gaidzik, V.I. et al. (2015). ASXL1 mutations
agement of AML in adults: 2017 ELN recommendations from an in younger adult patients with acute myeloid leukemia: a study by the
international expert panel. Blood 129: 424–447. German-Austrian Acute Myeloid Leukemia Study Group. Haemato-
Dohner, H., Weisdorf, D.J., and Bloomfield, C.D. (2015). Acute myeloid logica 100: 324–330.
leukemia. N. Engl. J. Med. 373: 1136–1152. Schnittger, S., Dicker, F., Kern, W. et al. (2011). RUNX1 mutations are
Falini, B., Macijewski, K., Weiss, T. et al. (2010). Multilineage dysplasia frequent in de novo AML with noncomplex karyotype and confer an
has no impact on biologic, clinicopathologic, and prognostic features unfavorable prognosis. Blood 117: 2348–2357.
of AML with mutated nucleophosmin (NPM1). Blood 115: 3776– Schuurhuis, G.J., Heuser, M., Freeman, S. et al. (2018). Mini-
3786. mal/measurable residual disease in AML: consensus document from
Gaidzik, V.I., Bullinger, L., Schlenk, R.F. et al. (2011). RUNX1 mutations ELN MRD Working Party. Blood 131: 1275–1291.
in acute myeloid leukemia: results from a comprehensive genetic and Stone, R.M., Mandrekar, S.J., Sanford, B.L. et al. (2017). Midostaurin
clinical analysis from the AML Study Group. J. Clin. Oncol. 29: 1364– plus chemotherapy for acute myeloid leukemia with a FLT3 mutation.
1372. N. Engl. J. Med. 377: 454–464.
Greenberg, P.L., Tuechler, H., Schanz, J. et al. (2012). Revised inter- Swerdlow, S.H., Campo, E., Pileri, S.A. et al. (2016). The 2016 revision
national prognostic scoring system for myelodysplastic syndromes. of the World Health Organization (WHO) classification of lymphoid
Blood 120 (12): 2454–2465. neoplasms. Blood 127: 2375–2390.
Greif, P.A., Dufour, A., Konstandin, N.P. et al. (2012). GATA2 zinc fin- Taskesen, E., Bullinger, L., Corbacioglu, A. et al. (2011). Prognostic
ger 1 mutations associated with biallelic CEBPA mutations define a impact, concurrent genetic mutations, and gene expression features
unique genetic entity of acute myeloid leukemia. Blood 120: 395–403. of AML with CEBPA mutations in a cohort of 1182 cytogenetically
Grimwade, D., Ivey, A., and Huntly, B.J. (2016). Molecular landscape of normal AML patients: further evidence for CEBPA double mutant
acute myeloid leukemia in younger adults and its clinical relevance. AML as a distinctive disease entity. Blood 117: 2469–2475.
Blood 127: 29–41. Wang, H.F., Takenaka, K., Nakanishi, A., and Miki, Y. (2011). BRCA2
Jongen-Lavrencic, M., Grob, T., Hanekamp, D. et al. (2018). Molecular and nucleophosmin coregulate centrosome amplification and form
minimal residual disease in acute myeloid leukemia. N. Engl. J. Med. a complex with the Rho effector kinase ROCK2. Cancer Res. 71:
378: 1189–1199. 68–77.
Chapter 5 Molecular basis of acute
lymphoblastic leukemia
Bela Patel & Fiona Fernando
Centre of Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK

Introduction, 59 Developmental origins, 62


Molecular methodologies, 59 Molecular genetic subsets, 62
Primary cytogenetic subgroups, 60 Molecular evaluation of Minimal Residual Disease, 69
Biological consequences of chromosomal alterations, 60 Conclusion, 70
Additional cooperating genetic events, 60 Further reading, 70

Introduction Molecular methodologies

Cure rates for childhood acute lymphoblastic leukemia Tools for interrogating the molecular pathophysiology of
(ALL) have markedly increased over the last four decades ALL encompass genomic, transcriptomic, and epigenomic
and now reach >80%. Despite these undoubtedly excellent methods, which synergistically contribute to in-depth molec-
results, challenges remain, particularly for relapsed ALL, ular profiling. The two time-honored methods for accurately
which is a major contributor to childhood mortality. In adult determining recurrent chromosomal abnormalities routinely
ALL, survival gains have been less pronounced despite the applied in clinical practice are chromosomal G banding
use of optimized approaches, and currently only 50% achieve on metaphase nuclei and fluorescence in situ hybridization
long-term survival. In both groups, treatment toxicity is a sig- (FISH). These methods evaluate macroscopic genetic change
nificant limitation leading to recurring short- and long-term and will discriminate major molecular subgroups in ALL.
morbidity. Thus, along with a clear need to improve treat- However, they have limited resolution.
ment efficacy, there is a requirement also to mitigate treat- Over the last decade, several technologies have evolved
ment toxicity. The need for more rationalized approaches to that are capable of mapping genetic change at both the
treatment is therefore pressing. submicroscopic and genome-wide levels. These method-
Recent efforts to molecularly characterize ALL have greatly ologies include microarray profiling of single-nucleotide
informed this priority area. Novel molecular-oncogenetic polymorphisms and comparative genomic hybridization to
groups have emerged providing new prognostic and pre- detect DNA copy number change, corresponding to ∼kilo
dictive biomarkers for informing novel and optimized mega base regions of genomic amplification and/or deletions
treatments. Moreover, molecular profiling has paved the way in potential candidate oncogenes and tumor suppressors.
for the discovery of key pathways essential for the develop- The development of next-generation high-throughput
ment and progression of ALL, thereby contributing essential sequencing technologies has paved the way for massively
knowledge not only of disease biology, but also of therapy parallel sequencing of targeted gene loci, the protein-coding
failure. Current advances in our understanding of the genetic exome and whole genome itself contributing an in-depth
basis of ALL and therapy failure are summarized in this chap- insight into the global cancer genome. Complementing these
ter, with particular emphasis given to how such insights are DNA approaches are methods that evaluate the transcrip-
evolving opportunities for clinical translation. tome and epigenome, including gene expression microarray
analysis together with direct mRNA sequencing. Insight into
the epigenomic mechanisms regulating gene expression in
leukemia has proceeded through high-density DNA methy-
lation arrays detecting CpG nucleotide status at transcrip-
tional start sites, as well as through chromatin immunopre-
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. cipitation assays (CHIP–CHIP array or CHIP sequencing),
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.

59
60 Molecular Hematology

where DNA-associated proteins regulating locus-specific chromosomal mutation. The single most common cause of
gene expression can be directly profiled. Collectively, these chromosomal mutation is chromosomal translocation, for
methods contribute an unprecedented depth of knowledge example t(12;21), t(9;22). Deletions, inversions, and isochro-
about the acquired molecular defects and genomic landscape mosomes represent further examples of structural chromo-
of ALL. somal change; however, these alterations do not give rise to a
recurrent entity in ALL and therefore are not currently rec-
ognized as a major cytogenetic subset.
The process of chromosomal translocation arises from
Primary cytogenetic subgroups
chromosomal breakage followed by recombination of sepa-
rated fragments without any loss or duplication of genetic
Altogether, approximately ∼30% of children with ALL and
material (i.e. balanced rearrangements). The biological con-
a further 40% of adults demonstrate a gross chromosomal
sequences of chromosomal displacement depends on the
alteration as a defining feature of their disease. The detection
individual gene targets affected and are therefore consid-
of a gross chromosomal anomaly relies on microscopic eval-
ered separately, but as a broad principle the net effect is dis-
uation of G-banded metaphase chromosomes, or the appli-
ruption in the homeostatic function of one of the affected
cation of locus-specific and centromeric FISH gene probes,
genes (Figure 5.2). It is also important to note that while the
together with targeted molecular techniques for determina-
pathogenic consequences of chromosomal rearrangements
tion of cytogenetically cryptic lesions such as t(12;21). These
have undoubtedly provided insight into the early initiation
tools are integral to the primary diagnostic evaluation of ALL,
of leukemogenesis, recently completed genome-wide stud-
as they assist in formalizing the diagnosis by determining the
ies have supplemented this understanding by defining addi-
presence of an ALL-specific chromosomal aberration, which
tional mutational targets, which collaborate with primary
in turn also informs the underlying prognosis.
acquired chromosomal defects (reviewed later) to establish
A list of the recurring cytogenetic abnormalities reported
the full disease phenotype.
in ALL, their prevalence, and prognostic association is pro-
vided in Figure 5.1. The nature of the chromosomal lesion
varies according to whether B- or T-lineage disease is present.
There are also striking differences in the distribution of cyto-
Additional cooperating genetic events
genetic abnormalities by age. The recurring hyperdiploid and
t(12;21) karyotypes both associate with excellent prognosis
Several lines of evidence suggest that primary chromosomal
and are almost exclusively observed in childhood ALL, where
lesions alone are not the full explanatory cause of ALL.
they account for the two largest cytogenetic subsets. By con-
The first and most compelling is the low concordance of
trast, both these good-risk subtypes are rarely observed in
ALL (10–15%) in monozygotic twins sharing a common
adult ALL. Conversely, the single largest cytogenetic subset
chromosomal translocation. The second is the failure of ALL
in adult ALL is formed by t(9;22), which confers poor out-
chromosomal lesions to induce malignant transformation
come and is seen in up to 25% of adults. The t(9;22) is rarely
in a variety of experimental models, and the last the finding
observed in childhood ALL, accounting for only 2–3% of
of ALL-associated gene rearrangements in the healthy pop-
cases. Hence, good-risk cytogenetic features highly predom-
ulation. Consequently, researchers have sought to identify
inate in childhood ALL, whereas there is an excess of poor-
additional complementary genetic lesions that complete
risk cytogenetic subtypes in adult ALL. Cases of pre-B ALL
disease development.
lacking a well-defined cytogenetic abnormality are now col-
Higher-resolution genetic methods utilizing DNA
lectively referred to as B-other ALL.
microarray and next-generation sequencing have provided
the critical insight here, by revealing an additional landscape
of submicroscopic genome-wide change accompanying ALL
Biological consequences of development. Critical findings are that compared to many
chromosomal alterations epithelial cancers the overall mutation burden in ALL is low,
with ∼6–8 copy number alterations per individual tumor,
The recurring cytogenetic aberrations reported in ALL suggesting that gross genomic instability is not a major
reflect either numerical changes in chromosome complement mechanism of malignancy. Among the alterations identified,
or an abnormality in chromosome structure, both leading to focal gene deletions dominate over gene amplifications and
a gross structural anomaly usually identifiable through con- sequence mutations. The most common targets of genetic
ventional cytogenetics. Both hyper- and hypodiploid ALL alterations are biological pathways controlling lymphoid
are defined by an altered chromosomal count, whereas most development (PAX5, IKZF1 EBF1, LMO2); cell cycle
other cytogenetic subsets have arisen through an acquired (CDKN2A/CDKN2B, TP53); cytokine signaling (CD200,
Molecular basis of acute lymphoblastic leukemia 61

Cytogenetic Molecular Children Adults Major pathogenic


abnormality alteration % % consequence

Pre-B ALL
Chimeric transcription factor with
t(12:21) (p13:q22) ETV6-RUNX1 22 <1 altered properties

High hyperdipoidy Associated with multiple 25-30 10


submicroscopic gene Determined by individual gene
alterations

Deregulated tyrosine kinase


t(9:22) (q34:q11) BCR-ABL1 2 19

t(4:11) (q21:q23) MLL-AF4 3 ~7–10

Other MLLq23

Epigenetic dysregulation
MLL-ENL: (t11:19) (q23:p13) <2 <2
MLL-AF9:
t(9:11) (p22:q23)
t(10:11)q13-14:q14-21)

IgH@–CRLF2,
P2RY8-CRLF2 Dysregulated CRLF2 expression
CRLF2 deregulation ~5–7 6
Associated with multiple
Determined by individual gene
Low hypodiploidy/Near submicroscopic gene
triploidy alterations ~1 4
Chimeric transcription factor with
t(1:19) (q23:p13) TCF3-PBX1 ~4 ~2 altered properties

RUNX1 amplification, miR-


802, and genes mapping to the Not well known
iAMP21 Down syndrome critical region 2 <1
ID4 6p22
CEBP Oncogene activation
IgH@-t EPOR 19p13 <5 8

None “B other” - 30 40 –

Pre-T ALL
Oncogene TF upregulation
1p32 deletion (SIL/TAL) ~15–25 Lower
t(1:14) (p34:q11)
t(1:7) (p32:q35) TAL1
(SIL/TAL 25%) (SIL/TAL ~5–10%)
5’ super-enhancer mutations

t(11:14) (p15:q11)
t(7:11) (q34:p15) LMO1
t(11:14) (p13:q11) 10–14 –
Oncogene TF upregulation
t(7:11) (q34:p13)
11p13 deletion LMO2

t(10:14) (q24:q11)
t(7:10) (q35:q24) Oncogene TF upregulation
del(10)(q24q26) HOX11/TLX1 3–7 30

t(5:14) (q35:q32) TLX3/HOX11L2/-BCL11B 20–25 5 Oncogene TF dysregulation


Intermediate risk
PICALM-MLLT10 (CALM-
t(10:11) (p13:q14) AF10) ~10 – Fusion TF oncogene
Adverse risk Fusion TF oncogene
t(11:19)(q23:p13) KMT2A–MLLT1 (MLL-ENL) 1% –

t(7:9)(q34:q34) Notch1 rearrangement <1 – Oncogene activation


good risk
t(8:14)(q24:q11) MYC ~1 – Proto-oncogene activation

NUP214-ABL1 ABL1 3-6 – Deregulated tyrosine kinase

t(6:7)(q23:q34) MYB 3 – Oncogene TF dysregulation

t(8:14)(q24:q11) MYC 1 – Oncogene activation

None - ~50 – –

Fig. 5.1 Cytogenetic subsets in acute lymphoblastic leukemia (ALL) and major molecular abnormalities. TF, transcription factor.
62 Molecular Hematology

Phosphorylation of
tyrosine residues on
multiple substrates

ATP

BCR-ABL
t(9;22)
kinase
BCR-ABL

• Cellular proliferation
• Cell survival
• Reduced stromal
A B dependance
Hybrid fusion gene

A B
Regulatory/enhancer Constitutive activation
Chromosomal Balanced
rearrangement translocation

T-cell
TAL1 target transformation
Molecular alterations (s) t(1;14) +++ genes
TAL1-TCRD
TAL1

Molecular pathogenesis

Fig. 5.2 Recurring targets of submicroscopic gene alterations in pre-B acute lymphoblastic leukemia (ALL) and association with disease subgroups.
ped, pediatric.

BTLA, and BLNK); drug response, such as NR3C gluco- representing an early event in leukemogenesis. Additional
corticoid receptor; as well as tumor suppressors (PTEN and oncogenetic lesions are acquired postnatally, indicating that
RB1), transcriptional activators of hematopoiesis (ETV6 and these are secondary “hits” in ALL development. Altogether
ERG), and epigenetic modifiers. While certain lesions are these findings, together with substantial evidence support-
enriched in specific cytogenetic groups and are considered ing the requirement for additional events other than primary
separately, a prevalent finding in pre-B ALL is inactivating chromosomal lesions, strongly support a multistep etiology
mutations in genes encoding key transcription factors underlying ALL development, wherein primary cytogenetic
involved in B cell development. The two most commonly alterations are founder events occurring very early in ALL
mutated B cell transcription factors are PAX5, observed in evolution, leading to the establishment of a pre-leukemic
>30% (deletions > point mutations > cryptic transloca- clone with an altered propensity for survival, self-renewal,
tions), and IKAROS1, (IKZF1) deletions, found in >10%. or transcription. The disruption in normal cellular behavior
Both transcription factors control cell transition from the endows a selective advantage to pre-leukemic clones which
pro to mature B cell stage, with gene inactivation predicted predisposes for further genetic events affecting multiple inde-
to impair B cell differentiation. pendent cellular pathways (e.g. B cell development), leading
to clone evolution and eventual full-blown clinical disease
development. Thus, the recent expanded knowledge of ALL
oncogenomics, derived in particular through high-resolution
Developmental origins
profiling, has led to a fundamentally enhanced understand-
ing of the evolutionary basis of the disease.
Collectively, these newly characterized molecular lesions not
only provide greater insight into the etiological cause of
ALL, but also significantly advance our understanding of the
natural history of this disease. In this respect, identical twin Molecular genetic subsets
leukemias have served as an ideal model for studying the
temporal sequence of genetic events leading to ALL develop-
B-precursor all
ment. Seminal studies here have identified a frequent prena-
tal origin of chromosomal lesions associated with prolonged Increasing knowledge of chromosomal aberrations and
latency to ALL development consistent with these defects, their molecular counterparts, together with an improved
Molecular basis of acute lymphoblastic leukemia 63

Pathway Gene(s) Frequency

B-cell transcription factor Pax-5 ~30%


>60% Ph-pos ALL
-Inactivating mutations IKZF1 ~30% Ph-neg adult ALL
~15% Ph-neg paed ALL
~30% Ph-like ALL

IKZF2 ~50% low hypodiploid ALL


IKZF3 ~13% near haploid ALL

Cell cycle regulation CDKN2A/B ~25%

~10% Ph-like ALL


Cytokine receptor/kinase
JAK 1/2 >95% CRLF2 mutated
signalling 25% Down syndrome
~5–16% Kinase-activating
-Activating mutations ~50% of Ph-like ALL mutations in Ph-like ALL
CRLF2
>50% Down syndrome
~50% JAK/2 mutated CRLF2, tyrosine kinase
genes (ABL1/2, CSFR1,
Kinase-activating PDGFRB), other JAK-
mutations >90% of Ph-like
STAT targets (e.g. JAK2,
EPOR) and RAS

RTK/RAS ~70% near haploid ALL

Tumour suppressor 90% low hypodiploid ALL


TP53
Inactivating mutations

RB1 ~40% low hypodiploid ALL

Transcriptional regulator
~7%
Inactivating deletions ERG –15% of B other

Fig. 5.3 Mechanisms of aberrant gene activities by chromosomal translocations.

genome-wide understanding of secondary oncogenetic BCR-ABL1 oncogene creation. Depending on the breakpoint,
events in ALL, has defined new molecular entities while also either the p210 or p190 BCR/ABL isoform results, which
enhancing our understanding of disease pathogenesis in can readily be detected ty reverse transcription-polymerase
well-established cytogenetic groups. These are now further chain reaction (RT-PCR). The resulting BCR/ABL1 fusion
discussed and summarized in Figure 5.3. gene encodes a chimeric BCR/ABL1 oncoprotein that
has constitutively activated ABL1 tyrosine kinase enzyme
activity, an activator of cellular signaling. Tyrosine kinases
Philadelphia chromosome positive t(9;22) (q34;q11) ALL
initiate cellular signal transduction cascades by catalyzing
(Ph-pos)
the transfer of the phosphate of adenosine triphosphate
The pathogenic consequence of balanced translocation (ATP) to tyrosine residues on target protein (signaling)
between chromosomes 9 and 22, t(9;22)(q34;q11), leads to a substrates, which drives a signal transduction program to
shortened chromosome 22, also known as the Philadelphia enhance cell proliferation, self-renewal, and reduce apop-
chromosome. Ph-pos ALL is perhaps the best-elucidated tosis – behaviors which typically contribute to malignant
molecular entity of ALL, owing partly to the enhanced transformation.
molecular pathophysiological understanding of chronic Molecular elucidation of BCR-ABL1-mediated oncoge-
myeloid leukemia (CML), which similarly is characterized nesis has blazed the trail for molecularly targeted therapies
by t(9;22) genetic rearrangement. Creation of the Philadel- in the form of tyrosine kinase inhibitors (TKIs). Imatinib, a
phia chromosome (Ph) 22q leads to fusion of the breakpoint first-generation BCR/ABL1 TKI, blocks the binding of ATP
cluster region gene (BCR) at chromosome 22q11 to the to the kinase domain of the BCR-ABL1 oncoprotein, which
Abelson gene (ABL) protein-tyrosine kinase gene at chro- in turn blocks the phosphorylation activity of BCR/ABL1
mosome 9q23 during t(9;22) recombination, with resultant oncoprotein, thereby extinguishing oncogenic cell signaling.
64 Molecular Hematology

Subsequent-generation TKIs nilotinib, dasatinib and pona- (MLL), but by far the most common is AF4, located on chro-
tinib have increased potency for kinase inhibition. Although mosome 4, which undergoes rearrangement with MLL on
TKIs broadly share the same mechanism of action, namely chromosome 11, resulting in t(4;11)(q21;q23) and MLL-AF4
competing with ATP, the phosphorylating entity, at the gene fusion. A typical MLL fusion protein contains the N ter-
catalytic binding site of tyrosine kinase, they differ from minus of MLL encoded by the first 8–13 exons and the C ter-
each other most notably in potency and in their interac- minus of one of over 50 fusion partner genes. MLL rearrange-
tion with either the closed/non-functional (imatinib and ment mediated by t(4;11) (q21;q23) is frequently present in
nilotinib) or open/functional conformation of the kinase infant leukemia, accounting for up to 50% of cases, with
domain (dasatinib). Despite the proven efficacy of TKIs in a diminishing incidence (2% in children, 7–10% in adults)
Ph ALL, resistance can occur by a multitude of mechanisms, thereafter.
including genetic mutations caused by acquired mutations A number of features define MLL-rearranged ALL as a
in the ABL-Kinase domain, which prevent drug binding. distinct clinicobiological subtype, namely poor outcome,
The most intractable ABL1 kinase mutation is the T315I, unique association with a pro-B immunophenotype, and
which confers pan-TKI resistance with sensitivity remaining excessive use of VH6 immunoglobulin gene – an early
to ponatinib alone. Overall, the molecular elucidation of the event in immunoglobulin gene rearrangement as well as
BCR-ABL oncogene is a shining example of how molecular a distint gene expression profile. The major mechanisms
scrutiny can directly inform underlying ALL biology, yield- underpinning MLL-rearranged leukemogenesis have now
ing novel predictors of response to targeted treatments and been clarified and point to disordered epigenetics as a major
facilitate individualization of treatment. mechanism of malignancy. The MLL gene encodes a large
However, unlike CML, where TKI inhibition effects a multifunctional epigenetic regulator (DNA methyltrans-
functional cure, Ph-pos ALL cannot be cured through ferase and histone H3K4 methyltransferase – a positive global
TKI inhibition alone, pointing to important biological regulator of gene transcription providing essential epigenetic
differences between these diseases. One key difference is maintenance of the HOX family of genes, master regulators
the target cell for transformation. ALL represents malignant of hematopoiesis. MLL fusion proteins corrupt the intrinsic
transformation of a lymphoid progenitor, as opposed to the epigenetic regulatory activity, leading to global changes in
hematopoietic stem cell (HSC), which is the cellular target of gene transcription and in particular inappropriate expression
malignant transformation in CML. Furthermore, additional of MLL target genes such as HOXA9 and MEIS1, which act
acquired defects in several tumor suppressor genes typify downstream to suppress differentiation and induce aberrant
Ph-associated ALL versus CML, strongly implicating the self-renewal.
disruption of these genes as contributory to Ph ALL patho- Extensive studies have characterized the mediators of tran-
genesis and aggressiveness. One of the key recurring genetic scriptional and epigenetic perturbations caused by MLL
inactivations associated with Ph ALL involves the IKZF1 mutations. These studies have identified co-factor epigenetic
gene encoding the B cell-specific transcription factor Ikaros, enzymes (LEDGF, DOT1L, CBX8, KDM1A, BRD4, P-TEFb,
leading to severely reduced expression of the functional BET) that assemble with MLL fusion proteins to form mul-
protein. Over 60% of Ph ALL exhibit focal deletions of tiprotein complexes to mediate the epigenetic disturbance.
IKZF1, making this the most prevalent co-occurring genetic The elucidation of these protein associates of MLL fusions
abnormality associated with Ph ALL; point mutations also has opened up several therapeutic opportunities. For exam-
contribute to IKZF1 haploinsufficiency, although these ple, identification of the DOT1L mediator of H3K79 histone
are less frequently observed. Recurring loss of function modification, which is significantly enriched at MLL target
in CDKN2A/B cell cycle control together with PAX5 and genes, has led to the targeting of histone methylation induced
EBF1 B cell development genes are also prevalent (∼50%) by DOT1L. Similarly, the BET family of chromatin adapter
in the Ph ALL subtype and likely contribute to BCR-ABL1 proteins, which associate with oncogenic MLL fusions, can
leukemogenesis. Loss of Ikaros has been shown by some be targeted by novel small-molecule inhibitors that displace
studies to confer a poor prognosis, yielding this as a potential BET proteins from chromatin. Both strategies are currently in
marker for clinical risk stratification. early-phase clinical trials and results are eagerly awaited. Tar-
geting of epigenetic enzymes that associate with MLL fusions
to induce leukemogenic suppression is therefore a promising
MLL-rearranged ALL
therapeutic avenue.
Chromosomal translocations involving the KMT2 gene (also
known as MLL) represent one of the most aggressive forms of
High hyperdiploidy
ALL. A large number of partner genes, AFF1 (AF4), MLLT1
(ENL), MLLT4 (AF6), MLLT3 (AF9), and MLLT10 (AF10), The hyperdiploid karyotype is characterized by mas-
have been shown to recombine with mixed-lineage leukemia sive aneuploidy. The formal definition is an increase in
Molecular basis of acute lymphoblastic leukemia 65

chromosomal copy numbers, with gains of between 51 consequences of chromosomal activation of CRLF2 appear
and 65 chromosomes. Hyperdiploid karyotypes constitute to be activation of the JAK-STAT, ERK, and mTOR/PI3K cel-
25–30% of pediatric ALL, making this one of the largest lular signaling pathways. Furthermore, JAK2 gene mutations
cytogenetic subsets in children. The prognosis of high- are concomitantly present in ∼50% of CRLF2-rearranged
hyperdiploid ALL in children is excellent, with overall ALL, which contributes to hyperactivation of this signal-
survival exceeding 90%. In contrast, high hyperdiploidy ing axis. Although an association of CRLF2-deregulated
occurs less frequently in adult ALL and does not confer ALL with clinical outcome has not been reproducibly con-
a similar magnitude of excellent prognosis, although cer- firmed, some studies in adults demonstrate an association
tain chromosomal amplifications may be associated with with higher rates of treatment failure. More significantly, the
increased survival gain. Chromosomal gains in hyperdiploid discovery of CRLF2 molecular alteration provides a rationale
ALL typically include +X, +4, +6, +10, +14, +17, +18, and target for signal transduction inhibitors of the JAK/STAT and
+21, and can also include trisomies (e.g. +4, +10, +17, PI3K pathways in CRLF2-overexpressing ALL.
+18), which may be specified as additional biomarkers
for improved survival. The pathogenic consequences of
Hypodiploid ALL
chromosomal hyperdiploidy are poorly understood, as the
specific target genes affected are not known, but it is generally Karyotypes with <45 chromosomes are collectively classi-
believed that gene dosage effects contribute to the oncogenic fied as hypodiploid. Further dissection by the degree of
role of a hyperdiploid karyotype. Secondary genetic events hypodiploidy generates two major subgroups: near haploid
(IKZF1 deletions, ETV6, and RAS pathway mutation) have (NH), with 24–31 chromosomes, which is more common,
been identified using high-resolution techniques, suggesting and low hypodiploid, with 32–39 chromosomes. While there
that high hyperdiploidy also arises through a multistep is no distinction in prognosis – both are uniformly poor
process. The presence of these secondary abnormalities has – recent genetic studies reveal variance in the molecular
not been shown to confer prognostic significance. pathologies of these groups. NH ALL is characterized by
a very high frequency of mutations, deletions, amplifica-
t(12;21) (p13;q22) ALL tions, and or/sequence mutations that activate RAS signaling
and several tyrosine receptor kinases (FLT3, NF1, MAPK1,
The t(12;21)(p13;q22) translocation is the most common PTPN11), in addition to a high prevalence of inactivating
chromosomal abnormality in pediatric ALL. The rearrange- alterations of the lymphoid transcription factor IKZF3 gene
ment creates a fusion gene encompassing the non-DNA bind- (∼13%), which is almost exclusive to the NH subtype. By con-
ing region of TEL (ETV6) and almost the entire locus of trast, LH is characterized in >90% of cases by TP53 alter-
AML1 (RUNX1), which retains its coding sequence. ETV6- ations, with further abnormalities of CDKN2A/B, RB1, and
RUNX1 resulting from t(12;21)(p13;q22) is cytogenetically inactivation of IKZF2 (∼50%). Interestingly, TP53 mutations
cryptic and cannot be discerned reliably in G-banded prepa- are constitutional in a significant proportion of childhood LH
rations, therefore FISH or RT-PCR is required for its accu- cases, indicative of a possible underlying Li–Fraumeni syn-
rate detection. TEL and AML1 are two well-characterized drome. The finding of hypodiploid cells harboring activating
transcription factors which are required for development and mutations in RAS signaling pathways provides a rationale for
definitive hematopoiesis. The TEL-AML1 fusion causes per- blockade therapies targeting MAPK/MEK/ERK signaling in
turbations in normal hematopoiesis, enhancing self-renewal this poor-risk ALL subset.
of B cell progenitors and altering HSC homeostasis, but is
insufficient on its own to induce the full leukemia pheno-
type. The multistep model of leukemogenesis which premises T ALL
a pre-leukemia initiating clone has been best demonstrated T ALL accounts for ∼20% of adult and 8–15% of child-
for this unique ALL subtype. hood ALL. Gross chromosomal alteration is less frequently
observed in T- versus B-lineage ALL, with just 50% of cases
harboring a primary lesion. A further contrast is that T ALL–
Cytokine receptor-like factor 2 deregulated ALL
specific abnormalities are not clearly prognostic for relapse
Deregulated expression of CRLF2, resulting from juxtapo- or overall survival, and are therefore not clinically applied
sitioning of the CRLF2 gene to either the Immunoglobu- for risk assignment. The nature of chromosomal aberrancy
lin heavy gene enhancer (IGH@-CRLF2) or the pseudo- in T ALL is dominated by balanced translocations, which
autosomal region or the P2RY8 promoter (P2RY8-CRLF2) occur in up to 20–25% of cases, with numerical changes being
of chromosomes X or Y, occurs in up 5–7% of B-ALL, exceedingly rare. Although the participating chromosome
with an increased incidence in B-other (30%) and in regions undergoing rearrangement can vary, the unifying
Down syndrome–associated ALL (>50%). The biochemical outcome of these events is induction of aberrant expression
66 Molecular Hematology

of hematopoietic transcription factor oncogenes, due to inap- Somatic mutations involving the NOTCH1 gene lead to
propriate positioning of these genes near constitutively active constitutive activation of Notch1 signaling, which is a promi-
T-cell-specific enhancers/regulatory regions within the TCR nent oncogenic pathway in T ALL. In both childhood and
beta (7q32-q36) or TCRA–TCRD (14q11) 7q32-q36 loci. adult ALL, the incidence of activating NOTCH1 mutations
These oncogenic transcription factors include basic helix- is between 50% and 70%. In addition, FBXW7 mutations,
loop-helix (bHLH) family members such as TAL1, LYL1, present in about 15% of T ALL cases, contribute to Notch
TAL2, and BHLHB1, LIM domain only (LMO) genes (LMO1 activation by impairing the proteasomal degradation of acti-
and LMO2), and HOX transcription factor oncogenes TLX1, vated Notch1 in the nucleus. The NOTCH1 gene encodes a
TLX3, and HOXA. Activation of these transcription factor type I transmembrane heterodimeric receptor, which func-
genes or transcription factor accessory proteins (LMO1 and tions as a ligand-activated transcription factor. Upon lig-
2) leads to enhanced activity of the transcriptional regulatory and binding, a series of proteolytic cleavages of the recep-
protein at the promoter or enhancer elements of a specific set tor, the last of which is catalyzed by gamma secretase,
of target genes involved in the development of hematopoietic releases intracellular Notch1, which acts downstream to
lineages. form a transcriptional activator complex for Notch1 tar-
A prototypical example of chromosomal translocation/ get genes influencing differentiation, proliferation, apoptotic
transcription factor dysregulation in T ALL is the disruption events, and in-cell fate determination/choices. Crucial effec-
of TAL1 (T-cell acute leukemia), also known as the SCL or tors of the oncogenic program downstream of NOTCH1-
TCL5 gene. The genetic events leading to TAL1 overexpres- mutated T ALL include MYC and the PI3K–AKT signal-
sion occur by a variety of mechanisms. Most commonly, ing pathway. A rare translocation t(7;9) (q34;q34.3) leads
TAL1 is activated by a cryptic interstitial deletion, TALd, to aberrant expression of a number of truncated Notch1
which results in the deletion of a ∼100 kb DNA fragment isoforms, referred to as TAN (for translocation-associated
next to the TAL1 locus and fusion with the 5′ part of the SIL Notch homolog). These truncated Notch1 isoforms (for
gene, leading to formation of the SIL-TAL1 chimeric gene. example TAN1) have a dominant oncoprotein effect, possess-
This deletion occurs in up to 25% of T ALL in children, but ing constitutive, ligand-independent activity. However, the
is much less common (∼5–10%) in adults. TAL1 overex- major cause of Notch1 dysregulation in T ALL is somatic
pression is also brought about by t(1;14)(p33;q11), which mutations of the extracellular Notch1 heterodimerization
leads to inappropriate TAL1 activation, due to its relocation domain or C terminal PEST domain, which confer consti-
close to the TCRAD locus. A recently discovered genetic tutive, ligand-independent activity to the Notch1 receptor.
mechanism delineating a novel basis for TAL1 dysregulation Discovery of molecularly altered Notch1 has led to tar-
in T ALL independent of chromosomal translocation is geted therapies that therapeutically terminate Notch1 sig-
the occurrence of somatic mutation in non-coding regions naling using γ-secretase inhibitors. Studies have shown that
downstream of TAL1, leading to the formation of an onco- combined with steroids, γ-secretase inhibitors can lead to
genic “super-enhancer” that drives transcription and over- clinically significant leukemia regressions, demonstrating the
expression of TAL1. Thus, multiple genetic defects in T ALL promising potential for Notch1 blockade therapies in T ALL.
converge on TAL-1 oncogene overexpression. A further frequent target of recurrent mutation in T ALL is
The second largest cytogenetic subgroup in T ALL, rep- CDKN2A deletions, occurring in 70% of cases. Multiple other
resenting ∼20% of cases in childhood, is characterized by components of cell cycle control are also targets of damaging
t(5;14)(q35;q32) which juxtapositions the TLX3/HOX11L2 mutations. These include chromosomal deletion of RB1 and
gene to BCL11B, leading to TLX3 transcription factor dysreg- CDKN1B genes and CCND, all seen in up to 15% of cases.
ulation via a non-TCR enhancer-driven mechanism. Occa- Thus cell cycle dysregulation plays a prominent role T ALL
sionally, chromosomal translocations in T ALL are seen that pathogenesis.
produce fusion genes encoding chimeric transcription factor The MYC oncogene is one of the most frequently acti-
oncogenes, such as t(11;19)(q23;p13) leading to MLL-ENL, vated oncogenes in T ALL. The mechanism of MYC upreg-
and also t(10;11)(p13;q14) resulting in PICALM-MLLT10 ulation is mostly post-translational, although it can result
(CALM-AF10). A common feature of leukemias harbor- from t(8;14)(q24;q11) chromosomal translocation in 1% of
ing these fusion transcription factor oncogenes is the aber- cases. Altered signal transduction programs also contribute
rant expression of developmentally important HOXA genes. to T ALL oncogenesis. Gain-of-function mutations within
Recently, a rare cryptic 9q34 deletion generating the SET- the JAK-STAT signaling axis are also a typical feature of T
NUP214 fusion gene has been described in T ALL which, ALL. Activating mutations target the IL7R gene (10%) encod-
similar to BCR-AB1, has constitutively activated tyrosine ing the IL-7 receptor, JAK1, and JAK3 (10%) genes, as well
kinase activity and is associated with sensitivity to inhibi- as more downstream effectors STAT5B (5–10%). Activating
tion with tyrosine kinase inhibitors, especially nilotinib and mutations that predict hyperactivity of the MAPK pathway
dasatinib. are also reported and result from PTEN tumor suppressor
Molecular basis of acute lymphoblastic leukemia 67

T-cell maturity

Early T precursor cell Early cortical Late cortical

Immunophenotype

sCD3 – +/– +
sCD4 – + +
sCD8 – + –
sCD1a – + –
sCD5 Weak
Myeloid marker +
Stem cell marker +
Genetic alterations

Notch1/FBXW7 + +++ ++
+/– +++ ++
CDKN2A

Myeloid tumor suppressors NUP214-ABL1 TAL1 activation


Hematopoietic genes Homebox genes PTEN deletion
Epigenetic regulators PHD finger protein 6
Cell signaling Wilms tumor 1
Protein tyrosine phosphatase
Non-receptor type 2
Fig. 5.4 Genetic abnormalities across T acute lymphoblastic leukemia (ALL) subtypes.

loss, as well as activation of HRAS, KRAS, and PTPN11 onco- genetic alterations of transcription factor oncogene activa-
genes. The finding of these molecular alterations has clearly tion. The mutations identified affect three principal path-
opened up opportunities for signal blockade therapies which, ways, namely loss-of-function mutations in transcription
although still in the early phase of pre-clinical and clinical factor–encoding genes involved in hematopoietic develop-
testing, hold tremendous promise. ment (e.g. RUNX1 IKZF1, ETV6 GATA3 EP300; ∼50%), acti-
Further insights into the molecular pathogenesis of T ALL vating mutations in factors mediating cytokine receptor and
have been garnered from recent genome-wide and transcrip- cellular signaling axis (NRAS, KRAS JAK1, JAK3, IL7R FLT3),
tomic studies. These efforts have led to the recognition of and inactivating mutations in several epigenetic modifiers
novel molecular oncogenic subtypes corresponding to devel- (PRC2, a H3K27 trimethylase, and loss of function of EZH2,
opmentally distinct stages of T ALL which are underpinned IDH1, IDH2, DNMT3A). Notably, some of the reported
by unique biology (Figure 5.4). A major T ALL entity iden- genetic alterations correspond to mutations in myeloid-
tified is the early T-cell precursor (ETP) subgroup, which specific oncogenes and tumor suppressor genes, supporting
exhibits a unique constellation of clinical, cellular, and molec- the demonstration of a myeloid gene expression program in
ular features. ETP ALL comprises 10–15% of childhood T ETP ALL. A particular prominence of aberrant activation
ALL and approximately 40–50% of adult T ALL. ETP ALL of JAK-STAT signaling in ETP tumors due to IL7 receptor
is defined by characteristic immunophenotypic features typ- and JAK1 and JAK3 as well as NRAS/KRAS pathway muta-
ical of early immature (early cortical) thymocytes: absence of tions suggests that these are rational targets for therapeutic
CD1a, CD4, and CD8 expression, weak CD5, and aberrant intervention.
expression of at least one myeloid or stem-cell marker such Crucially, genome-wide studies of T ALL have high-
as CD34 and myeloid surface antigens (CD13 and CD33). lighted the increasing importance of the non-protein cod-
Compared with more developmentally mature T ALL (e.g. ing regions in leukemogenesis. Several T ALL–specific non-
cortical T ALL), the clinical outcome of this group is poor. coding RNAs which are under the direct control of NOTCH1
Large-scale sequencing data have provided major insights have been identified which can contribute to the oncogenic
into the molecular mechanisms underlying this unique clini- state of T ALL. In addition, recurring chromosomal duplica-
cobiological subgroup. Transcriptionally, ETP T ALLs closely tions distal to MYC have also been reported. Such alterations
relate to HSCs and myeloid progenitors. The spectrum of create an enhancer for oncogenic NOTCH1, which in turns
acquired somatic alterations are distinct from classical T ALL drives MYC expression. Therefore, there is a broadening
68 Molecular Hematology

genomic basis for the core oncogenic programs implicated survival estimated at just ∼7%. The biological mechanisms
in T ALL oncogenesis, namely NOTCH1 and MYC, which giving rise to treatment resistance and eventual therapy
extends beyond just an intergenic cause. Recently, genomic failure have been greatly informed by increasing in-depth
profiling studies have identified recurrent mutations in ribo- molecular characterization of progressive ALL. In particu-
somal protein genes, in particular RPL10, RPL5, and RPL11, lar, the analysis of matched diagnosis and relapse pairs and
and CCR4–NOT transcription complex subunit 3 (CNOT3), of xenografted primary ALL tumors has defined key causal
which are thought to contribute to T ALL development by mechanisms contributing to leukemia clone evolution and
increasing basal levels of the protein synthesis that facilitates the specific genomic drivers of relapse (Figure 5.5). These
downstream malignant transformation. studies reveal that in the majority of cases, relapses are not
direct descendants of the original tumor, but are rather the
evolved products of a pre-ancestral clone that was concur-
Ph-like ALL
rently present (at a low level) in the primary tumor. This
Ph-like ALL is a recently characterized molecular subgroup is consistent with a model of intratumoral clonal diversity,
in which the underlying gene expression signature is simi- where, rather than a single genome or clone, there are multi-
lar to that of Ph-positive ALL, but where a detectable t(9;22) ple tumor genomes/clones present within a tumor.
chromosomal rearrangement or BCRABL1 oncogene is lack- Detailed evaluation of genetic patterning in sequential
ing. The incidence of Ph-like ALL increases with age, from samples together with xenografted primary ALL tumors
10% in children to up to 27% in young adults. The common reveals that instead of a linear sequence of mutational events,
features of this uniquely identified subgroup are poor clinical ALL clones appear to have evolved by a more branching
outcome and enrichment of CRLF2 rearrangement and over- subclonal diversification, resulting in a highly variegated
expression (∼50%), plus IKZF1 gene deletion (∼30%). The subclonal architecture comprising multiple variant subclones
identification of Ph-like ALL has led to fervent studies into which may share only minimal genetic relatedness. This
the underlying signaling pathway alterations mediating the branching clonal architecture, realized from the molecular
Ph-like gene expression signature. genetics of ALL, has now become a major tenet across the
Apart from identification of JAK1/2 mutations and rear- wider cancer field. This model predicts that under certain
rangement of CRLF2, both of which are highly preva- selective pressures – that is, those imposed for instance
lent in Ph-like ALL, a number of novel kinase-activating by cytotoxic therapy – variant subclones possessing a par-
mutations have been identified. These include kinase- ticular fitness advantage will survive to further diversify
activating gene fusions (∼12%) involving PDGFRB, ABL1, and/or cause relapsed disease. The precise determinants of
ABL2, and CSF1R; gene rearrangement of JAK2 (∼7%) “fitness” – that is, whether it reflects an advantage con-
and EPOR (∼3%); other mutations in JAK-STAT signal- ferred by a particular mutant genotype versus epigenetic
ing (∼12%, FLT3, IL7R, SH2B3, JAK1/3, TYK2, IL2RB, or microenvironmental interaction, or indeed whether it is
and TSLP); in addition to Ras pathway mutations (5%, entirely stochastic – continue to be debated, with evidence
KRAS, NRAS, NF1, PTPN11, and BRAF). The importance currently for all of these.
of delineating the underlying activating mutations of Ph- From the point of view of the key genomic drivers of
like ALL is underscored by the potential tractability of disease relapse, several have so far been identified. The main
these pathways to treatment intervention. For instance, Ph- driver mutations emerging at relapse include deletions of
like ALL would theoretically be amenable to inhibitors cell cycle regulator CDKN2A/B and lymphoid transcription
of the JAK pathway (CRLF2, JAK2, and EPOR) or tyro- factors ETV-6, IZKF1 ERG, SPI1, TCF4, and TCF7L2,
sine kinases such as dasatinib (ABL1, ABL2, PDGFRB, and tumor suppressors (TP53), RAS signaling, plus chromatin
CSF1R). Although large-scale data demonstrating the effec- modifiers SETD2 (histone methyltransferase) and CREBBP
tiveness of genetics-based treatment intervention in Ph- (histone acetyltransferase), the latter impairing response to
like ALL are currently lacking, the identification of Ph- glucocorticoid response genes as well as nucleoside response
like ALL offers real potential for novel treatments in this genes, NCOR1. These lesions can be subclonal at diagnosis,
highly distinct poor-risk group. Standardized diagnostics to in keeping with the evolutionary theory of cancer, where
facilitate Ph-like ALL identification will be a necessary devel- mutant-bearing fitter clones survive chemotherapeutic tar-
opment to realize the potential for optimizing therapy in geting. Potentially, the a priori knowledge of relapse-specific
these patients. mutations at diagnosis may in the future inform the applica-
tion of upfront therapies (cytokine blockade and drugs that
modulate histone marks, e.g. histone demethylases, histone
Biological basis of relapsed ALL
deacetylases, and epigenetic readers targeting bromodomain
Recurrence of ALL during or following primary ther- proteins) aimed at eliminating relapse-initiating clones
apy carries an extremely dismal prognosis, with long-term which may mitigate against ALL recurrence.
Molecular basis of acute lymphoblastic leukemia 69

Therapy

Branching
clonal
architecture

Emergence
of minor
(ancestral)
subclone

Diagnosis Relapse

Gene alterations

Major Ancestral CREBBP


Minor subclones Drug
clone clone NTC52 resistance
NCOR1
TP53
IKZF1

Fig. 5.5 Molecular mechanisms underlying acute lymphoblastic leukemia relapse.

sensitive quantitative technology for enumeration of MRD


Molecular evaluation of Minimal titers.
Residual Disease The major modality for measuring MRD in ALL is
polymerase chain reaction (PCR) analysis of the Ig/TCR
A principal feature of ALL reflecting the cellular origin of
rearrangements or chimeric fusion gene targets resulting
the disease – a B- or T-cell precursor – is rearrangement of
from chromosomal translocations (e.g. BCR-ABL), although
genes within the Ig and TCR locus. This is a normal physi-
immunological methods utilizing flow cytometry are
ological process active in developing lymphocytes and leads
increasingly being applied particularly outside Europe. Since
to antibody diversity. While Ig/TCR gene rearrangements in
Ig/TCR rearrangements occur in the majority (>90%) of pre-
ALL carry no pathophysiological consequence, their occur-
cursor B ALL (pre-B ALL) and T ALL patients, this approach
rence can be exploited for diagnostic purposes to measure
is widely applicable. Ig/TCR MRD quantification relies on
for the presence of minimal residual disease (MRD). The
the rearrangement and subsequent joining of gene compo-
concept of MRD arises from known limitations in the con-
nents within the V, D, and J regions of the immunoglobulin
ventional assessment of disease response, where despite <5%
and T-cell receptor locus. The joining of component V, D,
blasts by cytomorphology, signifying attainment of com-
and J genes results in loss of original nucleotides as well as
plete remission, there can remain up to 1010 submicroscopic
the de novo insertion of nucleotides (N region insertion)
tumor cells present, defined hereafter as the MRD. As the
at the junctional regions. The co-occurring insertion and
majority of treatment failures occur in patients achieving
loss of nucleotides at the junctional sites are unique to each
complete remission (CR), this points to the limited utility of
lymphocyte or lymphocyte clone. In the case of a leukemic
cytomorphology-defined CR for predicting disease relapse,
clone, these junctional regions serve as leukemic signatures
and highlights the potential clinical relevance of MRD.
or unique fingerprints of the patient’s leukemia cell, which
A very large body of evidence confirms the clinical impact
can be amplified by PCR using breakpoint-specific primers
of MRD in ALL and its role as a prognostic indicator of
for MRD assessment. The aim is to identify at least two MRD
relapse risk. Consequently, MRD evaluation is now routinely
PCR targets per patient. This is achieved by first determining
adopted for risk stratification in frontline protocols to delin-
the presence of clonal Ig and TCR gene rearrangements in a
eate the potential for either treatment success or treatment
diagnostic sample by heteroduplex analysis of PCR products.
failure. Measurement of MRD requires a trackable marker
This method relies on denaturation of PCR products, fol-
that is highly specific to the leukemia clone, combined with
lowed by rapid cooling to induce homoduplex (bearing PCR
70 Molecular Hematology

products with identical junctional regions) and heterodu- false positive results due to contamination. In addition, only
plex (bearing PCR products with heterogeneous junctional a minority (<40%) of patients carry these targets, thus appli-
regions) formation, corresponding to clonal and polyclonal cation is limited. Furthermore, RNA and therefore fresh sam-
cell populations, respectively. Homoduplexes and heterodu- ples (<48 hours) are required for reliable results.
plexes are separated on a polyacrylamide gel, where the
former migrate rapidly and the latter more slowly, forming a
background smear. Homoduplexes corresponding to clonal Conclusion
IG/TCR rearrangements are then sequenced to identify
junctional regions. Thereafter, PCR primers can be designed Insights into the genetic landscape of ALL have paved the
complementary to the junctional regions and tested for sensi- way for defining new and important clinical subgroups where
tivity and specificity for clone amplification in serially diluted the genetics not only inform the clinical behavior, but also
diagnostic DNA. Detection and quantitation of MRD after importantly reveal opportunities for targeted treatments.
treatment can then be performed by standard curve gener- Novel molecular subgroups in ALL defined by dysregu-
ation from the diagnostic dilution series using a real-time lated cellular signaling and epigenetic perturbation are key
fluorescence reporter PCR system. The main drawback of findings that have informed the field both scientifically and
Ig/TCR-based MRD quantification is the occurrence of con- clinically. Despite clear advances in our understanding, a
tinuing rearrangements of the original clone during the dis- notable area for further development is adult ALL, where
ease course, which can lead to false negative results. This can large-scale characterizations have yet to be performed.
be reduced by careful primer design, which avoids amplifica- Such endeavors should provide a comprehensive under-
tion of gene segments involved in continuing gene rearrange- standing of the landscapes defining a highly heterogeneous
ments and the use of at least two MRD targets per patient. disease.
In the case of chromosomal aberrations, the resultant
fusion genes can be used as reverse transcriptase PCR (RT
PCR) targets for MRD monitoring. The Philadelphia chro- Further reading
mosome, t(9;22), and resultant BCR/ABL fusion gene is by
far the most commonly assessed target for MRD monitoring Hunger, S.P. and Mullighan, C.G. (2015). Redefining ALL classifica-
in this category. In contrast to antigen gene receptor targets, tion: toward detecting high-risk ALL and implementing precision
these targets are not subject to clonal evolution and remain medicine. Blood 125: 3977–3987.
stable throughout disease course. Excellent sensitivities of Iacobucci, I. and Mullighan, C.G. (2017). Genetic basis of acute lym-
10−4 –10−6 can be reached. A disadvantage of this method is phoblastic leukemia. J. Clin. Oncol. 35 (9): 975–983.
Chapter 6 Chronic myeloid leukemia
Hagop Kantarjian, Jorge Cortes, Elias Jabbour & Susan O’Brien
Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston TX, USA

Introduction, 71 Molecular biology, 73


Epidemiology, 71 Therapy, 74
Clinical presentation and natural history, 71 Summary, 83
Prognostic models, 73 Further reading, 83

and the incidence increases with age. There are no known


Introduction causal etiologies; therefore, the disease is neither preventable
nor inherited. However, ionizing radiation is leukemogenic
Chronic myeloid leukemia (CML) is a clonal myeloprolif-
and CML has been observed in individuals exposed to the
erative neoplasm that arises from a pluripotent stem cell.
radiation of atomic bomb explosions in Japan in 1945. The
The Philadelphia (Ph) chromosome results from a recipro-
incidence of CML was 50-fold higher than in non-exposed
cal translocation between chromosomes 9 and 22, and con-
subjects and peaked approximately 10 years after the explo-
stitutes the cytogenetic hallmark of CML. It can be detected
sion, although patients younger than 15 years of age devel-
in myeloid, erythroid, megakaryocytic, B, and sometimes
oped CML earlier than those 30 years or older. In most cases
T lymphoid cells, but not in marrow fibroblasts. A criti-
of CML no antecedent radiation exposure is discernible.
cal milestone in CML research was the demonstration that
this translocation involved the ABL1 (v-abl Abelson murine
leukemia viral oncogene homolog 1) gene on chromosome
9 and the BCR (breakpoint cluster region) gene on chromo- Clinical presentation and natural
some 22, and resulted in the formation of the chimeric BCR- history
ABL1 fusion transcript that encodes the constitutively active
BCR-ABL1 tyrosine kinase. The discovery that BCR-ABL1 CML evolves typically in three phases. Approximately 90%
plays a pivotal role in the pathogenesis of CML set the stage of patients with CML are diagnosed in the chronic phase
for the development of therapeutic strategies aimed specifi- (CML-CP), characterized by high numbers of immature
cally at inhibiting this kinase and its downstream signals. In myeloid cells and mature granulocytes in the bone marrow
this chapter we summarize the current knowledge regarding and peripheral blood. Patients diagnosed in CML-CP are
the molecular biology of CML and the treatment modalities, often asymptomatic. If symptoms are present, they usually
including novel BCR-ABL1 tyrosine kinase inhibitors (TKIs). relate to the presence of splenomegaly (e.g. abdominal full-
ness, early satiety, pain). Other signs and symptoms that
may occur are anorexia, weight loss, fever, fatigue, or ane-
Epidemiology mia. Leukocytosis, with white blood cell (WBC) counts fre-
quently exceeding 100 × 109 l−1 , is frequently seen, but only
CML has an annual incidence of 1.6 per 100 000 adults, occasionally leads to signs and symptoms of hyperviscos-
and is slightly more frequent in men (male to female ratio ity (priapism, cerebrovascular accidents, dizziness, confu-
1.4 : 1). CML represents approximately 15% of all leukemias sion) or retinal hemorrhage. In CML-CP, CML cells retain
and accounts for up to 20% of all cases of adult leukemia the ability to differentiate and produce morphologically nor-
in Western societies. According to existing epidemiological mal blood elements capable of carrying out the physiologi-
databases, the median age of onset of CML is 60–65 years cal functions of normal counterparts. In about 50% of cases,
CML-CP is diagnosed after a routine blood test for unre-
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. lated reasons. The peripheral blood smear in CML-CP is
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. characterized by a left shift (i.e. increasing numbers of

71
72 Molecular Hematology

younger progenitors), although blasts typically comprise less of patients in CML-AP was one to two years before the TKI
than 5% of all WBCs. Basophilia is often present. The leuko- era, but with TKI therapy the estimated four-year survival
cyte alkaline phosphatase activity is reduced both in inten- has increased to 60–70%. In contrast to CML-AP following
sity and in the number of neutrophil band forms that stain CML-CP, de novo CML-AP has a better prognosis with front-
positively for this enzyme. The bone marrow aspirate and line TKI therapy, with an estimated eight-year survival rate
biopsy are hypercellular and demonstrate granulocytic and of 80%.
megakaryocytic hyperplasia, basophilia, and a blast per- A diagnosis of CML-BP requires the demonstration of at
centage of 5% or less. Historically, the median survival of least 30% blasts in the peripheral blood and/or the bone
patients in CML-CP was three to five years. Untreated, most marrow, or the presence of extramedullary blastic foci. The
patients progressed to a chronic myeloid leukemia blastic World Health Organization (WHO) classification has pro-
phase (CML-BP), characterized by a peripheral or bone mar- posed that the blast percentage that defines BP be changed
row blasts percentage of 30% or more, and frequently pre- from ≥30% to ≥20%, but this proposal has not been validated
ceded by an chronic myeloid leukemia accelerated phase in large studies using TKI. Immunophenotypically, CML-
(CML-AP). Before the introduction of TKI therapy, the esti- BP can display either myeloid or lymphoid features. In lym-
mated annual risk of transformation from CML-CP to CML- phoid CML-PB, blast cells often express both lymphoid and
BP was approximately 10% in the first two years after diag- myeloid cell surface markers (40–50%). The CML-BP is lym-
nosis and 15–20% thereafter. With TKI therapy, this annual phoid in 20–30% of patients, myeloid in 50%, and undiffer-
incidence was reduced to 2–3% in the first two years, and to entiated in 20%; megakaryocytic (M7) or erythroblastic (M6)
1% or less thereafter. Both CML-AP and CML-BP are char- in <5%. Cells from patients with lymphoid CML-BP exhibit
acterized by increasing arrest of maturation. high levels of the enzyme terminal deoxynucleotidyl trans-
A variety of classifications have been used to define the ferase (TdT). Most cases of lymphoid CML-BP arise from
different CML phases. One classification defines CML-AP progenitors of the B cell lineage, and express CD10, CD19,
as the presence of any of the following features: cytogenetic and CD22. T-cell CML-BP has been rarely described. The
clonal evolution, 15% or more blasts, 30% or more blasts phenotype of myeloid CML-BP cells resembles that of acute
plus promyelocytes, 20% or more basophils, or platelets myeloid leukemia cells, with blasts staining with myeloper-
lower than 100 × 109 l−1 unrelated to therapy (Table 6.1). oxidase and expressing CD13, CD33, and CD117. Clini-
Although other classification systems have been proposed cally, CML-BP is characterized by prominent hypercatabolic
for defining CML-AP and CML-BP, they have not been clin- symptoms, related to increasing tumor burden, and resis-
ically validated. Patients in CML-AP may present with fever, tance to conventional chemotherapeutic agents. The median
night sweats, weight loss, or bleeding associated with throm- survival of patients in CML-BP prior to the introduction of
bocytopenia. However, the transition from CML-CP to TKI therapy was two to six months; with TKI- based therapy
CML-AP is usually subclinical and laboratory monitoring is it improved to a median of one to three years, particularly
necessary to detect disease progression. The average survival when TKI is combined with chemotherapy.

Table 6.1 Diagnostic criteria of accelerated phase according to MD Anderson Cancer Center (MDACC), International Bone Marrow Transplant
Registry (IBMTR), and World Health Organization (WHO)

MDACC IBMTR WHO

Blasts 15–29% 10–29% 10–19%a


Blasts + promyelocytes ≥30% ≥20% NA
Basophils ≥20% ≥20%b ≥20%
Platelets (×109 l−1 ) <100 Unresponsively high or persistently low <100 or >1000 Unresponsive
Cytogenetics CE CE CE not at diagnosis
WBC NA Difficult to control, or doubling <5 days NA
Anemia NA Unresponsive NA
Splenomegaly NA Increasing NA
Other NA Chloromas, myelofibrosis Megakaryocyte proliferation, fibrosis

CE, clonal evolution; NA, not applicable; WBC, white blood cell count.
a In the WHO criteria, blastic phase is defined as a blast percentage of 20% or higher. In the MDACC and IBMTR, a definition of blastic phase

requires the presence of at least 30% blasts.


b Basophils plus eosinophils.
Chronic myeloid leukemia 73

In a study of DNA microarrays used to analyze gene


Prognostic models expression in 91 patients with CML, patients in CML-CP
or CML-BP revealed marked differences in gene expression.
The different CML phases predict for very different survival
There was a strong correlation in expression levels between
probabilities. However, the prognosis is also variable among
CML-AP and CML-BP (r = 0.81). Gene expression profiling
patients in the same phase of the disease. Several patient and
may facilitate a more comprehensive stratification of CML
disease characteristics have been found to be prognostically
that segregates genetically defined risk subgroups.
useful and have been used to generate prognostic models. In
1984, Sokal and colleagues studied 813 patients with CML
collected from six European and American series published Molecular biology
in the 1960s and 1970s. The following hazard ratio function
for death was derived from baseline patient and disease
characteristics: BCR-ABL1 oncogene
𝜆i (+) 𝜆0 (t) = Exp 0.0116 (age − 43.4) The fusion of the ABL1 and BCR genes resulting from
+ 0.0345 (spleen − 7.51) + 0.188 [(platelets∕700)2 the reciprocal translocation t(9;22)(q34;q11.2) gives rise to
the BCR-ABL1 oncogene that encodes for the constitutively
− 0.563] + 0.0887 (blasts − 2.10)
active BCR-ABL1 tyrosine kinase. Several experimental
This function segregated patients into three prognostic models, such as BCR-ABL1-expressing CD34+ cells in culture
groups with hazard ratios of <0.8, 0.8–1.2, and >1.2, and or retrovirally transduced BCR-ABL1-positive mouse cells,
median survivals of 2.5, 3.5, and 4.5 years, respectively. The have established a causal relationship between BCR-ABL1
Sokal prognostic score system was developed in the era and human leukemia. BCR-ABL1 can transform hematopoi-
before interferon alpha therapy. The Hasford score, which etic stem cells, but not committed progenitors lacking self-
takes into account age, platelet count, peripheral blast count, renewal capacity.
spleen size, eosinophils, and basophils, was developed to The breakpoint within ABL1 at 9q34 occurs in nearly all
identify risk groups in patients with CML treated with patients upstream of exon 2, with some variability in the
interferon. Both risk models have been validated in several specific site either upstream of exon Ib, downstream of exon
studies of patients treated with TKIs, but the prognostic Ia, or more frequently between the two (Figure 6.1). Break-
significance of several clinical factors has changed. Clinical points within BCR localize to three main breakpoint cluster
factors that maintain prognostic significance in the era of regions (bcr). In most patients with CML and in one-third of
TKIs include high blast (≥10–15%) and basophil (≥20%) those with Ph-positive acute lymphoblastic leukemia (ALL),
percentages, massive splenomegaly, and chromosome abnor- the breakpoint maps to the major breakpoint cluster region
malities involving 17p-/isochromosome 17, chromosome 7, (M-bcr), which spans BCR exons 12–16 (formerly called b1–
or 3q26.2. b5), giving rise to a fusion transcript with either e13a2(b2a2)
Genome-wide analyses of gene expression profiles have or e14a2(b3a2) junctions that translates into a 210-kDa
been used as prognostication tools in multiple malignancies. protein (p210BCR-ABL1 ). In two-thirds of patients with

Chromosome 22 Chromosome 9

e1 1b

e1’
e2’ 5’
m-bcr BCR 1a
e12 3’
M-bcr a2
e16 a3
5’
ABL
3’
e19
μ-bcr a11

e1a2 p190bcr-abl
e13a2
p210bcr-abl
e14a2
Fig. 6.1 The Philadelphia chromosome and e19a2 p230bcr-abl
associated BCR-ABL1 molecular abnormalities. 16
74 Molecular Hematology

Ph-positive ALL and rarely in CML, the breakpoints within Imatinib mesylate (Gleevec) is an orally bioavailable
BCR localize to an area of 54.4 kb between exons e2′ and e2, 2-phenylaminopyrimidine, relatively selective for the con-
termed the minor breakpoint cluster region (m-bcr), which stitutively active tyrosine kinase of the BCR-ABL1 fusion
translates to a 190-kDa protein (p190BCR-ABL1 ). A third protein. It also inhibits other kinases such as KIT, platelet-
breakpoint cluster region (μ-bcr) has been identified giving derived growth factor receptor (PDGFR)α and PDGFRβ,
rise to a 230-kDa fusion protein (p230BCR-ABL1 ), associated and ABL-related gene (ARG). Dasatinib (Sprycel) is a sub-
with a more indolent CML course and with a phenotype nanomolar inhibitor of BCR-ABL1 and SFKs, with potent
more akin to chronic neutrophilic leukemia (Figure 6.1). activity against KIT (IC50 13 nmol l−1 ), PDGFRβ (IC50
28 nmol l−1 ), and ephrin receptor EPHA2 (IC50 17 nmol l−1 )
tyrosine kinases. It is 350 times more potent than imatinib
BCR–ABL1 kinase-signaling pathways
in vitro. Dasatinib inhibits multiple imatinib-resistant BCR-
BCR-ABL1 signals through an intricate network of molecular ABL1 mutant isoforms, except for T315I. Nilotinib (Tasigna)
pathways that promote diminished growth factor/adhesion is a phenylaminopyrimidine derived from the crystal struc-
dependence, decrease apoptosis, and enhance proliferation. ture of imatinib in complex with ABL1 kinase. Nilotinib has
Phosphorylation of BCR Tyr177 provides a high-affinity 30–50 times improved affinity and inhibitory activity against
docking site for the SH2 domain of growth factor receptor- unmutated BCR-ABL1 and inhibits the tyrosine kinase activ-
bound protein 2 (GRB2), which in turn recruits SOS (a ity of 32 of 33 BCR-ABL1 mutants tested, the exception being
guanine-nucleotide exchanger of RAS), thus activating T315I. Bosutinib (Bosulif) is a potent SFK and ABL1 kinase
RAS and the adapter GRB2-associated binding protein 2 inhibitor with negligible activity against KIT or PDGFR,
(GAB2). BCR-ABL1-induced GAB2 phosphorylation acti- which may result in an improved toxicity profile, with less
vates the phosphatidylinositol 3-kinase (PI3K)/AKT and the myelosuppression (no inhibition of KIT) and fewer pleural
RAS/ERK pathways. BCR-ABL1 also phosphorylates the SRC effusions (no inhibition of PDGFR). Ponatinib (Iclusig) is a
family of kinases (SFKs) HCK, LYN, and FGR, which leads third-generation BCR-ABL1 multikinase inhibitor, and the
to activation of STAT5 (signal transducer and activation of only one which is effective against T315I, the gatekeeper
transcription 5). Upon dimerization, STAT5 translocates mutation. Ponatinib inhibits FLT3, FGFR, VEGFR, PDGFR,
to the nucleus and binds to cognate DNA sequences to and c-KIT. Omacetaxine mepesuccinate (Synribo) is a non-
modulate gene transcription. STAT5 also upregulates the TKI protein synthesis inhibitor approved by the FDA for the
anti-apoptotic protein BCLXL , which is repressed by the treatment of CML following failure of two TKIs. Allogeneic
tumor suppressor and negative regulator of granulocyte dif- stem cell transplantation (SCT) remains a curative modality
ferentiation ICSBP (transcription factor interferon consensus and a valid option for patients who fail TKI therapy. The
sequence binding protein). The RAC subfamily of guanosine structures of the five TKIs are shown in Figure 6.2.
triphosphatases (GTPases) RAC1, RAC2, and RAC3, as well Several general principles are emerging in CML therapy.
as the enzyme 12/15lipoxygenase (12/15-LO), have also been One is the importance of adherence to treatment, which is
identified as important elements in BCR-ABL1 downstream critical for the success of therapy. Patients who are less than
signaling. Although BCR-ABL1 signals through a multitude 90% adherent to TKI therapy as prescribed have a signifi-
of downstream components, the kinase activity of this cantly lower probability of achieving the deepest molecular
oncogenic enzyme is central to the pathogenesis of CML. responses compared with more compliant patients.
This provided the rationale to develop therapeutic strategies A second principle is the importance of daily continuous
that inhibit the kinase activity of BCR-ABL1. uninterrupted TKI therapy in both the chronic phase (single-
agent TKI; possible addition of other agents if hematologic
resistance and lack of better treatment alternatives), or in
Therapy transformation (CML-AP–TKI alone or in combination with
other standard therapies, e.g. hydroxyurea, cytarabine, azaci-
tidine, or decitabine; CML-BP–TKI combinations with acute
General
leukemia chemotherapies). TKI therapy is superior to non-
Since 2000, the treatment of CML has changed dramatically. TKIs (e.g. hydroxyurea), even when responses less than com-
Conventional chemotherapeutic agents such as busulfan plete cytogenetic response (CCyR) are achieved. In CML-BP
or hydroxycarbamide (hydroxyurea) are no longer used, the use of TKI in combination with chemotherapy is superior
except to achieve initial or transient cytoreduction. The to single-agent TKI or chemotherapy alone, even after failure
maturing experience of imatinib mesylate and other TKIs of TKI or chemotherapy. The only exceptions to not using
has brought about a change in the therapeutic algorithm TKIs in chronic or advanced CML phases are low-risk CML
for CML. Today, five TKIs have been approved by the Food as maintenance after allogeneic SCT; CML with sustained
and Drug Administration (FDA) to treat CML: imatinib, undetectable BCR-ABL1 transcript levels beyond two to
dasatinib, nilotinib, bosutinib, and ponatinib (Figure 6.2). three years, where TKI treatment discontinuation may be
Chronic myeloid leukemia 75

N CF3
H H
N N N N
CH3 O
H
N O N N N N
H N
N

N
Imatinib Nilotinib (AMN107)
N

CI CI
H
N N S O CI
HN OCH3
N N
HN
H3CO CN
N
N O N
N N
H3C
HO Dasatinib (BMS-354825) Bosutinib (SKI-606)

N CH3
N H N
N O N

N CF3
Ponatinib
Fig. 6.2 Chemical structures of imatinib and second/third-generation tyrosine kinase inhibitors used to treat chronic myeloid leukemia.

considered (treatment-free remission or molecular cures, newly diagnosed patients with CML-CP. After a follow-up of
discussed later); or persistent and intractable cytopenias eight years, the CCyR rate with imatinib was 87%, the MMR
related to TKIs not alleviated by optimization of the TKI (BCR-ABL1 transcript levels by International Standard [IS]
dose schedules. <0.1%) was 70%, and the rate of undetectable BCR-ABL1
A third general principle is to avoid discarding or changing transcripts was 40%. The estimated 10-year survival rate was
a TKI unless there is evidence of loss of CCyR (rather than 83%. The annual rate of progression from CML-CP to CML-
loss of major molecular response, MMR), or unless there is BP was 1.5–2% in the first two years and less than 0.5% there-
severe toxicity or chronic moderate toxicity affecting qual- after. In the IRIS trial, the depth of response achieved on ima-
ity of life not alleviated with optimization of the TKI dose tinib therapy was the most important prognostic factor. The
schedule (dose reduction) and symptom management. achievement of CCyR at 12 months and beyond was associ-
Because of the availability of five TKIs and the ease of chang- ated with a significant survival benefit. Achieving MMR was
ing therapy, there is a tendency to change TKIs in situa- associated with improved event-free survival (EFS) among
tions of perceived suboptimal response (e.g. increased BCR- patients who had achieved CCyR, but not with improved sur-
ABL1 transcripts to more than 0.1%, termed loss of MMR) or vival, because of the availability of effective salvage thera-
because of toxicities which could be minimized or eliminated pies if implemented optimally as soon as there is evidence
with TKI dose reductions and proper medical management. of cytogenetic relapse. More recently, achievement of unde-
This could result in discarding TKIs which could still offer tectable BCR-ABL1 transcripts sustained for at least two
clinical benefit to patients. years has offered the possibility of treatment discontinuation
(discussed later).
The DASISION study was a phase III randomized trial
Frontline CML therapy that compared dasatinib 100 mg daily with imatinib 400 mg
Based on promising preclinical data and efficacy in phase daily in 519 patients with newly diagnosed CML-CP. The
I and phase II trials in CML salvage (after interferon fail- confirmed CCyR rate at 12 months (primary study endpoint)
ure), the phase III randomized multinational IRIS (Interna- significantly favored dasatinib. In the final five-year study
tional Randomized study of Interferon-alpha plus cytarabine results, the cumulative rate of MMR by five years was 76%
versus STI571) study compared imatinib 400 mg orally daily with dasatinib and 64% with imatinib (p = 0.002); the cumu-
with interferon-alpha in combination with cytarabine in 1106 lative rate of MR4.5 (equivalent to a 4.5 log reduction in
76 Molecular Hematology

BCR-ABL1 transcripts from baseline; BCR-ABL1 transcripts Sokal group they were 94%, 97%, and 89%, respectively. In
[IS] less than 0.0032%) was 42% versus 33% (p = 0.025). The the high-risk Sokal cohort they were 89%, 91%, and 84%,
estimated five-year survival rate was 91% with dasatinib and respectively.
90% with imatinib. The rate of transformation to CML-AP A second trial from China used the same design and
or CML-BP by five years was 4.6% with dasatinib and 7.3% enrolled 267 patients. The MMR rate at 12 months was 52%
with imatinib. with nilotinib and 28% with imatinib. The rates of CCyR
In another multicenter trial, the North American Coop- (84% versus 87%) and progression-free survival (PFS; 95%
erative Groups randomized patients with newly diagnosed each) were similar at 24 months. The rates of progression to
CML-CP to dasatinib 100 mg once daily or imatinib 400 mg CML-AP/BP and of survival were similar with nilotinib and
once daily. Similar to the results of the DASISION study, imatinib.
dasatinib resulted in a higher rate of CCyR compared with While nilotinib therapy was well tolerated, there was a
imatinib (84% vs. 69%, p = 0.04). cumulative increased risk of cardiovascular events. In the
A third phase III randomized study, SPIRIT 2, also com- ENEST-nd trial, the six-year cumulative risk of a cardiovas-
pared dasatinib 100 mg daily to imatinib 400 mg daily. The cular event was 9.9% with nilotinib 300 mg twice daily, 15.9%
12-month MMR rate was 58% with dasatinib and 43% with with nilotinib 400 mg twice daily, and 2.5% with imatinib
imatinib (p < 0.001). The 12-month CCyR rates were 51% 400 mg once daily. Therefore, any potential added benefit of
versus 40% (p < 0.002). The progression rate to CML-AP was using nilotinib as initial therapy in the intermediate- and
0.5% with dasatinib and 0.7% with imatinib and to CML-BP high-risk Sokal groups should be weighed against the cumu-
1% and 1.7%, respectively. lative risk of cardiovascular complications.
The ENEST-nd was a large phase III international ran- At the MD Anderson Cancer Center (MDACC), several
domized study that compared two doses of nilotinib, 300 mg frontline studies ran sequentially and in parallel, and eval-
twice daily and 400 mg twice daily (the dose approved for uated the outcomes of patients with newly diagnosed CML
CML salvage), to imatinib 400 mg once daily. The rate of treated with imatinib 400 mg daily (n = 68), imatinib 800 mg
MMR by 12 months was 43–44% with nilotinib and 22% daily (n = 200), dasatinib 50 mg twice daily or 100 mg daily
with imatinib (p < 001). The cumulative incidence of CCyR (n = 106), and nilotinib 400 mg twice daily (n = 108). An
by 24 months was 85–87% with nilotinib and 77% with analysis of the total experience of 482 patients is summarized
imatinib (p < 0.001). With a minimum follow-up of five in Table 6.2. The experience showed similar benefits with
years, the cumulative incidence of MMR by five years was high-dose imatinib and second-generation TKIs in relation
77% with both nilotinib schedules and 60% with imatinib to early surrogate endpoints of long-term outcome (CCyR,
(p < 0.0001%). The incidence of BCR-ABL1 transcripts MMR, MR4.5). However overall survival was not signifi-
≤0.0032% IS (MR4.5) by six years were 55–56% with nilo- cantly different by treatment strategy. The table shows the
tinib and 33% with imatinib (p < 0.0001%). The incidence cumulative incidence of outcomes (usually reported in the lit-
of transformation to CML-AP/BP was 3.9% with nilotinib erature as outcome by a certain time), as well as the incidence
300 mg twice daily, 2.1% with nilotinib 400 mg twice daily, of outcomes at particular times, a more accurate reflection of
versus 7.4% with imatinib (p = 0.06 and 0.003, respectively). the benefit of TKIs.
However the definition of CML-AP used the pre-TKI
criteria, which are now known to be less relevant. The inci-
Selection of frontline TKI therapy
dence of CML-BP (a more relevant clinical endpoint) with
nilotinib versus imatinib was not reported. The estimated Current CML treatment guidelines offer imatinib 400 mg
five-year EFS rates were 95%, 97%, and 93%, respectively. daily, dasatinib 100 mg daily, or nilotinib 300 mg twice daily
When considering Sokal-risk category, nilotinib was more (on an empty stomach) as frontline treatment options in
effective in reducing the transformation rate in higher-risk CML. With the availability of generic imatinib, the choice of
Sokal disease. The rates of transformations were 1%, 1%, frontline TKI therapy may depend on several pre-treatment
and 0% among patients with low Sokal risk treated with considerations, as well as the endpoints of therapy. In 2019,
nilotinib 300 mg orally twice daily, nilotinib 400 mg orally the annual cost of generic imatinib is about $4000 for two
twice daily, or imatinib 400 mg orally once daily. The rates (among nine) least costly generics in the United States,
were 2%, 1%, and 10% among patients with intermediate- $3000–8000 in Canada, and $400 in India. Survival is sim-
risk Sokal, and 9%, 5%, and 11% among patients with ilar with all three TKIs, therefore generic imatinib may
high-risk Sokal. The estimated five-year survival rates were be the preferred treatment of choice for many patients. If
94% with nilotinib 300 mg twice daily, 96% with nilotinib second-generation TKIs are chosen over generic imatinib
400 mg twice daily, and 92% with imatinib once daily. The (estimated annual cost about 30% of patented imatinib), the
survival rates in patients with low-risk Sokal scores were incremental cost-effectiveness ratio (ICER = annual ratio of
97%, 99%, and 100% respectively. In intermediate-risk additional cost over incremental benefit) is about $800 000/
Chronic myeloid leukemia 77

Table 6.2 The MD Anderson Cancer Center long-term experience of chronic myeloid leukemia frontline therapy with tyrosine kinase inhibitors

% Response parameter Imatinib 400 mg Imatinib 800 mg Nilotinib Dasatinib

%Cumulative CCyR by 5 years 85 90 99 97


%CCyR at 5 years 87 90 93 96
% MMR at 5 years 76 86 91 90
% Cumulative MR4.5 by 5 years 56 74 80 78
% MR4.5 at 5 years 57 74 71 71

Compared with imatinib 400 mg daily, the 5-year event-free survival rate was significantly higher with imatinib 800 mg (p = 0.029), dasatinib
(p = 0.003), and nilotinib (p = 0.031). However, there were no differences in the rates of 5-year failure-free survival (p = 0.32, p = 0.075,
p = 0.332), transformation-free survival (p = 0.053, p = 0.038, p = 0.493), or overall survival (p = 0.563, p = 0.162, p = 0.981).
CCyR, complete cytogenetic response; MMR, major molecular response; MR4.5, see text.

quality-adjusted life-years (QALY), significantly higher than a previous history of cardiovascular event, pancreatitis, or
the accepted ICER of $50 000/QALY. severe diabetes.
Beyond survival, several factors may be considered in
deciding on TKI therapy: (i) the incidence of durable
Discontinuation of TKI therapy and
undetectable BCR-ABL1 transcripts for longer than two to
treatment-free remissions
three years (which may offer the option of treatment discon-
tinuation and treatment-free remissions); (ii) the CML Sokal Several studies have shown that TKI discontinuation in
risk; (iii) the out-of-pocket expenses to the patient; (iv) the patients with CML-CP on TKI therapy who have sustained
patient’s age; and (v) the pre-treatment medical comorbidi- undetectable BCR-ABL1 transcripts for at least two years
ties (e.g. hypertension, diabetes, chronic lung disease, pan- results in treatment-free remissions in 40–60%. The inci-
creatitis) and the associated risks for serious adverse events dence of MR4.5 (two years or more) on frontline TKI ther-
(such as arterio-thrombotic events, pleural effusions, pancre- apy is about 42% with second-generation TKIs, and 26%
atitis, pulmonary hypertension). with imatinib therapy (this increases to 40% with sequential
The incidence of treatment-free remissions, discussed second-generation TKIs). Therefore, the rates of treatment-
later in the chapter, is estimated at 20–25% with second- free remissions range from 13% to 21%. Treatment discon-
generation TKIs and 13–15% with imatinib. This therapeutic tinuation, when attempted in community practice, should
endpoint may be more important among young patients (e.g. adhere to strict criteria to avoid harm to patients. Patient
age ≤50–55 years or younger) in whom treatment discontin- must have low- or intermediate-risk disease (not high-risk),
uation after 5–10 years is relevant (to avoid long-term treat- a documented history of only chronic phase disease (no evi-
ment costs and side effects) compared with older patients dence of transformation), and an optimal response to front-
(e.g. age ≥65–70 years or older). Second-generation TKIs line TKI therapy. The combined duration of TKI treatments
may potentially improve outcome in patients with high-risk should be at least five or more years and undetectable BCR-
Sokal CML (10–20% of CML) and might be chosen for these ABL1 transcripts should be documented for at least two
patients as frontline therapy. to three years, with assessments every six months during
Out-of-pocket expenses are an increasing burden on that period. Patients must have quantifiable BCR-ABL1 tran-
patients, at least in the US. With 25% out-of-pocket expenses scripts (e13a2, e14a2) and comply with frequent monitoring
and a second-generation TKI annual cost of $150 000+, out- by polymerase chain reaction (PCR) every one to two months
of-pocket expenses could exceed $25 000 annually, close in the first two years after TKI discontinuation, and every
to half the annual income of an average family. This may three to six months thereafter. A recent analysis indicated
decrease patient compliance, which would be associated that, at current prices, second-generation TKIs do not offer
with a worse outcome. In such instances, generic imatinib a good treatment value for the endpoint of treatment-free
may reduce costs, improve compliance, and even provide remission.
better results (better compliance) than second-generation
TKIs.
Treatment endpoints in CML and other
Some relative contraindications or caution should be
endpoints of evolving “treatment value”
exerted based on prior medical history. Dasatinib should be
avoided in patients with a history of chronic lung disease or The achievement of CCyR (Ph-positive metaphases 0%;
pleural effusions. Nilotinib should be avoided in patients with BCR-ABL1 transcripts [IS] ≤1%) by/at 12 months and later
78 Molecular Hematology

on TKI therapy is associated with a significant survival that, among patients achieving CCyR, survival was similar
benefit compared with achievement of lesser degrees of whether they also achieved MMR or lesser degrees of molec-
response. Therefore, achieving CCyR is the primary end- ular response.
point of TKI therapy. Achievement of MMR (BCR-ABL1 Early response after three months of therapy was also
≤0.1% IS) is associated with modest improvements in EFS, reported to be an important treatment landmark associated
but not with a survival benefit. Achievement of sustained with differences in survival (discussed later). This implied
undetectable BCR-ABL1 transcripts offers the possibility of that at least 30% of patients on frontline imatinib would be
treatment discontinuation. Lack of achievement of MMR or switched to second-generation TKIs after only three months
of MR4.5 should not be interpreted as a need to change TKI of imatinib. As discussed shortly, the potential change of ther-
therapy or to consider allogeneic SCT. Other measurements apy after six months for lack of early response is more appro-
of outcome include EFS, transformation-free survival (TFS), priate and would apply to only 8% of patients.
and PFS. Event is defined differently in various studies and More recently there has been an emphasis on TKI treat-
could include instances of treatment discontinuation that ment discontinuation and treatment-free survival, an impor-
have no relevance to survival outcome (e.g. discontinuation tant therapeutic endpoint. However, the cost-effectiveness
for toxicity or non-compliance; death from causes unrelated analysis of such an approach, assuming treatment-free remis-
to CML, etc.). Transformation encompasses CML-AP, and sions of 20–25% with second-generation TKIs and 13–15%
many of the AP criteria have lost their prognostic relevance with imatinib, did not show a good treatment value for
on TKI therapy. second-generation TKIs at their current prices. A more rea-
At MDACC, the major treatment milestones are at six sonable annual price for second-generation TKIs is needed
and twelve months. Patients with BCR-ABL1 transcripts (IS) (perhaps $10 000–20 000 per year rather than $150 000+) to
>10% at six months, or not achieving CCyR with BCR-ABL1 offer a good treatment value for this endpoint.
transcripts (IS) ≤1% at one year, or with loss of response at
any later time, are offered a change of TKI therapy if possi-
Monitoring and determining treatment
ble. Patients on second TKIs with BCR-ABL1 transcripts (IS)
failure
>10% at six months should be observed closely for signs of
progression, but cannot be changed to “better” TKIs in gen- At baseline, a marrow examination is needed to establish the
eral, and have a better survival with continued TKI therapy diagnosis, determine the percentage of blasts and basophils
compared with proceeding to allogeneic SCT (five-year sur- (10% of patients may have higher percentages in the marrow
vival rates 85–90% vs. 65–70%). TKI therapy is not changed vs. blood that could change their CML phase), and perform
in patients in CCyR but not in MMR. cytogenetic analysis (confirm the presence of the Philadel-
All key randomized trials of imatinib compared with phia chromosome and assess for additional chromosomal
second-generation TKIs as initial therapy for CML-CP have abnormalities, or clonal evolution). A pre-treatment fluores-
shown a significant benefit of surrogate response endpoints, cence in-situ hybridization (FISH) analysis (assess Ph pos-
but similar survivals with second-generation TKIs versus itivity) and PCR (measure BCR-ABL1 transcripts) help to
imatinib. Generic imatinib is now available at a low annual avoid later false negative tests if alternative transcripts were
cost. Using second-generation TKIs as frontline therapy over present initially (2–3% of cases).
imatinib results in an ICER value of $800 000/QALY, an The current recommendation that follow-up marrow stud-
excessive “treatment value.” ies be done at 3, 6, and 12 months after starting therapy may
There has been a recent emphasis on treatment endpoints not be necessary. An alternative is to evaluate response by
other than survival. The three key ones include (i) achieve- FISH and PCR on peripheral blood. If a patient is responding
ment of MMR; (ii) achievement of early treatment response optimally, and the FISH study is negative at 6 or 12 months
(BCR-ABL1 transcripts <10% by three months of TKI ther- and/or BCR-ABL1 transcripts (IS) <1%, it may be reasonable
apy); and (iii) achievement of durable undetectable BCR- to omit marrow examinations, as the patient is likely to be in
ABL1 transcripts for at least two years, as a condition for CCyR.
treatment-free remission. These endpoints have been empha- For patients in CCyR receiving TKI therapy, periodic
sized in independent continuing medical education meetings molecular monitoring using FISH and PCR every six months
and symposia, and by CML experts. Therefore, it is important is acceptable. FISH and PCR are used as complementary
to explore in depth the supportive data. diagnostic tests to assess for response concordance (to detect
Achievement of “deeper molecular responses” (MMR) was possible false positive/high or negative/low values generated
suggested as an important treatment endpoint that may cor- by either assay). For patients in CCyR and MMR, a PCR
relate with improved survival. This implied that since MMR test every six months is sufficient to monitor response. For
rates are higher with second-generation TKI compared with patients with BCR-ABL1 transcripts between 0.1% and 1%
imatinib, they should be used as a better frontline therapy. and negative FISH analysis, more frequent monitoring every
However, several long-term follow-up studies have shown three months may be of value.
Chronic myeloid leukemia 79

Early molecular response has been shown to have strong conformation of the kinase to which imatinib binds, the cat-
prognostic value. In a study from the UK, patients with newly alytic domain, and the gatekeeper T315 residue. The T315I
diagnosed CML on imatinib therapy who achieved BCR- mutation causes steric hindrance to TKI binding and con-
ABL1 transcript levels >10% at three months had an eight- fers resistance to all TKIs, except ponatinib. Some patients
year survival rate of 54%, compared with 93% for those with with CML failing sequential TKI therapies carry more than
lower levels. However, patients did not have the option of one mutation within the same BCR-ABL1 molecule (“com-
an early change to second-generation TKIs. At MDACC, a pound” mutations or “polymutants”), which are associated
similar analysis showed 10-year survival rates of 94% ver- with increased oncogenic potency compared with each single
sus 98%; these patients accessed second-generation TKIs at mutation.
the first evidence of loss of CCyR. In the ENEST-nd and BCR-ABL1 mutations do not explain all cases of clinical
DASISION trials, the differences in survival rates were 86– resistance to TKI therapy. Other possible mechanisms of
87% versus 97%. These data suggested the need for a change resistance include BCR-ABL1-independent mechanisms
from imatinib to second-generation TKIs at three months (most common in patients with primary resistance); ima-
among patients with BCR-ABL1 transcripts (IS) >10%. This tinib plasma levels; excessive binding of imatinib to the
would require a change of therapy in 30% of patients on plasma protein α1 -acid glycoprotein 1 (AGP1); overexpres-
frontline imatinib therapy, and may not be appropriate con- sion of ABCB1 (MDR-1) transmembrane protein, which
sidering that it is a one-time test which has a high rate of regulates imatinib efflux from the cell; polymorphisms of the
variability around the PCR 10% value, and may be influ- human organic cation transporter (hOCT1), which regulates
enced by patient compliance and treatment interruptions imatinib influx; clonal evolution; SFK or BCR-ABL1 over-
over a short period of three months. Analysis from Aus- expression; and intrinsic low TKI sensitivity of quiescent
tralia and MDACC showed that an evaluation at six months CML stem cells (Lin− CD34+ BCRABL1-positive cells). Each
might be more appropriate. In these analyses, patients with of the mechanisms of resistance suggests different possible
BCR-ABL1 transcripts (IS) >10% at six months had a worse therapeutic interventions. The long-term updates of TKIs
long-term outcome (although still approximately 60% were in CML salvage are showing continued favorable results in
alive and free of events long term), regardless of the BCR- CML-CP–resistant disease. In a phase III study investigating
ABL1 transcript levels at three months. This requires a different dose schedules of dasatinib in 670 patients with
change of therapy in only 8% of patients, the ones who really CML-CP following imatinib resistance or intolerance, the
need it. cumulative CCyR rate was 50%, the MMR rate 42%, and the
A similar situation in a patient on second-generation TKIs estimated seven-year survival rate 65%. In a phase II trial of
(dasatinib, nilotinib) does not necessarily imply considera- bosutinib 500 mg orally daily in 288 patients with CML after
tion of a change of TKIs (no better one available) or of allo- imatinib resistance or intolerance, the CCyR was 48%, MMR
geneic SCT (five-year survival is better with continuation of 35%, and the estimated two-year survival rate 91%. The four-
TKI versus implementing allogeneic SCT). year follow-up study of nilotinib 400 mg twice daily in 321
patients with CML after imatinib failure resulted in a CCyR
rate of 45% and an estimated four-year survival rate of 78%.
Treatment of CML following frontline
In the phase II trial of ponatinib 45 mg orally daily in 267
failure
patients with heavily pretreated CML-CP, the CCyR rate was
Imatinib resistance can be categorized as primary (failure to 54%, the MMR rate 39%, and the four-year rates of PFS and
respond to TKI from the onset of therapy) and secondary survival were 60% and 81%, respectively. Among the subset
(after initial achievement of response). Mutations within the of 64 patients with a T315I mutation, the CCyR rate was 70%,
kinase domain of BCR-ABL1 are a frequent mechanism of MMR rate 58%, and estimated four-year survival rate 77%.
resistance to TKIs. The frequency of BCR-ABL1 mutations In a pivotal study of omacetaxine, 122 patients with CML-
in CML-CP after cytogenetic relapse on imatinib is 30% and CP in chronic (n = 81) or CML-AP (n = 41) who had received
after hematological relapse 50%. Mutation detection is rare two or more prior TKIs were treated. In the chronic phase,
in patients responding to frontline therapy (less than 5%). the major cytogenetic response rate was 20% (complete 10%),
Mutation rates are higher (50–90%) in patients experienc- the median duration of response was 17.7 months, and the
ing CML transformation. More than 100 different BCR-ABL1 median survival was 34 months.
point mutations encoding for single amino acid substitutions
have been reported, conferring different degrees of imatinib
Drug dose schedules
resistance (Table 6.3). The most frequently reported muta-
tions map to the P-loop region of the kinase domain, which All five BCR-ABL1 TKIs are given orally. Imatinib 400 mg
serves as a docking site for phosphate moieties of adenosine daily, nilotinib 300 mg twice daily (on an empty stomach),
triphosphate (ATP). Mutations also frequently map to the and dasatinib 100 mg daily are FDA-approved frontline ther-
activation loop, which impair the achievement of the inactive apies for the treatment of CML. They are also approved for
80 Molecular Hematology

Table 6.3 Relative activity profile of BCR-ABL1 tyrosine kinase inhibitors in imatinib-resistant mutants

IC50 -fold increase relative to wild type (W = 1)

Mutation Imatinib Bosutinib Dasatinib Nilotinib Ponatinib

M244V 0.9 0.9 2 1.2 3.2


L248R 14.6 22.9 12.5 30.2 6.2
L248V 3.5 3.5 5.1 2.8 3.4
G250E 6.9 4.3 4.4 4.6 6
Q252H 1.4 0.8 3.1 2.6 6.1
Y253F 3.6 1 1.6 3.2 3.7
Y253H 8.7 0.6 2.6 36.8 2.6
E255K 6 9.5 5.6 6.7 8.7
E255V 17 5.5 3.4 10.3 12.9
V299L 1.5 26.1 8.7 1.3 0.6
T315A 1.7 6 58.9 2.7 0.4
T315I 17.5 45.4 75 39.4 3
T315V 12.2 29.3 738.8 57 2.1
F317L 2.6 2.4 4.5 2.2 0.7
F317R 2.3 33.5 114.8 2.3 4.9
F317V 0.4 11.5 21.3 0.5 2.3
M351T 1.8 0.7 0.9 0.4 1.2
F359I 6 2.9 3 16.3 2.9
F359V 2.9 0.9 1.5 5.2 4.4
H396R 3.9 0.8 1.6 3.1 5.9
F486S 8.1 2.3 3 1.9 2.1

White = very sensitive (IC50-fold increase <2); yellow = moderately sensitive (IC50 2.1–4); orange = resistant (IC50 4.1–10); red = highly resistant
(IC50 > 10).
Source: Adapted from Redaelli S., Mologni L., Rostagno R., et al. (2012). Three novel patient-derived BCR/ABL mutants show different sensitivity
to second and third generation tyrosine kinase inhibitors. Am. J. Hematol. 87: E125–E128.

the treatment of CML following failure of frontline therapy. lower dose schedule of 20–70 mg daily, which is still effective
Nilotinib 400 mg orally daily is the approved dose for CML and significantly less toxic. Nilotinib can also be used at
after failure of frontline therapy. Dasatinib 100 mg daily is a lower dose schedule of even 150–200 mg orally twice
the approved dose for CML after failure of frontline therapy; daily. Bosutinib can be used in a dose schedule range of
dasatinib 140 mg daily is indicated for patients with CML 300–500 mg daily. While the approved ponatinib dose is
in transformation. In addition, bosutinib 500 mg daily and 45 mg daily, dose schedules of 15–30 mg daily appear to be
ponatinib 45 mg daily are approved for the treatment of CML effective and significantly less toxic.
following failure of frontline therapy. Omacetaxine is given
at the dose of 1.25 mg m−2 twice daily subcutaneously for
Choice of TKIs
14 days during induction, and for 7 days every 1–2 months
during maintenance. This schedule may be too myelosup- The FDA-approved indications for the five TKIs are sum-
pressive. Omacetaxine for five to seven days during induc- marized in Table 6.4. The choice of TKI depends on several
tion, and for two to five days every one to two months during factors in both the frontline and salvage settings. In frontline
maintenance (alone or in combination with a TKI), may be therapy, TKI selection may be based on the cost of the
less myelosuppressive and equally effective. TKI (generic imatinib vs. more expensive patented second-
As long-term experience is gained with the TKIs, it is generation TKIs), CML Sokal risk (higher vs. lower), patient
clear that drug dose modifications can eliminate or reduce age and treatment aim (survival vs. treatment-free remis-
significant toxicities and allow continuation of therapy. For sion), and prior medical history (e.g. diabetes, hypertension,
example, in a case of pleural effusions, dasatinib can be cardiac or other vasospastic or vaso-occlusive problem,
held, steroids initiated for a short period (e.g. prednisone pancreatitis, pulmonary problems including chronic
40–60 mg daily × 3–5 days), and dasatinib restarted at a obstructive pulmonary disease or pleural effusions). This
Chronic myeloid leukemia 81

Table 6.4 Food and Drug Administration-approved indications of tyrosine kinase inhibitors (TKIs) and omacetaxine in chronic myeloid leukemia

Frontline Salvage Accelerated Blastic

Imatinib 400 mg QD 400 mg QD 600 mg QD 600 mg QD


(IFN failure) (IFN failure) (IFN failure)
Dasatinib 100 mg QD 100 mg QD 140 mg QD 140 mg QD
R1 R1 R1
Nilotinib 300 mg BID 400 mg BID 400 mg BID —
R1 R1
Bosutinib — 500 mg QD 500 mg QD 500 mg QD
R2 R2 R2
Ponatinib — 45 mg QD 45 m QD 45 mg QD
R2 R2 R2
Omacetaxine — 1.25 mg m−2 SQx14d 1.25 mg m−2 SQx14d —
Q28d (induction), Q28d (induction),
then x7d Q28d then x7d Q28d
R3 R3

QD, daily; BID, twice daily; IFN, interferon alpha.


R1 = resistance or intolerance to prior therapies including imatinib.
R2 = resistance or intolerance to prior TKIs.
R3 = resistance or intolerance to two prior TKIs.

was detailed earlier. In CML salvage therapy, two additional then be restarted on lower doses of dasatinib 20–70 mg
factors are considered: prior TKI exposures, and whether daily. Myelosuppression (particularly thrombocytopenia) is
there are any BCR-ABL1-guiding mutations. more common with dasatinib compared with imatinib. Pul-
monary hypertension can occur occasionally and manifests
clinically as shortness of breath, normal chest radiograph,
Side effects of TKIs and right-side heart failure. It is reversible with dasatinib
discontinuation.
Imatinib is associated with mild to moderate toxicities,
Notable side effects with nilotinib include headache and
including fatigue, nausea, vomiting, diarrhea, skin rashes,
skin rashes (common in 20–30%, but mild to moderate; alle-
muscle cramps and bone aches, periorbital or leg edema,
viated by dose reduction), self-limited elevation of indirect
and weight gain. Serious but uncommon toxicities (<2%)
bilirubin (10%), elevations of blood sugar (10–20%), and
include a decreased glomerular filtration rate, which is
rarely pancreatitis (1–2%). The six-year cumulative incidence
reversible with treatment discontinuation or interruption
of vaso-spastic/vaso-occlusive disease is 10% with nilotinib
and resuming the drug at a lower dose. Hepatic or car-
300 mg twice daily and 15% with nilotinib 400 mg twice daily.
diopulmonary adverse events are rare, as well as peripheral
The most significant toxicities of bosutinib include
neuropathies and central neurological toxicities, which are
myelosuppression (30%), liver function abnormalities (17%,
at times misdiagnosed as dementia or Alzheimer’s disease.
mostly mild to moderate; severe in 7%), and gastrointestinal
These reverse slowly after TKI discontinuation. Drug-
problems (nausea 45%; diarrhea 70–80% – early and self-
related myelosuppression occurs in 10–30% of patients and
limited, but requiring dose reductions to 300–400 mg daily,
is managed with brief treatment interruptions and/or dose
severe in 8%).
modifications, or with growth factors (e.g. erythropoietin
Significant ponatinib toxicities include pancreatitis in
for anemia, filgrastim for neutropenia).
10–15%, severe thrombocytopenia in 35%, severe skin rashes
On average, the chronic mild to moderate toxicities affect-
in 5–10%, vaso-spastic and vaso-occlusive disease in 15–20%,
ing quality of life are lower with second-generation TKIs
and hypertension in 35% (severe in 10–15%).
compared with imatinib (fatigue, fluid retention and perior-
bital edema, muscle cramps and bone aches, weight gain).
Dasatinib is associated with pleural effusions in up to 30%
Other therapies
of patients, which may be severe, requiring thoracentesis in
2–3%. Most resolve with dasatinib treatment interruptions, Interferon alpha was developed as a CML therapy in
diuretics, and short courses of corticosteroids. Patients can the 1980s and became a standard of care in 1990–2000.
82 Molecular Hematology

Interferon therapy induced survival benefits, but was toxic. degree of matching, CML phase, and others. It is most
A CCyR was obtained in 10–35% of patients and was effective in CML-CP. Among patients in first CML-CP who
associated with a 10-year survival rate of 78%, establishing undergo matched related sibling transplant, the 20-year sur-
the concept of achievement of CCyR to improve survival. vival rate is 40–50%. The International Bone Marrow Trans-
Of interest is that 30% of patients in CCyR had undetectable plant Registry (IBMTR) data of >6000 patients showed a 5-
BCR-ABL1 transcripts and most had not relapsed after more year survival rate of 60% in CML-CP/sibling donor SCT, but
than 10 years of follow-up, establishing the uncommon but a 20-year survival rate of 40–45%. About 10% of patients
possibly curative effect of interferon alpha in CML. Today, still die in years 5–20 from transplant-related complications
interferon alpha is rarely used as a standard of care; it can rather than CML relapse. Chronic morbidities following
be used in women with CML who become pregnant and SCT include graft-versus-host disease (GVHD), second neo-
cannot receive TKIs, in combination with TKIs to overcome plasm, cataracts, infertility, hip necrosis, and GVHD-caused
CML resistance, in investigational TKI combination studies organ damage (pulmonary, hepatic) or immune-mediated
to improve therapeutic results, or in investigational studies complications. Disease-free survival (DFS) rates are 60–80%
aimed at eradication of CML minimal residual disease. among patients <30–40 years of age, and 30–40% in patients
Hydroxyurea, a ribonucleotide reductase inhibitor, is a >50 years. DFS rates are 30–50% in patients with CML-AP
well-tolerated oral agent that controls CML counts rapidly. and 5–30% in patients with CML-BP. Patients with clonal
Rare side effects include nausea, rashes, mouth ulcers, and evolution as the only CML-AP criterion have DFS rates of
hand or leg ulcers. The dose range is 1–10 g daily × 1–5 days 60%. Patients receiving SCT in second chronic-phase CML
depending on the degree of leukocytosis. The dose is adjusted have DFS rates of 40%. Non-myeloablative preparative regi-
to keep the WBC count around 3–10 × 109 cells l−1 . Hydrox- mens are safer in older patients and have reduced transplant-
yurea can be used for initial cytoreduction, or as part of a associated complications and mortality. Haplo-identical and
combination approach with TKIs. It should not be used alone cord blood transplants are showing promising results.
as a treatment in CML. The role and timing of allogeneic SCT in patients with
Busulfan (1,4-dimethane-sulfonyl-oxybutane) is the first CML are changing based on the maturing experiences
alkylating agent to demonstrate activity in CML. It is asso- with TKIs and SCT, as well as the cost of therapies and
ciated with significant toxicities: severe prolonged myelo- regional considerations. Patients who present with or evolve
suppression, myelofibrosis (“spent phase of CML”), and into CML-AP/BP should receive TKI combinations with
Addison-like syndrome. Today, the role of busulfan is only chemotherapy to reduce the CML burden and proceed to
as part of conditioning regimens in allogeneic SCT. allogeneic SCT. The exception may be de novo CML-AP,
which may still respond well to long-term TKI therapy,
particularly if a CCyR is achieved early. Among patients
Pregnancy, CML, and TKIs
whose CML manifests resistance to TKI therapy in CML-
If a pregnant woman is diagnosed as having CML, she can CP, the treatment choice depends on whether mutations
be managed with leukapheresis as indicated during the first are detected, and whether there is clonal evolution. Patients
trimester of pregnancy, and with hydroxyurea therapy sub- with T315I mutation should receive ponatinib, and proceed
sequently until delivery, then with more definitive therapy. to allogeneic SCT (pending longer-term results with pona-
Some experts recommend interferon alpha therapy during tinib). Patients with particular mutations may be treated with
pregnancy, but this may have adverse effects on the fetus. If a mutation-selective TKI (nilotinib, dasatinib, bosutinib, or
woman with CML becomes pregnant while on TKI therapy, ponatinib), based on the mutation sensitivity to particular
the drug should be discontinued. Among 125 infants born to TKIs. Patients with clonal evolution or with mutations unre-
women with CML on imatinib (drug discontinued after preg- sponsive to TKIs have short response durations; allogeneic
nancy documented), 12 had birth defects, including 3 with a SCT should be considered as a definitive therapy, particularly
syndrome of ocular, renal, and skeletal abnormalities. Chil- in patients who do not achieve a cytogenetic response after
dren of women whose male partners have CML and are on 6–12 months of TKI therapy. Older patients (≥65–70 years)
imatinib have all been normal. The incidence of fetal mal- and patients who lack a donor may forgo the option of cura-
formations after dasatinib exposure in a similar scenario is tive allogeneic SCT in favor of years of good disease control
higher, about 10%. in CML-CP with TKI-based therapies.
Most relapses after SCT occur in the first three to
five years. Patients usually exhibit molecular relapse prior
Allogeneic stem cell transplantation
to cytogenetic-hematologic relapse. CML relapse usually
Allogeneic SCT is curative, but is associated with signifi- responds to TKI therapies (response rates 50–60% if a molec-
cant morbidities and with a one-year mortality of 5–30% ular or cytogenetic relapse in CML-CP). Other options
depending on several factors: patient age, source of cells, include donor lymphocyte infusions, interferon alpha, or a
Chronic myeloid leukemia 83

second SCT. Donor lymphocyte infusions may exacerbate hypomethylating agents (azacitidine, decitabine), hydrox-
GVHD (which can be fatal) and cause severe myelosuppres- yurea, cytarabine or AML-type therapy, and considered for
sion (20%). allogeneic SCT, depending on the clinical condition and
prognosis.
Atypical CML is a diagnosis of exclusion, an entity that
Therapy for CML in accelerated and blastic does not meet the WHO criteria for BCR-ABL1-positive
phases CML, primary myelofibrosis, polycythemia vera, or essential
thrombocythemia. There are SETBP1 mutations in 15–30%.
All five TKIs and omacetaxine have shown efficacy in CML-
Patients with chronic neutrophilic leukemia have signif-
AP/BP. The single-agent TKI activity is modest in CML-AP
icant neutrophilia without dysplasia. There are mutations of
and even lower in CML-BP. In CML-AP, TKIs are associated
CSF3R in 90% and mutations of SETBP1 in 50%. Patients with
with CCyR rates of 20–30% and four-year survival rates of
chronic neutrophilic leukemia and CSF3R mutations may
40–50%, depending on the CML-AP definition and extent
respond to ruxolitinib (JAK2 inhibitor) therapy (response
of prior TKI therapies. The second-generation TKIs produce
rate 50–55%).
better results than imatinib, either alone or in combination.
MDS/MPN-RS-T manifests as anemia with 15% or more
All TKIs have shown fewer responses, which are also less
ring sideroblasts in the erythroid precursors and thrombocy-
deep and of shorter duration in CML-BP, and combination
tosis ≥450 × 109 . There are mutations of SF3B1 in 50–70%,
modalities should be pursued. These include acute myeloid
which is associated with a better prognosis (median survival
leukemia regimens (e.g. idarubicin + cytarabine) plus TKIs
6.9 years versus 3.3 years with wild-type SF3B1).
or azacitidine/decitabine plus a TKI (if intensive chemother-
Patients with MDS-MPD and translocations involving
apy is not indicated) in myeloid-undifferentiated CML-BP,
5q33 – t(5q33; other); PDGFR-beta rearrangement – may
and acute lymphoid leukemia regimens plus TKIs (e.g. hyper-
respond to imatinib therapy.
CVAD + dasatinib or ponatinib) in lymphoid CML-BP. Cen-
tral nervous system prophylaxis should also be administered
in lymphoid CML-BP, since 30% of patients may develop cen-
Summary
tral nervous system disease. In all cases, patients should be
referred for allogeneic SCT as soon as possible. They may also
BCR-ABL1 TKIs have significantly improved prognosis in
benefit from post-SCT TKI maintenance.
CML. Today, TKI frontline therapy in CML-CP is associ-
ated with an estimated 10-year survival rate of 85%. With
TKI therapy, good patient compliance, and optimal monitor-
Philadelphia chromosome-negative CML
ing, the relative survival in patients with CML is similar to
Some patients present with a morphological picture of CML that of normal individuals. Frontline therapy with imatinib
without detectable Ph-positive disease by cytogenetic anal- and new-generation TKIs results in the durable disappear-
ysis. This condition encompasses diverse entities. A third ance of BCR-ABL1 transcripts, which may lend itself to possi-
of patients with typical CML morphology have the BCR- ble treatment discontinuation and treatment-free remission.
ABL1 molecular abnormality detected by FISH or PCR (Ph- Combinations of TKIs with other active agents that target
negative, BCR-ABL1-positive CML) and have similar clinical the downstream signaling events or are directed against the
features, response to TKI therapy, and prognosis to patients “dormant” CML stem cells may increase the proportion of
with Ph-positive CML. patients with durable complete molecular responses, and the
Patients who have atypical CML morphologies (ane- rate of treatment-free remissions.
mia, thrombocytopenia, absent basophils or eosinophils,
presence of monocytosis) and who do not have the
BCR-ABL1 molecular abnormality (Ph-negative, BCR- Further reading
ABL1-negative) are categorized by the WHO classification
into (i) chronic myelomonocytic leukemia (CMML); (ii) Molecular biology of CML
atypical CML, BCR/ABL1-negative; (iii) myelodysplastic/
Daley, G.Q., Van Etten, R.A., and Baltimore, D. (1990). Induction of
myeloproliferative neoplasm, unclassifiable (MDS-MPN-
chronic myelogenous leukemia in mice by the P210bcr/abl gene of
U); and (iv) myelodysplastic/myeloproliferative neoplasm the Philadelphia chromosome. Science 247: 824–830.
with ringed sideroblasts and marked thrombocytosis Deininger, M.W., Goldman, J.M., and Melo, J.V. (2000). The molecular
(MDS-MPD-RS-T). biology of chronic myeloid leukemia. Blood 96: 3343–3356.
Patients with CMML are diagnosed based on periph- Groffen, J., Stephenson, J.R., Heisterkamp, N. et al. (1984). Philadelphia
eral monocytosis greater than 109 l−1 and ≥10% periph- chromosomal breakpoints are clustered within a limited region, bcr,
eral monocytes. Patients with CMML can be treated with on chromosome 22. Cell 36: 93–99.
84 Molecular Hematology

Heisterkamp, N., Jenster, G., ten Hoeve, J. et al. (1990). Acute leukemia Weisdorf, D.J., Anasetti, C., Antin, J.H. et al. (2002). Allogeneic bone
in bcr/abl transgenic mice. Nature 344: 251–253. marrow transplantation for chronic myelogenous leukemia: compar-
Melo, J.V. (1996). The diversity of BCR-ABL fusion proteins and their ative analysis of unrelated versus matched sibling donor transplanta-
relationship to leukemia phenotype. Blood 88: 2375–2384. tion. Blood 99: 1971–1977.

Prognostic factors and models Imatinib

Cortes, J., Talpaz, M., O’Brien, S. et al. (2006). Staging of chronic myeloid Björkholm, M., Ohm, L., Eloranta, S. et al. (2011). Success story of tar-
leukemia in the imatinib era. Cancer 106: 1306–1315. geted therapy in chronic myeloid leukemia: a population-based study
Gratwohl, A., Hermans, J., Goldman, J.M. et al. (1998). Risk assessment of patients diagnosed in Sweden from 1973 to 2008. J. Clin. Oncol. 29:
for patients with chronic myeloid leukemia before allogeneic blood 2514–2520.
or marrow transplantation. Chronic Leukemia Working Party of the Cortes, J. and Kantarjian, H. (2012). How I treat newly diagnosed
European Group for Blood and Marrow Transplantation. Lancet 352: chronic phase CML. Blood 120: 1390–1397.
1087–1092. Druker, B.J., Guilhot, F., O’Brien, S.G. et al. (2006). Five-year follow-up
Hasford, J., Pfirrmann, M., Hehlmann, R. et al. (1998). A new prognostic of patients receiving imatinib for chronic myeloid leukemia. N. Engl.
score for survival of patients with chronic myeloid leukemia treated J. Med. 355: 2408–2417.
with interferon alfa. Writing Committee for the Collaborative CML Druker, B.J., Sawyers, C.L., Kantarjian, H. et al. (2001). Activity of a spe-
Prognostic Factors Project Group. J. Natl. Cancer Inst. 90: 850–858. cific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of
Kantarjian, H.M., Dixon, D., Keating, M.J. et al. (1988). Characteristics chronic myeloid leukemia and acute lymphoblastic leukemia with the
of accelerated disease in chronic myelogenous leukemia. Cancer 61: Philadelphia chromosome. N. Engl. J. Med. 344: 1038–1042.
1441–1446. Druker, B.J., Tamura, S., Buchdunger, E. et al. (1996). Effects of a selec-
Sokal, J.E., Cox, E.B., Baccarani, M. et al. (1984). Prognostic discrimi- tive inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl pos-
nation in “good-risk” chronic granulocytic leukemia. Blood 63: 789– itive cells. Nat. Med. 2: 561–566.
799. Kantarjian, H., O’Brien, S., Jabbour, E. et al. (2012). Improved survival in
chronic myeloid leukemia since the introduction of imatinib therapy:
a single-institution historical experience. Blood 119: 1981.
Interferon alpha Kantarjian, H., Sawyers, C., Hochhaus, A. et al. (2002). Hematologic and
cytogenetic responses to imatinib mesylate in chronic myelogenous
Guilhot, F., Chastang, C., Michallet, M. et al. (1997). Interferon alfa-2b leukemia. N. Engl. J. Med. 346: 645–652.
combined with cytarabine versus interferon alone in chronic myel- O’Brien, S.G., Guilhot, F., Larson, R.A. et al. (2003). Imatinib compared
ogenous leukemia. N. Engl. J. Med. 337: 223–229. with interferon and low-dose cytarabine for newly diagnosed chron-
Kantarjian, H.M., Smith, T.L., O’Brien, S. et al. (1995). Prolonged sur- icphase chronic myeloid leukemia. N. Engl. J. Med. 348: 994–1004.
vival in chronic myelogenous leukemia after cytogenetic response to
interferon-alpha therapy. Ann. Intern. Med. 122: 254–261.
Talpaz, M., Kantarjian, H.M., McCredie, K. et al. (1986). Hematologic
Second- and third-generation TKIs
remission and cytogenetic improvement induced by recombinant Cortes, J.E., Kim, D.W., Pinilla-Ibarz, J. et al. (2013). A phase 2 trial of
human interferon alpha A in chronic myelogenous leukemia. N. Engl. ponatinib in Philadelphia chromosome-positive leukemias. N. Engl.
J. Med. 314: 1065–1069. J. Med. 369: 1783–1796.
Cortes, J., Nicolini, F., Wetzler, M. et al. (2013). Subcutaneous omacetax-
Allogeneic stem cell transplantation ine mepesuccinate in patients with chronic-phase chronic myeloid
leukemia previously treated with 2 or more tyrosine kinase inhibitors
Barrett, J. and Ito, S. (2015). The role of stem cell transplantation for including Imatinib. Clin. Lymphoma Myeloma Leuk. 13 (5): 584–591.
chronic myelogenous leukemia in the 21st century. Blood 125: 3230– Cortes, J.E., Saglio, G., Kantarjian, H.M. et al. (2016). Final 5-year
3235. study results of DASISION. The dasatinib versus imatinib study
Crawley, C., Szydlo, R., Lalancette, M. et al. (2005). Outcomes of in treatment-naı̈ve chronic myeloid leukemia patients trial. J. Clin.
reduced intensity transplantation for chronic myeloid leukemia: an Oncol. 34: 2333–2340.
analysis of prognostic factors from the Chronic Leukemia Working Gambacorti-Passerini, C., Brummendorf, T., Kim, D. et al. (2014). Bosu-
Party of the EBMT. Blood 106: 2969–2976. tinib efficacy and safety in chronic phase chronic myeloid leukemia
Gratwohl, A., Hermans, J., Niederwieser, D. et al. (1993). Bone mar- after imatinib resistance or intolerance: minimum 24-month follow-
row transplantation for chronic myeloid leukemia: long-term results. up. Am. J. Hematol. 89: 732–742.
Chronic Leukemia Working Party of the European Group for Bone Hochhaus, A., Saglio, G., Hughes, T.P. et al. (2016). Long-term
Marrow Transplantation. Bone Marrow Transplant. 12: 509–516. benefits and risks of frontline nilotinib vs imatinib for chronic
Hansen, J.A., Gooley, T.A., Martin, P.J. et al. (1998). Bone marrow myeloid leukemia in chronic phase: 5-year update of the randomized
transplants from unrelated donors for patients with chronic myeloid ENESTnd trial. Leukemia 30: 1044–1054.
leukemia. N. Engl. J. Med. 338: 962–968. Kantarjian, H.M., Giles, F.J., Bhalla, K.N. et al. (2011). Nilotinib is effec-
Oehler, V.G., Gooley, T., Snyder, D.S. et al. (2007). The effects of ima- tive in patients with chronic myeloid leukemia in chronic phase after
tinib mesylate treatment before allogeneic transplantation for chronic imatinib resistance or intolerance: 24-month follow-up results. Blood
myeloid leukemia. Blood 109: 1782–1789. 117: 1141–1145.
Chronic myeloid leukemia 85

Kantarjian, H., Giles, F., Wunderle, L. et al. (2006). Nilotinib in imatinib Mechanisms of resistance to TKIs
resistant CML and Philadelphia chromosome-positive ALL. N. Engl.
J. Med. 354: 2542–2551. Gorre, M.E., Mohammed, M., Ellwood, K. et al. (2001). Clinical resis-
Shah, N.P., Rousselot, P., Schiffer, C. et al. (2016). Dasatinib in imatinib- tance to STI-571 cancer therapy caused by BCR-ABL gene mutation
resistant or-intolerant chronic-phase, chronic myeloid leukemia or amplification. Science 293: 876–880.
patients: 7-year follow-up of study CA180-034. Am. J. Hematol. (9): Shah, N.P., Nicoll, J.M., Nagar, B. et al. (2002). Multiple BCR-ABL kinase
869–874. domain mutations confer polyclonal resistance to the tyrosine kinase
Talpaz, M., Shah, N.P., Kantarjian, H. et al. (2006). Dasatinib in ima- inhibitor imatinib (STI571) in chronic phase and blast crisis chronic
tinib resistant Philadelphia chromosome-positive leukemias. N. Engl. myeloid leukemia. Cancer Cell 2: 117–125.
J. Med. 354: 2531–2541. Shah, N.P., Skaggs, B.J., Branford, S. et al. (2007). Sequential ABL
Weisberg, E., Manley, P.W., Breitenstein, W. et al. (2005). Characteriza- kinase inhibitor therapy selects for compound drug-resistant BCR-
tion of AMN107, a selective inhibitor of native and mutant Bcr-Abl. ABL mutations with altered oncogenic potency. J. Clin. Invest. 117:
Cancer Cell 7: 129–141. 2562–2569.
Chapter 7 Myeloproliferative neoplasms
Jyoti Nangalia1,2,3 , Anthony J. Bench4 , Anthony R. Green3,5 & Anna L.
Godfrey6
1 Wellcome Sanger Institute, Hinxton, UK
2
Department of Haematology, University of Cambridge, Cambridge, UK
3
Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK
4 Laboratory Medicine, NHS Lothian, Edinburgh, UK
5
Department of Haematology, Cambridge Institute for Medical Research, Cambridge, UK
6 Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK

Introduction, 87 Chronic neutrophilic leukemia and chronic eosinophilic leukemia, 95


The JAK2V617F mutation in Philadelphia-negative MPNs, 87 Integration of molecular analysis into diagnostic and management
Other JAK2 mutations in MPNs, 91 pathways, 95
MPL mutations in ET and PMF, 92 Conclusions and future directions, 96
CALR mutations in ET and PMF, 93 Further reading, 97
Other somatic mutations in PV, ET, and PMF, 94

diagnostic tool in PV, were an early demonstration of the


Introduction hypersensitivity of hematopoietic progenitors from MPN
patients to cytokines.
The classical Philadelphia-negative myeloproliferative neo-
The biological behavior of PV, ET, and PMF, and the
plasms (MPNs) comprise polycythemia vera (PV), essential
clinical similarities between them, became clearer with
thrombocythemia (ET), and primary myelofibrosis (PMF).
the identification in 2005 of the JAK2V617F mutation in
These disorders share features including altered stem cell
the majority of PV patients and approximately half of
behavior, overproduction of myeloid lineages, propensities to
patients with ET and PMF. Subsequently, the findings of
thrombosis, hemorrhage and splenomegaly, and, in a minor-
mutations in exon 12 of the JAK2 gene in JAK2V617F-
ity of patients, transformation to acute myeloid leukemia
negative PV, and of CALR and MPL mutations in patients
(AML). They were originally grouped together by Dameshek
with JAK2V617F-negative ET and PMF, have further clar-
along with chronic myeloid leukemia (CML) in 1951. The
ified the biology of MPNs and provided major tools for
World Health Organization (WHO) categorizes the MPNs
molecular diagnosis that are now integrated into interna-
along with CML and rarer clonal disorders, including chronic
tional diagnostic criteria. However, it has also become clear
neutrophilic leukemia (CNL). Clonal mast cell diseases are
not only that the pathogenesis of these disorders depends on
now classified separately and will not be discussed in this
these “phenotypic” mutations that are relatively specific to
chapter.
MPNs, but that a range of other genetic lesions may also have
Evidence that the MPNs represent clonal disorders was
roles in initiation of disease and in driving transformation.
originally derived from the presence of recurrent abnor-
mal karyotypes in approximately one-third of PV and PMF
patients, together with the finding that X-chromosome inac-
The JAK2V617F mutation in
tivation patterns in the majority of PV and PMF patients, and
Philadelphia-negative MPNs
a proportion of ET patients, demonstrate clonally derived
blood cells. Biological studies of hematopoietic progenitor
The JAK2V617F mutation is predominantly found in
cells by Prchal and Axelrad in 1974 showed that in semisolid
patients with PV (>95% of patients), ET, and PMF (50–
media, erythroid progenitors from the bone marrow of PV
60%). Among other myeloid neoplasms it can be found
patients could be grown without the addition of erythro-
in association with SF3B1 mutations in about half of
poietin (EPO), in contrast to those of normal individuals
patients with the uncommon overlap entity myelodysplas-
which required the presence of EPO. These “endogenous ery-
tic/myeloproliferative neoplasm with ring sideroblasts and
throid colonies” (EECs), which were subsequently used as a
thrombocytosis (MDS/MPN-RS-T), but at much lower fre-
quencies in AML and other chronic myeloid disorders.
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. JAK2 is crucial for effective signaling through a number
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. of cytokine receptors, including those for erythropoietin

87
88 Molecular Hematology

(a)
pseudokinase kinase

JAK2 FERM SH2 JH2 JH1

617
Val Phe

(b)

EPO

P P

V617F
V617F
JAK2

JAK2

JAK2

JAK2

JAK2

JAK2
EPOR

EPOR

EPOR

EPOR

EPOR

EPOR
P P
P P P P
P P P P
P P P P

No signal JAK/STAT JAK/STAT


PI3K/AKT PI3K/AKT
ERK/MAPK ERK/MAPK
Fig. 7.1 The JAK2V617F mutation. (a) JAK2 protein showing the valine to phenylalanine substitution at codon 617. (b) Role of JAK2 in cytokine
signaling. The cytokine receptor EPOR binds JAK2 as a homodimer (left). On ligand binding, a conformational change within EPOR brings the two
JAK2 molecules into close proximity, initiating a cascade of phosphorylation events and activation of downstream pathways (middle). The JAK2
V617F molecule is constitutively active, leading to erythropoietin (EPO)-independent activation of signaling pathways (right).

(EPOR) and thrombopoietin (TPOR). Homozygous knock- stabilizing the activation loop within JH1 in the inactive
out of murine Jak2 results in embryonic lethality due to the form.
complete absence of erythropoiesis. In the unstimulated
state, EPOR forms a transmembrane homodimer, the cyto-
Biochemical and cellular effects of the
plasmic domain of which is bound to two JAK2 molecules
JAK2V617F mutation
(Figure 7.1). Ligand binding to the extracellular domain
results in transphosphorylation and activation of the JAK2 The valine at codon 617 of JAK2 lies at an evolutionarily
molecules (Figure 7.1). Activated JAK2 phosphorylates conserved position within the JH2 domain (Figure 7.1) and
tyrosine residues within EPOR which, in turn, promotes is important for the interaction between the JH1 and JH2
recruitment and JAK2-mediated phosphorylation of other domains. The V617F mutation results in the substitution
JAK2 substrates such as STAT5 and phosphatidylinositol of valine by a bulkier phenylalanine residue, which appears
3-kinase (PI3K). Activation of various intracellular signaling to interfere with JH2-mediated auto-inhibition by a num-
pathways ensues (Figure 7.1). JAK2 possesses two JAK ber of mechanisms and leads to constitutive activation of
homology (JH) domains (Figure 7.1). The JH1 domain pos- the tyrosine kinase (Figure 7.1). The downstream effects of
sesses tyrosine kinase activity, while the JH2 or pseudokinase JAK2 V617F may depend on its association with homod-
domain is homologous to JH1 and predominantly plays an imeric cytokine receptors such as EPOR and TPOR, but also
auto-inhibitory role, by directly interacting with and with heterodimeric receptors (e.g. interferon-γ receptors)
Myeloproliferative neoplasms 89

and with related tyrosine kinases (e.g. JAK1 and TYK2). It is typically higher in granulocytes and erythroblasts com-
also has a scaffold role in some cytokine receptor complexes pared to more primitive (CD34+ ) cell fractions. Consistent
and is required for effective trafficking of receptors to the with this, knock-in mouse models carrying JAK2V617F show
cell surface. The cellular effects of the mutation will there- increased numbers of lineage-restricted hematopoietic pro-
fore depend on the other signaling molecules expressed by a genitors, including those of the megakaryocytic/erythroid
specific cell type. and granulocyte/macrophage lineages.
The biochemical effects of the JAK2V617F mutation reflect Conversely, the effects of JAK2V617F on hematopoietic
both canonical and non-canonical targets. A variety of stem cells (HSCs) have been much less clear. Xenotransplan-
experimental systems have confirmed that the mutation is tation experiments, in which CD34+ cells from PV patients
associated with activation of canonical pathways including were transplanted into non-obese diabetic/severe combined
JAK/STAT, PI3K/AKT, and MAPK/ERK signaling. Constitu- immunodeficiency (NOD/SCID) mice, found that the pro-
tive activation of STAT5 in erythroid progenitors can induce portion of JAK2V617F-positive cells was reduced in reconsti-
the formation of EECs and, consistent with this, STAT5 tuted marrow compared to the original donor cells, arguing
activation appears particularly important in the erythrocy- against a selective advantage for the mutant cells. Studies of
tosis associated with JAK2-mutant MPNs: impairment of the number and function of HSCs in JAK2V617F knock-in
STAT5 signaling can abrogate JAK2V617F-induced cytokine mice have yielded different conclusions between the differ-
independence in cell lines, as well as the erythrocytosis ent models, perhaps due to variation in whether the human
observed in JAK2V617F-driven mouse models of MPNs. or protein mutant gene was used and how this was intro-
Altered expression of many downstream genes has been duced into the Jak2 gene locus. Studies of MPN patients are
implicated in disease pathogenesis. For example, the anti- further complicated by the potential presence of additional
apoptotic protein Bcl-xL is required for the growth of ery- mutations in JAK2-mutant or wild-type cells, and uncertainty
throid progenitors and is upregulated in PV erythroid pro- about the processes that determine HSC number and regen-
genitors. Interestingly, JAK2 V617F may not only upregulate eration, particularly during aging. At present there is no con-
Bcl-xL through STAT5-driven transcription, but may also clusive evidence that JAK2V617F confers an advantage on
inhibit the normal DNA damage response by interfering with human HSCs, and additional genetic factors, either consti-
changes in intracellular pH and Bcl-xL deamidation. Many tutional or acquired, may well be required to promote the
other apoptotic mediators have also been implicated down- expansion and persistence of JAK2-mutant clones in MPNs.
stream of JAK2 V617F, including the anti-apoptotic protein
FLIPshort, ID1, Pim1/2 kinases, the BH3-only proteins Bad
and Bim, and the Bcl2-like protein Mcl-1, downstream of the Factors influencing disease presentation in
STAT3, STAT5, PI3K/AKT, and MAPK/ERK pathways. JAK2V617F-positive MPNs
Non-canonical effects of JAK2 were identified following Following the identification of the JAK2V617F mutation,
the observation that JAK2 was not confined to the cyto- many studies have attempted to determine the additional fac-
plasm but was also present in the nucleus. The kinase has tors that influence which specific MPN phenotype develops
histone phosphorylation activity and induces a specific chro- in a patient carrying the JAK2V617F mutation. For exam-
matin modification, phosphorylation of H3Y41, which alters ple, patients with JAK2V617F-positive ET, as compared to
gene expression through impaired binding of the protein JAK2V617F-negative ET, show certain clinical characteris-
HP1α. Moreover, JAK2 V617F can also phosphorylate and tics reminiscent of PV, including higher hemoglobin levels
alter the activity of a histone arginine methyltransferase, and neutrophil counts, lower platelet counts, greater bone
PRMT5. It therefore seems that canonical activation of gene marrow cellularity, erythropoiesis, and granulopoiesis, lower
transcription (e.g. downstream of activated STATs) and per- serum ferritin and mean corpuscular volume. These find-
turbations of chromatin modification may be complemen- ings suggested that JAK2-mutated ET could represent a
tary mechanisms through which mutant JAK2 alters cellular “milder” form of PV and prompted further studies investi-
signaling. gating which factors might lead to a phenotype of ET or PV
At the cellular level, studies of JAK2V617F in different in the context of JAK2V617F. Homozygosity for JAK2V617F
hematopoietic cell compartments suggested that while there is acquired by many patients, but is more frequent in PV
may be preferential expansion of the mutant cells, this may than ET, with homozygous-mutant precursors detectable in
occur predominantly in the later stages of erythroid and approximately 80% and 50% of patients, respectively. More-
granulocytic differentiation. For example, when quantitative over, the homozygous-mutant subclones are much larger in
assays are used to estimate the relative proportions of cells patients with PV than those in ET. These findings suggested
that are mutant and wild type for JAK2 in the bone mar- that homozygosity for the mutation might have a role in
row of patients with PV and ET, the V617F allele burden driving a PV phenotype. Quantitative polymerase chain
90 Molecular Hematology

(a)

Wild-type allele

al F+
7
P1 P2

or 61
V
er
K2

m
at
JA
W

N
P3
G
Mutant allele
Control
P1 P2
Mutant specific

T P3

(b)
Wild-type allele R

R Q Q JA
JAK2
0.1
V617F+

G G 0.01

∆Rm
Mutant allele Normal
R Q 0.001
R Q
0.0001
T T 20 22 24 26 28 30 32 34 36 38 40
Cycle

Fig. 7.2 Detection of the JAK2V617F mutation. (a) Allele-specific polymerase chain reaction (PCR). Amplification with primers P1 and P3 yields
a product of 364 bp (control), whereas amplification with primers P1 and P2 yields a 203 bp PCR product from the mutant allele only. (b) Real-time
PCR using a dual-labeled probe specific for the mutant allele. Amplification from the wild-type allele results in displacement but not destruction of
the probe, resulting in no release of fluorescence. In contrast, amplification from the mutant allele results in Taq-dependent destruction of the
probe, releasing the reporter (R) whose fluorescence can then be detected.

reaction (PCR) assays can be used in patient samples not mild thrombocytosis with a heterozygous mutation, whereas
only to detect the presence of the V617F mutation, but homozygosity was associated with marked erythrocytosis,
also to accurately quantify the proportion of mutated alleles confirming that the acquisition of JAK2V617F homozygos-
(Figure 7.2). Among patients with PV, higher V617F allele ity can be associated with PV-like, rather than ET-like, dis-
burdens (measured in peripheral blood granulocytes) were ease. However, other knock-in models have shown significant
associated with more extreme PV features, such as higher erythrocytosis even with a heterozygous mutation, and the
hemoglobin levels and white cell counts and lower platelet explanations for these discrepant phenotypes are not entirely
counts at diagnosis, again consistent with the concept that clear.
JAK2V617F homozygosity might be important in the PV Early mouse models also demonstrated that in the con-
phenotype. text of different genetic backgrounds, the JAK2V617F muta-
Mouse models have also been informative in investigat- tion may be associated with variable hematological features.
ing the factors determining phenotype in the context of This supported the concept that constitutional genetic differ-
JAK2V617F. These models have been generated by a num- ences may affect the presentation of human MPNs, which has
ber of approaches: the earliest models used transduction of been further confirmed by genome-wide association stud-
murine bone marrow with retroviruses carrying JAK2V617F; ies (GWAS) examining the prevalence of single-nucleotide
several transgenic models were then developed; and most polymorphisms (SNPs) in MPN patients. A specific constitu-
recently knock-in mouse models have been generated with tional haplotype at the JAK2 locus (“46/1”) is strongly associ-
mutant JAK2 in a heterozygous or homozygous state. In ated with the development of JAK2V617F-positive MPNs, as
general these mice have developed convincing myeloprolif- well as with JAK2 exon 12-mutated and MPL-mutated MPNs.
erative phenotypes with features including erythrocytosis, It remains uncertain whether the mechanism for this reflects
leukocytosis, splenomegaly, and increased marrow fibrosis, an increased risk of developing a JAK2 mutation, or whether
but the specific features have varied substantially between the presence of the 46/1 haplotype facilitates the development
models. With respect to PV and ET, a knock-in model car- of clinical disease following the acquisition of JAK2V617F.
rying a mutant human JAK2 gene showed a phenotype of Additional GWAS have identified other SNPs that are
Myeloproliferative neoplasms 91

associated with MPNs, including at the TERT, TET2, and (a)


SH2B3 gene loci. Interestingly, an SNP at the HBS1L/MYB
locus was not only associated with MPNs, but within JAK2-
mutated patients was associated with an increased risk of
ET and not PV, further supporting the concept that consti-
tutional genetic differences may influence not only disease
development but also phenotype.
Additional mutations may coexist with JAK2V617F and
there is evidence that interactions between mutations may
also influence phenotype. For example, mutations in the
gene for the transcription factor NF-E2 may coexist with the
JAK2V617F mutation in PV patients, and may cooperate in
driving erythrocytosis and other myeloproliferative features.
TET2 mutations were first identified in patients with very
large JAK2V617F-heterozygous clones, in keeping with the
concept that additional mutations could confer an advantage
that promotes the growth of JAK2-mutant progenitors. In
such patients with multiple driver mutations, the precise
order in which these are acquired leads to the establishment
of a particular clonal substructure within HSC and progeni-
tor compartments, and may therefore determine the relative
expansion of the myeloid lineages. Additional mutations are (b)
likely to be particularly important in JAK2V617F-positive
PMF and in patients who transform from PV or ET to 0 Normal
myelofibrosis. Both primary and post-PV/ET myelofibrosis K539L
are characterized by a greater average number of somatic
F537-K539delinsL
mutations than PV or ET, and the spectrum of mutations –5
found in these disorders shows significant overlap with N542-E543del
AML and other chronic myeloid malignancies, as discussed E543-D544del
shortly. F537-I546dupF547L
–10
HRM Difference Plot

Other JAK2 mutations in MPNs 70 75 80


Fig. 7.3 Types and detection of JAK2 exon 12 mutations. (a)
Approximately 2% of well-characterized PV patients do not Three main types of mutation can be described: lysine to leucine
carry a JAK2V617F mutation. In 2007 it was shown that substitution at codon 539 (shaded); deletion of glutamic acid at codon
many of these patients carry a mutation within JAK2 exon 543 (shaded); duplication affecting codons 537–546 (underlined). (b)
Detection of JAK2 exon 12 mutations by high resolution melting
12. Over 20 mutations have now been described, clustering
analysis.
within a small region spanning codons 537–547 and most
Source: Frequency of mutations taken from Passamonti F., Elena C.,
often affecting Leu539 or Glu543 (Figure 7.3). This SH2– Schnittger S., et al. (2011). Molecular and clinical features of the
JH2 linker region lies just upstream of the JH2 pseudoki- myeloproliferative neoplasm associated with JAK2 exon 12 mutations.
nase domain (Figure 7.3) and is known to undergo a con- Blood 117: 2813–6.
formational change in the presence of the V617F mutation.
Like JAK2V617F, JAK2 exon 12 mutations lead to cytokine
independence and increased phosphorylation of JAK2 sub-
strates. Murine models carrying exon 12 mutations show ele- the incidence of thrombosis, myelofibrosis, acute leukemia,
vated hematocrit, but normal or mildly elevated neutrophil and death appears to be similar between PV patients with
and platelet counts. Similarly, patients with PV who carry a JAK2V617F and exon 12 mutations. JAK2 exon 12 mutations
JAK2 exon 12 mutation often display an isolated erythrocy- have not been detected within JAK2V617F-negative ET or
tosis, together with isolated bone marrow erythroid hyper- PMF patients and are therefore specific to PV. Hence, muta-
plasia without granulocytic or megakaryocytic expansion, in tions within JAK2 exon 12 lead to increased kinase activ-
contrast to JAK2V617F-positive patients who characteristi- ity that drives the HSC toward erythropoiesis, but not other
cally show hyperplasia of all three myeloid lineages. However, hematopoietic lineages.
92 Molecular Hematology

A number of germline JAK2 mutations have been reported 10 have also been identified in JAK2/CALR-unmutated ET,
in familial MPNs, including V617I and other mutations and recently some of these have been confirmed to have
within and outside of the pseudokinase domain. These muta- functional effects. Many patients acquire homozygous MPL
tions have most often been associated with hereditary throm- mutations, especially those with PMF and/or with a W515K
bocytosis and have shown to have weaker functional effects mutation, while others have compound mutations (on the
on JAK2 compared to JAK2V617F. More recently, non- same or different chromosomes) or a coexistent JAK2V617F
canonical, activating JAK2 mutations were also identified in mutation.
a minority of individuals with JAK2/MPL/CALR-unmutated MPL mutations can confer cytokine independence in cell
ET and in a rare case of isolated erythrocytosis, and again lines, together with activation of targets downstream of the
these were mostly confirmed to be germline. thrombopoietin receptor, including JAK2, STAT3, STAT5,
Akt, and ERK. Introduction of the W515K and W515A
mutations into murine bone marrow was associated with
MPL mutations in ET and PMF a myeloproliferative phenotype of thrombocytosis, leukocy-
tosis, extramedullary hematopoiesis, megakaryocytic hyper-
Mutations in the gene for the thrombopoietin receptor, MPL, plasia, and fibrosis of the bone marrow and spleen. These
were first reported in 2006 and have since been found in mice did not develop erythrocytosis, in keeping with a selec-
4–9% of patients with PMF and 1–11% of those with ET. tive effect on megakaryocytic precursors and with the obser-
The commonest acquired mutations are W515L and W515K vation that these mutations are not associated with PV in
within exon 10 of the protein, but other mutations are found humans. Patients with MPL-mutated PMF are more likely to
at the W515 residue and elsewhere (Figure 7.4). The S505N be female, older, and have more severe anemia than those
variant was originally described in hereditary thrombocyto- with MPL-unmutated PMF, while those with MPL-mutated
sis, but has also been found as an acquired mutation in ET ET tend to present with lower hemoglobin levels, higher
and PMF. While a number of other variants cause heredi- platelet counts, and lower bone marrow cellularity compared
tary thrombocytosis, additional rare mutations outside exon to those with JAK2V617F-positive ET.

(a) S505

Extracellular TM JM Cytoplasmic

W515
(b)

2 HRM Difference Plot

1 Normal

0 W515L
–1
W515A
–2
S505N
–3
W515K
–4
W515R
–5

–6 Fig. 7.4 Detection of MPL exon 10 mutations.


–7 (a) The S505 and W515 amino acid residues lie
within the transmembrane domain (TM) and
–8 juxtamembrane domain (JM), respectively.
–9 (b) Detection of MPL exon 10 mutations by
79.5 80.0 80.5 81.0 81.5 82.0 82.5 83.0 83.5 84.0 84.5 85.0 85.5 86.0 86.5 87.0 87.5 88.0 high-resolution melting analysis.
Myeloproliferative neoplasms 93

with the remainder of mutations being alternative insertions


CALR mutations in ET and PMF or deletions affecting the same DNA region (Figure 7.5).
Mutations in CALR result in a 1 bp shift of the codon-reading
Despite the landmark discoveries of mutations in JAK2 and
frame of the DNA, resulting in a neomorphic CALR pro-
MPL in MPNs, just under half of patients with ET and
tein with a novel carboxyl (C)-terminus, which suggests a
PMF still lacked a phenotypic driver mutation that accounted
gain of function. This is in contrast to other genes affected by
for their disease. In 2013, next-generation sequencing tech-
insertion or deletion mutations, where there is often prema-
niques enabled the identification of mutations in the gene
ture truncation of the protein sequence and resultant loss of
calreticulin in around 80% of such JAK2/MPL-unmutated
function.
patients. CALR mutations are insertions or deletions affect-
Unlike JAK2 and MPL, which are proteins involved in
ing the terminal exon 9 of the gene, and 85% of mutations are
cytokine receptor signaling in hematopoietic cells, CALR
either a 52-bp deletion (type 1; c.1099_1150del; L367fs*46) or
resides within the lumen of the endoplasmic reticulum (ER),
a 5-bp insertion (type 2; c.1154_1155insTTGTC; K385fs*47),

c.1154_1155insTTGTC
(a) p.K385fs*47 (Type 2)

N domain P domain C domain KDEL

c.1092_1143del
p.L367fs*46 (Type 1)

(b)
Normal

50 75 100 125 150 175 200


Size (nt)

c.1092_1143del
(Type 1)

50 75 100 125 150 175 200

c.1154_1155insTTGTC
(Type 2)
Wild-type allele

Fig. 7.5 Detection of CALR exon 9 mutations.


(a) CALR mutations lie within the C-terminal Mutant allele
portion of the CALR protein. (b) Detection of CALR
exon 9 mutations by fluorescent polymerase chain
reaction followed by high-resolution capillary
electrophoresis. 50 75 100 125 150 175 200
94 Molecular Hematology

where it functions as a chaperone protein that ensures the However, 10–15% of patients with ET and PMF still lack a
proper folding of newly synthesized glycoproteins prior to recurrent genetic driver of their disease. It is possible that
their exportation to the cell surface or extracellular excretion these patients are a heterogeneous group, with a subset still
via the Golgi apparatus. Mutant CALR loses its C-terminal– carrying as yet unidentified somatic drivers of their disease,
KDEL amino acid ER retention motif, as well as numer- and others harboring either reactive myeloproliferation or
ous calcium binding sites. Studies exploring the mechanism other myeloid disorders.
by which mutant CALR results in myeloproliferation have
revealed a novel paradigm for MPN pathogenesis. Mutant
CALR is able to transform Ba/F3 cells expressing the throm- Other somatic mutations in
bopoietin receptor MPL and render cells cytokine indepen- PV, ET, and PMF
dent. Mutant but not wild-type CALR has been shown to
activate MPL and result in downstream STAT, MAPK, and In addition to mutations in JAK2, MPL, and CALR, a num-
PI3K activation in cell lines in the absence of thrombopoietin. ber of other somatically acquired mutations are found in
Mutant CALR is now known to directly interact with MPL MPNs. In general, these additional mutations are not exclu-
within the ER. This interaction has been shown to be depen- sive to MPNs and are found across myeloid malignancies, and
dent on both the extracellular N-glycosylation residues of in some cases are also found in lymphoid malignancies and
MPL and the mutant carboxyl-terminus of CALR. It is possi- solid tumors. Exome sequencing of MPNs has shown that PV
ble that the loss of –KDEL allows mutant CALR, coupled with and ET have on average six to seven mutations per case, and
MPL, to leave the ER. Activation of MPL is believed to occur PMF has a significantly higher number of mutations, in keep-
while in complex with CALR in a TPO-independent manner, ing with its being a more advanced phase of disease. Over-
which subsequently results in downstream constitutive JAK- all, the burden of somatic mutations in MPNs is similar to
STAT signaling. Mice that are recipients of bone marrow cells that observed in other chronic and acute hematological neo-
retrovirally transduced with either CALR 52-bp deletion or plasms, and is roughly one log-fold lower than that seen in
CALR 5-bp insertion develop PMF and ET, respectively, reca- common solid tumors, such as epithelial cancers.
pitulating the disease seen in patients and demonstrating that Up to one-third of MPN patients harbor additional
mutant CALR is sufficient to cause sustained megakaryocyte mutations in genes that regulate the epigenome, either via
proliferation and increased platelet production. DNA methylation, such as TET2, DNMT3A, and IDH1/2,
Given this novel interaction between mutant CALR and or chromatin remodeling, such as EZH2 and ASXL1.
MPL, it is unsurprising that mutations in CALR result in an TET (Ten-eleven translocation methylcytosine dioxyge-
MPN phenotype that is remarkably similar to that caused by nase) proteins are involved in the demethylation of DNA
MPL mutations, with both being associated with ET or PMF. through the conversion of 5-methylcytosine (5-mc) to
Both MPL- and CALR-mutated ET are characterized by an 5-hydroxymethylcytosine (5-hmc). Loss-of-function muta-
isolated thrombocytosis, unlike JAK2-mutated ET, which is tions are found in 10% of MPNs and are associated with HSC
associated with an increase in trilineage hematopoiesis and expansion and myeloid progenitor expansion in murine
modest increases in hemoglobin and white cell counts, in models and TET2-mutated MPN patients. DNMT3A is a de
addition to thrombocytosis. Clinically, patients with CALR novo DNA methyltransferase that is also mutated in around
mutations are younger and suffer fewer thromboses com- 10% of MPNs. Mutations are also believed to be loss of
pared to JAK2-mutated counterparts. Elevated white blood function, and the commonest single-nucleotide mutation at
cell counts correlate with an increased risk of thrombosis R882 has been shown to confer a dominant negative effect
in ET. Therefore, it is possible that the differences in the over wild-type DNMT3A. Loss of DNMT3A also results in
incidence of thrombosis between JAK2V617F- and CALR- marked HSC compartment expansion in mouse models, and
mutated patients reflect the association between JAK2V617F in human cells has been shown to confer a stem cell advan-
and leukocytosis in ET patients, or this may be due to tage over DNMT3A-wild-type cells. Clinical studies have
mutant JAK2-related platelet and leucocyte activation. The not demonstrated consistent associations between DNMT3A
thrombotic tendency of JAK2V617F-mutated ET may also be or TET2 mutations and alterations to MPN phenotype or
related to the increased erythrocytosis that is evident in this outcome. However, these mutations are some of the most
subgroup compared to JAK2-unmutated ET patients. Within common somatically acquired genetic aberrations to be
PMF, molecular status also affects clinical outcome, with found in the blood of elderly individuals who have normal
CALR-mutated patients carrying a better overall survival blood count parameters, and their presence is associated
compared to patients with either JAK2 mutations or those with clonal hematopoiesis. Mutations affecting histone
that are triple negative (JAK2, CALR, and MPL unmutated). methylation are common in advanced MPNs. Affected genes
The identification of mutations in JAK2, MPL, and CALR code for proteins of the Polycomb repressive complex 2
has dramatically improved diagnosis for MPN patients and (PRC2) that normally trimethylates histone H3 at lysine
molecular testing is now used routinely in the clinical setting. 23 to repress transcription. Loss-of-function mutations
Myeloproliferative neoplasms 95

in ASXL1 (Additional sex combs like 1) are present in a Other mutations reported in patients with CNL include those
quarter of PMF patients and have been shown to cause in ASXL1 (50–60% of patients), SETBP1 (14–43%), TET2
myeloproliferation and anemia in murine models. Mutations (30%), and CALR (<10%), with ASXL1 mutations appearing
in EZH2 (PcG Enhancer of Zeste Homolog 2) are also seen to confer an adverse prognosis.
in <5% of MPNs, and are more frequent in advanced disease. Among the clonal stem cell disorders associated with
A further distinct category of mutations observed in MPNs eosinophilia, the most common molecular abnormalities are
reside in genes that code for proteins of the intracellular fusion genes that involve the PDGFRA, PDGFRB, or FGFR1
spliceosome machinery, such as SF3B1, U2AF1, SRSF2, and genes and result in constitutive activation of a tyrosine
ZRSR2. Mutations are present in 2–5% of MPNs, less frequent kinase. This group includes both myeloid and lymphoid
than in myelodysplastic syndromes. In some cases, muta- diseases, and these are now categorized separately within the
tions (such as those affecting U2AF1 or SRSF2) are asso- WHO classification as “Myeloid/lymphoid neoplasms with
ciated with a poorer prognosis when associated with PMF. eosinophilia and rearrangement of PDGFRA, PDGFRB, or
Patients with splicing factor mutations often have myelodys- FGFR1, or with PCM1-JAK2.” The commonest lesion is a
plastic features on bone marrow examination, and in some submicroscopic (800 kb) deletion of chromosome 4q12 that
cases distinct morphological features are seen, such as the leads to the generation of a FIP1L1–PDGFRA fusion gene,
presence of ringed sideroblasts in association with mutated which is not visible with standard G banding but can be
SF3B1. Mutations result in distinct alterations to mRNA detected by nested reverse transcriptase-polymerase chain
motif recognition sites and mis-spliced target genes are often reaction (RT-PCR) or by fluorescence in situ hybridization
important players in myeloid leukemogenesis. Interestingly, (FISH) using a probe (CHIC2) within the deleted region.
mutations in splicing factors are generally mutually exclu- Other lesions can be detected by FISH or as rearrangements
sive. This suggests either that cells cannot tolerate more than within an abnormal karyotype. Patients who lack these
one aberration to the splicing machinery, or that one mutated fusion genes but demonstrate a significant unexplained
component is sufficient to alter overall function, such eosinophilia can however be diagnosed with chronic
that additional mutations do not provide a further clonal eosinophilia leukemia, not otherwise specified (CEL, NOS)
advantage. if an alternative clonal marker is identified. For example, the
Additional mutations in MPNs are found more frequently JAK2V617F mutation and KIT D816V (typically associated
in the context of disease progression to post-MPN AML, such with mastocytosis) have been identified in 3–4% of patients
as RUNX1, IDH1/2, and TP53. In some cases monoallelic loss with hypereosinophilia of unknown significance and may
of TP53, either through del17p or genetic mutation, can be coexist with a variety of other mutations, such as in TET2,
detected during chronic-phase disease, with acquisition of SRSF2, EZH2, and ASXL1.
biallelic loss associated with the progression of the disease.

Integration of molecular analysis into


Chronic neutrophilic leukemia and diagnostic and management pathways
chronic eosinophilic leukemia
In recent years, clinical classification schemes have been
CNL is a rare MPN characterized by expansion of seg- greatly enhanced in their ability to aid diagnosis, prognosis,
mented neutrophils in the blood and bone marrow granulo- and therapy by the incorporation of their relevant molecular
cytic hyperplasia, without an increase in immature blast cells features. In the case of the MPNs, the impact of newly identi-
or morphological dysplasia. Mutations in the gene for the fied mutations has been so significant that schemes have had
colony-stimulating factor 3 receptor, CSF3R, had previously to change dramatically to accommodate them. Classification
been found in individuals with severe congenital neutrope- of the MPNs, first attempted by the Polycythemia Vera
nia, but in 2013 were identified in a significant proportion of Study Group (PVSG), was a tool for distinguishing these
patients with CNL. Although mutations were also reported disorders from phenotypically similar clonal and non-clonal
in the related MDS/MPN atypical CML, subsequent stud- blood disorders. The identification and characterization
ies have suggested that CSF3R mutations are relatively spe- of JAK2, MPL, and CALR mutations have offered a set of
cific for CNL and can be found in most patients with CNL highly specific diagnostic tests for the MPNs. New simplified
defined strictly according to WHO criteria. There are two classification systems have been devised that incorporate
classes of activating CSF3R mutations. Membrane-proximal the molecular changes underlying the MPNs (Tables 7.1 and
mutations, the commonest of which is T618I, cause ligand- 7.2). As shown in these tables, both approaches empha-
independent signaling through the JAK-STAT pathway and size the importance of a JAK2, MPL, or CALR mutation,
may be amenable to pharmacological inhibition with JAK although they differ in the significance attached to these
inhibitors such as ruxolitinib. Truncation mutations, by con- markers as compared to other clinical and histological
trast, cause dysregulation of SRC family and TNK2 kinases. criteria.
96 Molecular Hematology

Table 7.1 The 2016 World Health Organization (WHO) diagnostic criteria for polycythemia vera, essential thrombocythemia, and primary
myelofibrosisa

Polycythemia vera
Major

1. Hb >165 g l−1 (men), >160 g l−1 (women) or Hct >0.49 (men), >0.48 (women) or Elevated red cell mass >25% above mean normal
predicted value
2. BM biopsy features: hypercellularity; trilineage proliferation (erythroid, granulocytic, and megakaryocytic); pleomorphic, mature
megakaryocytes
3. JAK2V617F or JAK2 exon 12 mutation

Minor

Subnormal serum erythropoietin level

Essential thrombocythemia
Major
1. Platelet count ≥450 × 109 l−1
2. BM biopsy features: predominantly megakaryocytic hyperplasia; large, mature megakaryocytes
3. Not meeting criteria for another myeloid neoplasm (e.g. CML, PV, PMF, MDS)
4. JAK2, CALR, or MPL mutation
Minor

Another clonal marker; or absence of reactive thrombocytosis

Primary myelofibrosis (overt)


Major

1. Megakaryocytic hyperplasia and atypia; reticulin and/or collagen fibrosis


2. Not meeting criteria for another myeloid neoplasm (e.g. CML, ET, PV, MDS)
3. JAK2, CALR, or MPL mutation; in the absence of these mutations, another clonal marker (e.g. ASXL1, EZH2, TET2, IDH1/2 mutation) or
absence of reactive myelofibrosis

Minor
1. Unexplained anemia
2. Leukocytosis ≥11 × 109 l−1
3. Splenomegaly (palpable)
4. Increased serum LDH
5. Leucoerythroblastic blood film

BM, bone marrow; CML, chronic myeloid leukemia; Hb, hemoglobin; Hct, hematocrit; LDH, lactate dehydrogenase; MDS, myelodysplastic
syndrome.
a Diagnosis of polycythemia vera (PV) requires either all three major criteria, or the first two major criteria and the minor criterion.

Diagnosis of essential thrombocythemia requires all four major criteria or the first three major criteria and the minor criterion.
Diagnosis of overt primary myelofibrosis (PMF) requires all three major criteria and at least one minor criterion. Separate criteria exist for the
diagnosis of pre-fibrotic primary myelofibrosis, the principal difference being the absence of reticulin fibrosis >grade 1.

Molecular markers do not only offer a simplified strat- inform the potential benefit of targeted therapies, an exam-
egy for diagnosis, but also add prognostic information that ple being the use of JAK inhibitors in patients with CNL and
can guide management. For example, mutations in ASXL1, a membrane-proximal CSF3R mutation.
SRSF2, EZH2, and IDH1/2 are associated with an adverse
prognosis in PMF, and in the case of ASXL1 this is inde-
pendent of the International Prognostic Scoring System. Conclusions and future directions
Mutational analysis may therefore aid in decisions such as
the appropriateness of allogeneic stem cell transplantation. Identification of JAK2, MPL, and CALR mutations that
In the future, molecular information also may increasingly are relatively specific to PV, ET, and PMF has reinforced
Myeloproliferative neoplasms 97

Table 7.2 British Committee for Standards in Haematology (BCSH) diagnostic criteria for polycythemia vera, essential thrombocythemia, and
primary myelofibrosisa

Polycythemia vera
A1 High hematocrit (men >52%, women >48%) or an increased red cell mass (>25% above predicted value)
A2 Mutation in JAK2

Essential thrombocythemia
A1 Platelet count >450 × 109 l−1
A2 Presence of an acquired pathogenetic mutation (e.g. in JAK2, CALR, or MPL genes)
A3 No other myeloid malignancy, especially JAK2-positive PV, PMF, CML, or MDS
A4 No reactive cause for thrombocytosis and normal iron stores
A5 Bone marrow aspirate and trephine biopsy showing increased megakaryocytes with a spectrum of morphology, predominantly large
with hyperlobated nuclei and abundant cytoplasm. Reticulin generally not increased.

Primary myelofibrosis
A1 Reticulin grade 3 or higher (on a 0–4 scale)
A2 Pathogenetic mutation or absence of both BCR-ABL1 and reactive causes of bone marrow fibrosis
B1 Palpable splenomegaly
B2 Unexplained anemia
B3 Leucoerythroblastosis
B4 Teardrop red cells
B5 Constitutional symptoms (drenching night sweats, weight loss >10% over six months, unexplained fever, or diffuse bone pain)
B6 Histological evidence of extramedullary hematopoiesis

Diagnosis of essential thrombocythemia requires A1 − A3, or A1 + A3 − A5.


Diagnosis of primary myelofibrosis (PMF) requires A1 + A2 and any two of the B criteria.
CML, chronic myeloid leukemia; MDS, myelodysplastic syndrome.
a Diagnosis of polycythemia vera (PV) requires both criteria to be present. Separate criteria exist for the diagnosis of JAK2-negative PV.

Source: Adapted from McMullin M.F., Harrison C.N., Ali S. et al. (2019). A guideline for the diagnosis and management of polycythaemia vera. A
British Society for Haematology Guideline. Br. J. Haematol. 184: 176–191. Harrison C.N., Butt N., Campbell P., et al. (2014). Modification of British
Committee for Standards in Haematology diagnostic criteria for essential thrombocythaemia. Br. J. Haematol. 167: 421–3; Reilly J.T., McMullin
M.F., Beer P., et al. (2012). Guideline for the diagnosis and management of myelofibrosis. Br. J. Haematol. 158: 453–71, with permission.

Dameshek’s dictum that these disorders are closely linked. AML with its poor prognosis. It is also increasingly evident
It has also revolutionized all aspects of the MPNs, including that clinical heterogeneity in all of the chronic myeloid dis-
our understanding of pathogenetic mechanisms, clarification orders reflects significant underlying mutational heterogene-
of diagnostic criteria, and, importantly, development of new ity, and in future more comprehensive mutational analysis
therapies. The discovery of the JAK2V617F mutation led to might be able to facilitate a more “personalized” approach to
the rapid development of JAK inhibitors, the first of which therapy.
was approved for use in PMF just seven years later in 2012,
and was subsequently also licensed for some patients with
PV. A greater understanding of the additional mutations in
MPNs, especially those affecting histone modifications, has Further reading
prompted clinical trials of other novel drugs such as histone
deacetylase inhibitors. Introduction
In spite of these advances, important challenges remain in
Arber, D.A., Orazi, A., Hasserjian, R. et al. (2016). The 2016 revision to
the understanding of MPN disease biology and its translation
the World Health Organization classification of myeloid neoplasms
into improving clinical outcomes. Studies of mutational allele
and acute leukemia. Blood 127: 2391–2405.
burdens in patients taking pharmaceutical agents demon- Bench, A.J. and Pahl, H.L. (2005). Chromosomal abnormalities and
strate that none of these have a reliable, selective, inhibitory molecular markers in myeloproliferative disorders. Semin. Hematol.
effect on the HSCs that drive disease. This issue must be tack- 42: 196–205.
led if therapy is to reverse the underlying disease process Campbell, P.J. and Green, A.R. (2006). The myeloproliferative disorders.
and reduce the risk of disease transformation, particularly to N. Engl. J. Med. 355: 2452–2466.
98 Molecular Hematology

Dameshek, W. (1951). Some speculations on the myeloproliferative syn- Other JAK2 mutations in MPNs
dromes. Blood 6: 372–375.
Prchal, J.F. and Axelrad, A.A. (1974). Letter: Bone-marrow responses in Grisouard, J., Li, S., Kubovcakova, L. et al. (2016). JAK2 exon 12 mutant
polycythemia vera. N. Engl. J. Med. 290: 1382. mice display isolated erythrocytosis and changes in iron metabolism
favoring increased erythropoiesis. Blood 128: 839–851.
Kapralova, K., Horvathova, M., Pecquet, C. et al. (2016). Cooperation
The JAK2V617F mutation in of germ line JAK2 mutations E846D and R1063H in hereditary ery-
throcytosis with megakaryocytic atypia. Blood 128: 1418–1423.
Philadelphia-negative MPNs
Mead, A.J., Rugless, M.J., Jacobsen, S.E., and Schuh, A. (2012). Germline
Anand, S. and Huntly, B.J. (2012). Disordered signaling in myelopro- JAK2 mutation in a family with hereditary thrombocytosis. N. Engl.
liferative neoplasms. Hematol. Oncol. Clin. North Am. 26: 1017– J. Med. 366: 967–969.
1035. Milosevic Feenstra, J.D., Nivarthi, H., Gisslinger, H. et al. (2016).
Baxter, E.J., Scott, L.M., Campbell, P.J. et al. (2005). Acquired mutation Whole-exome sequencing identifies novel MPL and JAK2 mutations
of the tyrosine kinase JAK2 in human myeloproliferative disorders. in triple-negative myeloproliferative neoplasms. Blood 127: 325–332.
Lancet 365: 1054–1061. Passamonti, F., Elena, C., Schnittger, S. et al. (2011). Molecular and clin-
Godfrey, A.L., Chen, E., Pagano, F. et al. (2012). JAK2V617F homozy- ical features of the myeloproliferative neoplasm associated with JAK2
gosity arises commonly and recurrently in PV and ET, but PV is char- exon 12 mutations. Blood 117: 2813–2816.
acterized by expansion of a dominant homozygous subclone. Blood Scott, L.M., Tong, W., Levine, R.L. et al. (2007). JAK2 exon 12 mutations
120: 2704–2707. in polycythemia vera and idiopathic erythrocytosis. N. Engl. J. Med.
Godfrey, A.L. and Green, A.R. (2012). Genotype–phenotype inter- 356: 459–468.
actions in the myeloproliferative neoplasms. Hematol. Oncol. Clin.
North Am. 26: 993–1015.
James, C., Mazurier, F., Dupont, S. et al. (2008). The hematopoietic MPL mutations in ET and PMF
stem cell compartment of JAK2V617F-positive myeloproliferative
Beer, P.A., Campbell, P.J., Scott, L.M. et al. (2008). MPL mutations in
disorders is a reflection of disease heterogeneity. Blood 112: 2429–
myeloproliferative disorders: analysis of the PT-1 cohort. Blood 112:
2438.
141–149.
James, C., Ugo, V., Le Couedic, J.P. et al. (2005). A unique clonal JAK2
Guglielmelli, P., Pancrazzi, A., Bergamaschi, G. et al. (2007). Anaemia
mutation leading to constitutive signalling causes polycythaemia
characterises patients with myelofibrosis harbouring Mpl mutation.
vera. Nature 434: 1144–1148.
Br. J. Haematol. 137: 244–247.
Jones, A.V. and Cross, N.C. (2013). Inherited predisposition to myelo-
Pecquet, C., Staerk, J., Chaligne, R. et al. (2010). Induction of myelo-
proliferative neoplasms. Ther. Adv. Hematol. 4: 237–253.
proliferative disorder and myelofibrosis by thrombopoietin receptor
Jutzi, J.S., Bogeska, R., Nikoloski, G. et al. (2013). MPN patients harbor
W515 mutants is mediated by cytosolic tyrosine 112 of the receptor.
recurrent truncating mutations in transcription factor NF-E2. J. Exp.
Blood 115: 1037–1048.
Med. 210: 1003–1019.
Pikman, Y., Lee, B.H., Mercher, T. et al. (2006). MPLW515L is a novel
Keil, E., Finkenstadt, D., Wufka, C. et al. (2014). Important scaffold
somatic activating mutation in myelofibrosis with myeloid metapla-
function of the Janus kinase 2 uncovered by a novel mouse model
sia. PLoS Med. 3: e270.
harboring a Jak2 activation-loop mutation. Blood 123: 520–529.
Vannucchi, A.M., Antonioli, E., Guglielmelli, P. et al. (2008). Character-
Kralovics, R., Passamonti, F., Buser, A.S. et al. (2005). A gain-of-function
istics and clinical correlates of MPL 515W>L/K mutation in essential
mutation of JAK2 in myeloproliferative disorders. N. Engl. J. Med.
thrombocythemia. Blood 112: 844–847.
352: 1779–1790.
Levine, R.L., Wadleigh, M., Cools, J. et al. (2005). Activating mutation
in the tyrosine kinase JAK2 in polycythemia vera, essential thrombo- CALR mutations in ET and PMF
cythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell 7:
387–397. Araki, M., Yang, Y., Masubuchi, N. et al. (2016). Activation of the throm-
Li, J., Kent, D.G., Chen, E., and Green, A.R. (2011). Mouse models of bopoietin receptor by mutant calreticulin in CALR-mutant myelo-
myeloproliferative neoplasms: JAK of all grades. Dis. Model Mech. 4: proliferative neoplasms. Blood 127: 1307–1316.
311–317. Chachoua, I., Pecquet, C., El-Khoury, M. et al. (2016). Thrombopoietin
Ortmann, C.A., Kent, D.G., Nangalia, J. et al. (2015). Effect of mutation receptor activation by myeloproliferative neoplasm associated calreti-
order on myeloproliferative neoplasms. N. Engl. J. Med. 372: 601–612. culin mutants. Blood 127: 1325–1335.
Tapper, W., Jones, A.V., Kralovics, R. et al. (2015). Genetic variation at Klampfl, T., Gisslinger, H., Harutyunyan, A.S. et al. (2013). Somatic
MECOM, TERT, JAK2 and HBS1L-MYB predisposes to myeloprolif- mutations of calreticulin in myeloproliferative neoplasms. N. Engl. J.
erative neoplasms. Nat. Commun. 6: 6691. Med. 369: 2379–2390.
Vainchenker, W., Delhommeau, F., Constantinescu, S.N., and Bernard, Kollmann, K., Warsch, W., Gonzalez-Arias, C. et al. (2017). A novel
O.A. (2011). New mutations and pathogenesis of myeloproliferative signalling screen demonstrates that CALR mutations activate essen-
neoplasms. Blood 118: 1723–1735. tial MAPK signalling and facilitate megakaryocyte differentiation.
Zhao, R., Follows, G.A., Beer, P.A. et al. (2008). Inhibition of the Bcl-xL Leukemia 31: 934–944.
deamidation pathway in myeloproliferative disorders. N. Engl. J. Med. Marty, C., Harini, N., Pecquet, C. et al. (2014). Calr mutants retroviral
359: 2778–2789. mouse models lead to a myeloproliferative neoplasm mimicking an
Myeloproliferative neoplasms 99

essential thrombocythemia progressing to a myelofibrosis. Blood 124: Pardanani, A., Lasho, T.L., Laborde, R.R. et al. (2013). CSF3R T618I
157–157. is a highly prevalent and specific mutation in chronic neutrophilic
Nangalia, J., Massie, C.E., Baxter, E.J. et al. (2013). Somatic CALR muta- leukemia. Leukemia 27: 1870–1873.
tions in myeloproliferative neoplasms with nonmutated JAK2. N. Schwaab, J., Umbach, R., Metzgeroth, G. et al. (2015). KIT D816V and
Engl. J. Med. 369: 2391–2405. JAK2 V617F mutations are seen recurrently in hypereosinophilia of
unknown significance. Am. J. Hematol. 90: 774–777.
Other somatic mutations in PV, ET, and PM
Integration of molecular analysis into
Grinfeld, J., Nangalia, J., and Green, A.R. (2017). Molecular determi-
diagnostic and management pathways
nants of pathogenesis and clinical phenotype in myeloproliferative
neoplasms. Haematologica 102: 7–17. Harrison, C.N., Butt, N., Campbell, P. et al. (2014). Modification of
Guglielmelli, P., Lasho, T.L., Rotunno, G. et al. (2014). The number British Committee for Standards in Haematology diagnostic criteria
of prognostically detrimental mutations and prognosis in primary for essential thrombocythaemia. Br. J. Haematol. 167: 421–423.
myelofibrosis: an international study of 797 patients. Leukemia 28: McMullin M.F., Harrison C.N., Ali S. et al. (2019). A guideline for the
1804–1810. diagnosis and management of polycythaemia vera. A British Society
Vannucchi, A.M., Lasho, T.L., Guglielmelli, P. et al. (2013). Mutations for Haematology Guideline. Br. J. Haematol. 184: 176–191.
and prognosis in primary myelofibrosis. Leukemia 27: 1861–1869. Reilly, J.T., McMullin, M.F., Beer, P.A. et al. (2012). Guideline for the
diagnosis and management of myelofibrosis. Br. J. Haematol. 158:
Chronic neutrophilic leukemia and chronic 453–471.
Vannucchi, A.M., Lasho, T.L., Guglielmelli, P. et al. (2013). Mutations
eosinophilic leukemia
and prognosis in primary myelofibrosis. Leukemia 27: 1861–1869.
Cools, J., DeAngelo, D.J., Gotlib, J. et al. (2003). A tyrosine kinase
created by fusion of the PDGFRA and FIP1L1 genes as a thera- Conclusions and future directions
peutic target of imatinib in idiopathic hypereosinophilic syndrome.
https://doi.org/10.1056/NEJMoa025217. N. Engl. J. Med. 348: 1201– Andersen, C.L., McMullin, M.F., Ejerblad, E. et al. (2013). A phase II
1214. study of vorinostat (MK-0683) in patients with polycythaemia vera
Maxson, J.E., Gotlib, J., Pollyea, D.A. et al. (2013). Oncogenic CSF3R and essential thrombocythaemia. Br. J. Haematol. 162: 498–508.
mutations in chronic neutrophilic leukemia and atypical CML. N. Harrison, C., Kiladjian, J.J., Al-Ali, H.K. et al. (2012). JAK inhibi-
Engl. J. Med. 368: 1781–1790. tion with ruxolitinib versus best available therapy for myelofibrosis.
Meggendorfer, M., Haferlach, T., Alpermann, T. et al. (2014). N. Engl. J. Med. 366: 787–798.
Specific molecular mutation patterns delineate chronic neu- Verstovsek, S., Mesa, R.A., Gotlib, J. et al. (2012). A double-blind,
trophilic leukemia, atypical chronic myeloid leukemia, and chronic placebo-controlled trial of ruxolitinib for myelofibrosis. N. Engl. J.
myelomonocytic leukemia. Haematologica 99: e244–e246. Med. 366: 799–807.
Chapter 8 Lymphoma genetics
Jennifer L. Crombie1 , Anthony Letai1 & John G. Gribben2
1 Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston MA, USA
2
Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK

Introduction, 101 Follicular lymphoma, 106


Techniques, 101 Lymphoplasmacytic lymphoma, 106
Commonly occurring translocations in lymphoma, 103 Mucosa-associated lymphoid tissue lymphomas, 107
Burkitt lymphoma, 104 Anaplastic large cell lymphoma, 107
Diffuse large B-cell lymphoma, 105 Chronic lymphocytic leukemia, 107
High-grade B-cell lymphomas with MYC and BCL2 or BCL6 Hodgkin lymphoma, 108
translocations, 105 Conclusions, 108
Mantle cell lymphoma, 106 Further reading, 109

Introduction Techniques

As with all cancers, lymphomas were originally categorized Techniques for studying genetic abnormalities in tumor spec-
based primarily on morphology and clinical behavior. The imens have undergone a revolution in the past two decades
use of antibodies against cell surface markers allowed the (Table 8.1). Initial genetic analyses were based on the tech-
study of lymphoma specimens with antibody panels that nique of chromosomal study by Giemsa-trypsin banding. In
could, along with morphological criteria, usually place a these studies, cells are grown in short-term culture, usually
given lymphoma into a diagnostic category. Even within a in the presence of mitogens. Colcemid treatment results in
given lymphoma category, however, there is considerable cell accumulation in metaphase, at which point the cells are
heterogeneity of clinical behavior. A prominent example is fixed and dropped on glass slides. The slides are treated with
diffuse large B-cell lymphoma (DLBCL), in which over two- trypsin followed by Giemsa to give a banding pattern. An
thirds of patients are cured with conventional chemotherapy, experienced cytogenetic technician can then identify normal
whereas those with relapsed/refractory disease have dismal chromosomes, translocations, numerical abnormalities, and
outcomes. In this case, as with many of the lymphomas, sometimes more subtle deletions. The technique can iden-
a prognostic index (the International Prognostic Index tify only genetic changes large enough to disrupt a Giemsa-
or IPI) based on pre-treatment clinical criteria is able to stained band, requiring a change of many megabases.
provide very useful prognostic information. However, even More modern techniques are able to detect abnormalities
among patients with the same IPI score, significant clinical with greater sensitivity. Southern hybridization starts with
heterogeneity persists. Furthermore, it is likely that the IPI the electrophoretic separation of tumor DNA on a gel, fol-
defines subclasses of lymphomas based upon biological lowed by transfer to a membrane. This membrane is then
differences among these lymphomas. Studies of genetic probed with radioactively labeled polynucleotide probes spe-
abnormalities have proved to be important tools to further cific for certain genes of interest. Changes in the expected
classify and prognosticate lymphomas. In addition, a better size or intensity of the band of interest can indicate muta-
understanding of the molecular pathophysiology has led to tion, translocation, amplification, or deletion of the gene of
improvements in therapeutic options. interest. It can also be used to evaluate the presence of clonal

Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.

101
102 Molecular Hematology

genome-wide scan for abnormalities, requiring no prior sus-


Table 8.1 Techniques to study lymphoma genetics
picion of a particular abnormality which can have a clinical
r Cytogenetic analysis impact, are comparative genomic hybridization (CGH), gene
r Southern blot analysis expression profiling (GEP), single-nucleotide polymorphism
r Polymerase chain reaction (PCR) analysis (SNP) arrays, micro-RNA arrays, next-generation sequenc-
r Fluorescent in situ hybridization (FISH) ing (NGS), and clustered regularly interspersed short palin-
r Comparative genomic hybridization (CGH)
dromic repeats (CRISPR)-Cas9 based lethality screens.
r CGH microarray
In original CGH techniques, DNA is isolated from the
r Gene expression profiling
tumor sample and a normal control sample. The DNA in
r Single-nucleotide polymorphism (SNP) array
r microRNA expression profiling
each is labeled with a different fluorescent dye, for example
r Next-generation sequencing (NGS): whole-genome sequencing, green for the tumor DNA and red for the normal DNA. These
samples are mixed and hybridized onto slides of metaphase
whole-exome sequencing, RNA sequencing, and ChIP
sequencing
spreads of normal cells. Images of metaphase spreads are
r Clustered regularly interspersed short palindromic repeats then analyzed for the green-to-red color ratio. Regions of
(CRISPR)-Cas9 functional genetic screening chromosomes that have a high green-to-red ratio contain
a putative area of amplification. Regions that have a low
green-to-red ratio contain a putative deletion. In this way,
the entire genome may be examined for abnormalities. Other
techniques, generally beyond what is performed in clinical
rearrangements of the immunoglobulin loci in B cells or the laboratories, are required to determine the critical genes
T-cell receptor locus in T cells. involved in areas of amplification and deletion. Small abnor-
Polymerase chain reaction (PCR) technology has allowed malities and balanced translocations cannot be observed
for the detection of genetic abnormalities using only a small using this technique. Currently in more common use is a
amount of tumor DNA. Using primers designed to flank variation of the CGH technique (known as “array CGH”)
the genomic region of interest, repetitive cycles of anneal- which hybridizes the DNA to defined arrays of genomic
ing, DNA polymerization, and thermal melting eventually DNA fragments. These arrays can cover the genome and
yield a PCR product. The presence and size of this product improve resolution to the size of the DNA fragments within
may be analyzed by gel electrophoresis to determine the pres- the microarray, currently down to the level of 1 MB.
ence of a translocation. Furthermore, a PCR product may be GEP, which allows for a comprehensive, quantitative
sequenced to look for point mutations. Like Southern blot- examination of the mRNA transcripts of a tumor sample, a
ting, PCR can also be used to evaluate the presence of clonal group of molecules that has been termed the “transcriptome,”
rearrangements of the immunoglobulin loci in B cells or the was one of the original tools to provide a deeper insight
T-cell receptor locus in T cells. into the molecular underpinnings of lymphoma. In this tech-
Fluorescent in situ hybridization (FISH) uses fluores- nique, mRNA is purified from a fresh or frozen tumor sam-
cently labeled DNA probes to bind to specific regions of ple, amplified, and fluorescently labeled. The labeled RNA
genomic DNA. Images are then analyzed under a fluorescent is then hybridized to oligonucleotide probes, which can
microscope. Numerical chromosomal abnormalities may be be quantitated by microscopy and image analysis software.
detected by simply counting the number of signals per cell: Comparison of different tumor samples, and comparison
more than two indicates the addition of a chromosome, with wild type, can then allow the determination of tran-
whereas less than two indicates a deletion. To investigate a scripts that are over- or underrepresented in certain con-
potential translocation, two probes are used, one to detect ditions or tumors. Tumors may be categorized using these
the genomic DNA on each side of the known translocation. If profiles, and subgroups of messages may be used to create
the two probes are consistently approximated, this indicates predictors of clinical behavior. For example, GEP has been
the presence of a translocation. FISH may be performed on used use to characterize two molecular subgroups of DLBCL,
interphase cells, so that growth in culture is not a requirement germinal center B-cell (GCB) subtype and activated B-cell
for this type of analysis as it is for conventional cytogenetics. (ABC) subtype, which have both prognostic and therapeu-
Tests for small deletions or other subtle abnormalities may be tic implications. In recent years, RNA sequencing using NGS,
better performed on metaphase cells. FISH requires knowl- which is discussed in greater detail shortly, has been replac-
edge of the area to be labeled. ing microarray assays for evaluation of RNA expression.
Since they rely on the annealing of a labeled specific DNA Whole-genome analyses of SNPs have also been facili-
probe or primer, Southern hybridization, PCR, and FISH are tated by the development of SNP arrays. DNA fragments
techniques to determine the presence or absence of a known are digested and annealed to oligonucleotide linkers, ampli-
genetic abnormality. Modern techniques which provide a fied, and hybridized to an SNP array plate. Hundreds of
Lymphoma genetics 103

thousands of oligonucleotides, representing all known SNPs, allowing physicians access to the genomic landscape and the
can be assessed for using this method. This can identify ability to personalize management.
which SNPs are present in a given tumor sample. Simi- Similarly, high-throughput RNA sequencing (RNA-Seq),
larly, consistent loss of all SNPs along a region indicates which involves NGS of cDNA, is able to measure the pres-
a region of chromosomal loss, whereas consistent increase ence and quantity of RNA in a biological sample. This tech-
in SNP signal at a particular region indicates amplification. nique can identify alternatively spliced gene transcripts, gene
As with GEP, newer NGS-based methods are replacing this fusions, and post-transcriptional modifications. RNA-Seq
technology. can also identify different populations of RNA in a sample,
miRNAs are small non-coding RNA transcripts that act including total RNA, miRNA, and tRNA.
primarily by modulating the expression of other genes. miR- Lastly, technologies such as chromatin immunoprecipita-
NAs exert this function by annealing to mRNAs, the con- tion (ChIP) assays can be combined with NGS in a pro-
sequence of which can be shortened mRNA half-life or cess known as chromatin immunoprecipitation–sequencing
decreased translation. Any single miRNA can modulate the (ChIP-Seq). This is a powerful method to determine genome-
expression at many different genes. It has been difficult, how- wide binding sites for transcription factors and other pro-
ever, to use the primary sequence of the miRNA to predict teins. Together, these technologies allow for high-throughput
the genes at which it acts. Several studies have demonstrated sequencing of thousands of genes from small clinical sam-
that by measuring the levels of the hundreds of known miR- ples, readily providing massive amounts of data on lym-
NAs in the genome, one can segregate cancers into different phoma genomics and transcriptomics.
groups. Furthermore, such segregation may provide infor- Genome editing technology based on the prokaryotic
mation about prognosis, progression, and response to ther- CRISPR–Cas9 system has also revolutionized genome engi-
apy. Supporting the concept that miRNAs play an important neering and our ability to identify genes that are critical for
role in determining cancer behavior, non-coding regions of survival. In this technique, libraries of small-guide RNAs
the genome that are frequently deleted in cancer often con- (sgRNAs) can guide the Cas9 enzyme to target a host of
tain miRNA genes. Chronic lymphocytic leukemia (CLL) is genes in lymphoma models. Next-generation DNA sequenc-
an example of a lymphoid cancer for which miRNA biology ing can subsequently be used to identify genes of interest.
has been informative. For instance, it has been found that in This functional genomics approach identifies mutations with
many cases of CLL, particularly those of indolent behavior, functional consequence and thus has the potential to identify
there is downregulation or deletion of mir-15-A and mir-16- optimal therapeutic targets.
1, located at 13q14.3. Both of these miRNAs have the ability
to decrease BCL-2 levels, so their deletion may explain the
high levels of BCL-2 found in nearly all CLL cells. In addi- Commonly occurring translocations
tion, expression levels of a limited number of miRNAs may in lymphoma
distinguish between indolent and aggressive clinical subtypes
in CLL. Commonly occurring translocations in lymphoma may
More recently, NGS has revolutionized DNA and RNA be crudely divided into two main classes: those that foster
analysis. Whole-exome sequencing (WES), for example, has increased proliferation, and those that inhibit programmed
been utilized on paired tumor and germline DNA sam- cell death, or apoptosis. The classical gene in lymphomagen-
ples to identify recurrently, somatically mutated protein- esis that induces proliferation is c-MYC. Burkitt’s lymphoma
coding genes in lymphoma, allowing for the identification of (BL), one of the most rapidly dividing lymphomas, is the
genes not previously recognized as drivers of cancer. Whole- archetype of a lymphoma that overexpresses c-MYC by
genome sequencing (WGS), which as the name implies the t(8;14). c-MYC is a helix-loop-helix leucine zipper
results in sequencing of the entire genome, has the ability transcription factor, which requires heterodimerization
also to interrogate variants that may be important for con- with the protein MAX to activate transcription and induce
trolling gene transcriptional regulation or splicing. Results proliferation. Targets of this dimer include genes controlling
from these sequencing tools have allowed for the develop- cell cycle progression, cell growth, metabolism, differenti-
ment of clinical gene panels able to identify frequently occur- ation, and apoptosis. The net effect of c-MYC expression is
ring mutations with potential therapeutic impact. These generally an increase in proliferation; however, this effect is
panels are transforming the management of hematological context specific. In some cells, c-MYC overexpression can
malignancies, such as leukemia, providing physicians with induce cell cycle arrest or apoptosis via p53. Therefore, it
rapid knowledge of mutations that can guide the use of tar- may require a concurrent apoptotic defect to permit c-MYC
geted therapy and risk stratify patients to various treatment overexpression.
approaches. While these panels are not yet in clinical use for BCL-2 is an oncogene that does not directly foster
lymphoma, they will likely be incorporated in the near future, increased proliferation, but rather opposes apoptosis. It does
104 Molecular Hematology

Table 8.2 Chromosomal translocations in non-Hodgkin lymphomas (NHL)

% of cases Proto- Mechanism of activation


NHL histological type Translocation involved oncogene Function of oncogene

Burkitt lymphoma t(8;14) 80% c-MYC Cell proliferation and Transcriptional deregulation
t(2;8) 15% growth
t(8;22) 5%
Diffuse large cell lymphoma der(3) 35% BCL-6 Transcriptional repressor, Transcriptional deregulation
required for germinal
center formation
Mantle cell lymphoma t(11;14) >70% Cyclin D1 Cell cycle regulator Transcriptional deregulation
(BCL-1)
Follicular lymphoma t(14;18) 90% BCL-2 Anti-apoptotic Transcriptional deregulation
Lymphoplasmacytic lymphoma t(9;14) 50% PAX-5 Transcription factor Transcriptional deregulation
regulating B cell
proliferation
Mucosa-associated lymphoid t(11;18) 50% API-2-MLT API-2 is anti-apoptotic Fusion protein
tissue (MALT) lymphoma t(1;14) rare BCL-10 ? Anti-apoptotic Transcriptional deregulation
Anaplastic large cell t(2;5) 60% in adults NPM/ALK Anaplastic lymphoma kinase Fusion protein
lymphoma 85% in children (ALK) is a tyrosine kinase

this at least in part by binding and sequestering pro-apoptotic or which have been demonstrated to have the greatest impact
BCL-2 family members, preventing them from communicat- on prognosis or treatment, are described there.
ing or executing death signals, especially at the mitochon- While these translocations are commonly seen in lym-
drion. It is classically overexpressed in follicular lymphoma phoma, and can represent the defining feature of subtypes
due to the t(14;18). This is also the most commonly occurring such as follicular lymphoma, other lymphomas are less com-
translocation to occur in conjunction with t(8;14) in double- monly associated with chromosomal translocations. GEP
hit lymphomas, an aggressive subtype of lymphoma that is and other sequencing techniques have been used to iden-
often refractory to conventional chemotherapy, occurring in tify the drivers of lymphogenesis in many of these cases.
approximately 80% of cases. For example, while lymphoplasmacytic lymphoma (LPL) was
A frequent hallmark of translocations found in B-cell previously thought to have no clear translocation of onco-
lymphomas is their exploitation of immunoglobulin gene gene, we now know that over 90% of LPLs have MYD88
regulatory elements to drive expression of an oncogene in L265P mutations. Similarly, BRAF mutations have been
a malignant B-cell or B-cell precursor. Burkitt’s lymphoma found in nearly all cases of hair cell lymphoma.
is an example of a lymphoma characterized by the overex-
pression of c-MYC. While the most common translocation
is t(8;14), which puts c-MYC under the control of the
immunoglobulin heavy chain (IgH) transcription elements, Burkitt lymphoma
the less common t(2;8) and t(8;22) are also found, putting
c-MYC transcription under the control of the light chain Burkitt lymphoma (BL) is a very high-grade B-cell malig-
κ and λ transcription elements, respectively. The t(14;18) nancy. Pathologically it is characterized by small, non-
found in follicular lymphoma drives BCL-2 expression using cleaved cells. The presence of many apoptotic malignant
IgH transcription elements. The BCL-6 expression found cells gives rise to tingible body macrophages and the “starry
in many DLCBL cases is often driven by IgH, Igκ, and Igλ sky” appearance characteristic of this and other very rapidly
elements in the t(3;14), t(2;3), and t(3;22), respectively. dividing tumors. Frequent mitotic figures also demonstrate
PAX5 expression in lymphoplasmacytic lymphoma and the rapid cell division typical of this tumor. Though rapidly
cyclin D1 expression in mantle cell lymphoma are likewise dividing, it is one of the most curable lymphomas, with
driven by the t(9;14) and t(11;14) which exploit the IgH >90% of adults enjoying long-term survival when treated
locus. Improved techniques of genetic study have allowed with a regimen similar to that proposed by MacGrath. As the
for the identification of a large number of chromosomal MacGrath regimen is quite different, and yields much
translocations, the most common of which are shown in improved results when compared to the R-CHOP regimen
Table 8.2. Those abnormalities which are the most common, that is used for other aggressive B-cell lymphomas, it is
Lymphoma genetics 105

important to make the diagnostic distinction between BL data to further subclassify DLBCLs into four groups. While
and DLBCL. this formulation does provide a useful refinement of prog-
Genetic testing plays a key role in making the diag- nosis, it still falls short of the ideal predictor: one that would
nosis. The genetic hallmark of BL is overexpression of definitively determine, prior to a particular therapy, whether
the c-MYC oncogene due to a translocation which places that therapy would work. As this scoring system was devel-
c-MYC transcription under the control of elements at an oped in the pre-rituximab era, its ability to predict outcomes
immunoglobulin locus. The most common translocation, with novel therapy remains unknown.
t(8;14), is a chromosomal rearrangement involving c-MYC While the ideal predictor of response to therapy may
and the IgH locus. Other translocations involve c-MYC not yet be available in practice, progress has been made in
with the κ(t(2;8)) or λ(t(8;22)) light chain loci. It is difficult improving prognosis using the wealth of molecular data pro-
to make the diagnosis of BL in the absence of a c-MYC vided by GEP. Two groups, one based at the US National Can-
translocation by either cytogenetics, FISH, or PCR. The cer Institute and one based at Dana-Farber Cancer Institute,
c-MYC (myelocytomatosis) oncogene is a helix-loop-helix, have published results of applying GEP to lymphoma samples
zinc finger containing transcription factor that is associated for which clinical data were available. In both cases, predic-
with a proliferative phenotype. There have been reports tors generated by GEP were able to identify new subclasses of
of lymphomas that resemble BL, though they lack MYC lymphomas and also to further refine prognosis even within
rearrangements. These lymphomas are associated by 11q IPI subgroups.
alterations, and are now recognized as a new provisional The two main molecular subgroups that define cell of
entity in the revised WHO classification of 2016. origin (COO) by GEP are GCB-like and ABC-like lym-
NGS has further improved our understanding of the phomas. There are somatic mutations common to both
underlying biology of BL. For example, 70% of sporadic and subtypes, including activating mutations of TP53, genes
40% of endemic BLs harbor mutations in TCF3, a transcrip- involved in immunosurveillance (B2M, CD58), epigenetic
tion factor that regulates the B-cell receptor and phospho- regulators (CREBBP/EP300, KMT2D/C, MEF2B), and BCL6.
inositide 3-kinase (PI3K), or their negative regulator ID3. Other mutations appear to cluster by COO. In particular,
The mechanism through which these mutations cooperate mutations affecting genes involved in histone modification
with MYC and the potential to target these mutations ther- (EZH2) and translocations involving BCL-2 are more com-
apeutically remain to be explored. mon in the GCB subtype, while genes involved in B-cell
Evidence for latent Epstein–Barr virus (EBV) infection is receptor signaling (MYD88, CD79A, CARD11, TNFAIP3)
also found in nearly all of the African endemic BL, but in only and the NF–κB pathway are common in the ABC sub-
20% of the sporadic form found outside Africa. It has been type. These classifications also have prognostic significance,
suggested that EBV plays a causative role by opposing apop- with patients with ABC subtype DLBCL having worse out-
tosis, though the exact mechanism is unknown. comes. The Hans algorithm can be used to identify these sub-
groups immunohistochemically, using antibodies to CD10,
BCL6, and IRF4/MUM1, though categorization does not
Diffuse large B-cell lymphoma exactly mirror the findings detected by GEP. Newer meth-
ods in which RNA is extracted from formalin-fixed paraffin-
DLBCLs, the most common subtype of non-Hodgkin embedded tissue have the potential to improve classification
lymphoma (NHL), are a heterogeneous group of lymphomas of COO, though they are not routinely available.
of aggressive clinical behavior. The majority likely derive As the molecular signature of DLBCL become more clearly
from follicular center cells, and roughly one-fifth of DLBCLs defined, the molecules involved in that signature can be
derive from transformation of a pre-existing follicular immediately identified as potential targets of anti-cancer
lymphoma. As their name suggests, DLBCLs have a diffuse therapy, a feat not possible when prognosis is determined by
histological pattern of large lymphoid cells. Approximately purely clinical criteria. Although the clinical significance of
two-thirds of patients with this disease will be cured many of the mutations remains unknown, there are ongoing
with combination chemotherapy and the CD20 antibody efforts to exploit these mutations with targeted therapies and
rituximab. While approximately half of patients with to tailor therapy by COO.
relapsed/refractory disease can be rescued by autologous
stem cell transplant following high-dose therapy, the remain-
ing patients have limited therapeutic options and dismal High-grade B-cell lymphomas with
outcomes. MYC and BCL2 or BCL6 translocations
In an attempt to better divide the heterogeneous group
of diseases encompassed by the label DLCBL, an IPI was While there was previously an entity known as DLBCL
created. The IPI uses just five pieces of clinical and laboratory unclassifiable, with features intermediate between DLBCL
106 Molecular Hematology

and BL, the criteria was imprecise and not uniformly used. As is characterized by a follicular pattern in the lymph node.
DLBCL unclassifiable was found to be overlap with DLBCL The appearance can be similar to that of non-malignant
with MYC and BCL-2 and/or BCL-6 translocations by mor- follicular hyperplasia. Light chain restriction can be useful
phology and GEP, a new category known as high-grade B-cell in suggesting the clonality of the tumor, which distinguishes
lymphoma with MYC and BCL2 or BCL6 translocations, also it from benign hyperplasia.
known as “double-hit” and “triple-hit” lymphomas, has been A t(14;18) translocation is found in >85% of follicular
incorporated into the WHO criteria. These patients often lymphomas. This rearrangement puts the BCL-2 gene under
present at an advanced stage and have rapidly progressive dis- the transcriptional control of elements from the igH locus.
ease that is refractory to therapy. GEP has identified a host of The BCL-2 protein functions to oppose programmed cell
mutations in epigenetic mediators, genes involved in signal- death. It is presumed that BCL-2 expression in malignancies
ing pathways, and tumor suppressors. such as follicular lymphoma permits survival of cancer cells
under conditions (cell cycle checkpoint violation, metastatic
location, genomic instability) which would otherwise trigger
Mantle cell lymphoma programmed cell death. The cloning of BCL-2 led to the
identification of a family of related proteins. While some are
Mantle cell lymphoma is a B-cell lymphoma thought to be anti-apoptotic like BCL-2, many are pro-apoptotic, but all
the malignant counterpart to the memory B cells found in function in the control of apoptosis.
the mantle zone of lymphoid follicles. It has characteristic While follicular lymphoma can be cured by local ther-
cell surface markings of CD5+, CD10−, and CD23−. While apy in very localized disease, it is more usually diagnosed in
it is classically thought of as an aggressive lymphoma, clin- an advanced stage where cure is exceedingly rare. It is gen-
ically indolent variants with low proliferation indexes have erally quite responsive to chemotherapy, but almost always
been identified. These indolent variants are often character- relapses. The clinical course is commonly marked by a series
ized by a mutated IGHV heavy chain and negative staining of chemotherapy-induced remissions followed by relapses,
for SOX11. with the interval between these decreasing over time. The end
While mantle cell lymphoma often responds to cyto- stage of the disease may be characterized by insuperable resis-
toxic chemotherapy, it has frustrated attempts at cure with tance to chemotherapy or by transformation to an aggressive
chemotherapy, though there are reports of long-term sur- large B-cell phenotype. Despite the very low cure rate, many
vivors following allogeneic stem cell transplantation. As in patients nonetheless survive for more than a decade due to
DLBCL, a mantle cell IPI score has been developed, known the indolent nature of the disease. Follicular lymphoma can
as the MIPI score, which places patients into one of three risk transform into a higher-grade lymphoma with DLBCL mor-
groups, with low-risk scores correlating with a five-year over- phology. Numerous genetic changes have been associated
all survival (OS) of 60% and high-risk scores correlating with with this transformation, including trisomy 7, loss of p53, and
an OS of approximately two years. c-MYC rearrangements.
Mantle cell lymphoma is almost uniformly characterized As outcomes in follicular lymphoma are heterogeneous, a
via classical cytogenetics or PCR by a t(11;14) which puts variety of prognostic scoring systems have been developed.
the cyclin D1 (also known as BCL-1, or B-cell leukemia/ The FLIPI score and the more recent FLIPI2 score incor-
lymphoma 1) gene under control of the igH transcription porate clinical criteria to predict OS and progression-free
control elements. Cyclin D1 binds to and activates cyclin- survival (PFS), respectively. There has also been integration
dependent kinases (CDKs). An important target of this acti- of gene mutations into a prognostic model known as the
vated CDK complex is the retinoblastoma (RB) gene product. m7-FLIPI score. This scoring system incorporates the muta-
In its hypophosphorylated state, RB inhibits entry into the S tional status of seven genes (EZH2, ARID1A, MEF2B, EP300,
phase of the cell cycle by binding the transcription factor E2F. FOXO1, CREBBP, and CARD11), along with the FLIPI score
When RB is phosphorylated, E2F is freed to activate the tran- and ECOG performance status, to create a more predictive
scription of genes which propel the cell into S phase. There- prognostic tool.
fore, overexpression of cyclin D1 acts to overcome this late
G1 phase checkpoint and maintain continuous proliferation.
Lymphoplasmacytic lymphoma

Follicular lymphoma Lymphoplasmacytic lymphoma (LPL) is an indolent lym-


phoma. The cells of this lymphoma have a phenotype that lies
Follicular lymphoma is an indolent lymphoma. The COO midway between mature lymphocytes and plasma cells, and
is thought to be the follicular center B cell. Histologically it for this reason they are often nicknamed “lymphocytes.” This
Lymphoma genetics 107

lymphoma commonly expresses IgM, which can lead to the leading to NF-κB activation. Others have shown that API2-
syndrome of Waldenstrom macroglobulinemia (WM). WM MALT 1 correlates with the nuclear location of BCL-10.
is characterized by IgM expression, hyperviscosity, bleeding, These findings suggest that these two translocations may be
Raynaud’s phenomenon, visual disturbances, and other neu- involved in activating the same pathway.
rological symptoms. Trisomy 3 is observed in 20–60% of all MALT lymphomas.
Roughly half of all LPL cases will demonstrate the t(9;14) The oncogenic properties of this numerical chromosomal
which juxtaposes the PAX5 gene and the IgH locus. PAX5 abnormality are not understood.
encodes the BSAP (B-cell specific activator protein), which
is a transcription factor. Its expression is associated with
increased expression of genes important in early B cell Anaplastic large cell lymphoma
development and decreased expression of the p53 tumor
suppressor. Anaplastic large cell lymphoma (ALCL) is characterized by
WGS of patients with WM has also identified the somatic strong surface expression of the CD30 (Ki-1) antigen, a
mutation MYD88 L265P to occur in more than 90% of cytokine receptor in the tumor necrosis factor receptor fam-
patients. Mutated MYD88 triggers tumor growth through ily. The majority of ALCLs demonstrate T cell surface mark-
the activation of NF-κB by Bruton’s tyrosine kinase (BTK). ers and/or clonal rearrangements of the T-cell receptor locus.
Mutations in MYD88 suggest susceptibility to ibrutinib, a tar- There are two main clinical forms, systemic and cutaneous.
geted inhibitor of BTK, while CXCR4 mutations alternatively The cutaneous form is particularly indolent. While it is clin-
convey resistance. ically aggressive, systemic ALCL is generally sensitive to
chemotherapy.
Approximately 50% of systemic ALCLs carry the t(2;5),
Mucosa-associated lymphoid tissue which confers good prognosis. The t(2;5)(p23;q35) results
lymphomas in a chimeric gene encoding a fusion of the nucleophosmin
(NPM) and anaplastic lymphoma kinase (ALK) proteins.
The mucosa-associated lymphoid tissue (MALT) lymphomas NPM is a multifunctional protein which has been implicated
are thought to arise from the extranodal counterpart to post- in ribosome assembly, control of centrosome duplication,
follicular memory B cells found in the marginal zone of and nuclear transport as a shuttle protein; it also possesses
lymph node follicles. These tumors are often localized, and chaperonin and ribonuclease activities. ALK is a member
their behavior is generally indolent. At least some have a of the insulin family of receptor tyrosine kinases. Its natural
dependence on continued antigen stimulation for survival, ligand is unknown. The NPM–ALK fusion contains the
as demonstrated by the prolonged complete responses that oligomerization domain of NPM and the tyrosine kinase
are seen when early-stage gastric MALT lymphoma is treated domain of ALK. It results in a self-oligomerizing, constitu-
with an antibiotic regimen to eradicate chronic helicobacter tively active tyrosine kinase with transforming properties.
pylori infection. NPM–ALK can activate numerous downstream effectors,
t(11;18)(q21;q21) is found in more than half of all low- including phospholipase C-γ PI3K, and RAS. Patients with
grade MALT lymphomas, with a preference for gastric lym- ALK mutations may also derive benefit from the targeted
phomas. The translocation is not typically found in high- inhibitor of ALK, crizotinib.
grade MALT lymphomas. API-2-MALT 1 fusion protein is
expressed from the mutant locus. API-2 (also known as
IAP-2) belongs to a family of inhibitors of apoptosis that pre- Chronic lymphocytic leukemia
vent death likely due to their direct interaction with caspases,
the proteases activated by programmed cell death. The phys- CLL is a low-grade lymphoma marked by a peripheral lym-
iological function of the MALT 1 protein is less well under- phocytosis of CD5+, CD20+, and CD23+ small lymphocytes,
stood, though it possesses a caspase-like domain at its C ter- similar in morphology to normal lymphocytes. Staging based
minus. The function of the fusion protein is unclear, though on the presence of lymphadenopathy, organomegaly, anemia,
there is some evidence that it activates NF-κB, perhaps lead- or thrombocytopenia can provide prognostic information,
ing to inhibition of apoptosis. with those cases in the best prognostic groups enjoying near
BCL-10 is overexpressed in a minority of MALT lym- normal mean survival times.
phoma cases via the t(1;14)(p22;q32), putting the coding Prognosis can also be estimated by purely molecular
region of BCL-10 under the influence of the IgH enhancer. criteria. In about half of CLL cases, VH genes exhibit
The function of this protein is unclear, but some researchers somatic hypermutation, while the others lack VH mutations.
have suggested an interaction between BCL-10 and MALT-1, CLL with IgVH genes that exhibit more than 2% somatic
108 Molecular Hematology

hypermutation, or mutated IGHV status, have significantly overexpression in CLL is rarely if ever due to a t(14;18), it is
better responses to chemoimmunotherapy and OS than the thought that the deletion of two miRNA genes, mir-15-A and
latter. mir-16-1, which downregulate BCL-2 levels, are responsible
Results of FISH cytogenetics, which detect genomic for overexpression. Nearly all CLL cases are dependent on
aberrations in over 80% of cases, also have prognostic BCL-2 for survival, which is largely due to the requirement
implications. As FISH is a directed rather than screen- for BCL-2 to tonically sequester the large amounts of the
ing technique, these abnormalities have previously been pro-death molecule BIM that are generated in CLL cells.
demonstrated by conventional banding techniques. The When BCL-2 function is abrogated, BIM is released, the
abnormalities include del 13q (50%), del 11q (18%), +12q mitochondria are permeabilized, and the cell dies. Veneto-
(16%), del 17p (7%), and del 6q (7%). Regression analysis clax, a targeted BCL-2 inhibitor, has had dramatic clinical
allowed the assignment of 90% of these cases to one of activity in patients with relapsed/refractory CLL, including
five prognostic classes based on genetic abnormalities. The those with high-risk biology, and is now a fundamental
best prognostic group included those that had 13q deletion therapeutic option for patients with CLL.
as their sole abnormality, whereas the worst prognostic
group contained those with a 17p deletion. CLL with 17p
and 11q deletions was more likely to have extensive lym-
Hodgkin lymphoma
phadenopathy, splenomegaly, cytopenias, and B symptoms.
These data raise the question of whether the classical clinical
Classical Hodgkin lymphoma (cHL), which arises from
staging, which can be used to predict survival, is partly just
germinal center or post-germinal center B cells, is char-
a surrogate for particular genetic abnormalities, and it is the
acterized histologically by small numbers of malignant
genetic abnormalities and resulting expression patterns that
Reed–Sternberg (RS) cells surrounded by an extensive but
are more important in determining prognosis.
ineffective inflammatory cell infiltrate. It has a bimodal
While there are no disease-defining mutations in CLL,
distribution, with one peak occurring in young adults and
sequencing has revealed a number of mutations that occur
another in adults of older age. While HL is curable in
at low frequency. Mutations in TP53, NOTCH1, SF3B1, and
majority of patients treated with conventional chemotherapy
BIRC3, for example, are of clinical interest because of their
with or without radiation, there is a subset of patients with
prognostic and therapeutic implications. There are ongoing
relapsed/refractory disease who are more difficult to treat.
efforts to further understand how these mutations and oth-
cHL lack surface immunoglobulin expression and B-cell
ers impact pathogenesis and whether they can be targeted
receptor signaling, and rely on alternative survival pathways,
therapeutically.
such as constitutive activation of NF-κB. Despite advances
While genetic changes in CLL have yet to result in major
in understanding the genomics of cHL, there had been few
therapeutic advances, further understanding of the signaling
therapies aimed at targeting known mutations. More recently,
pathways implicated in CLL has recently yielded a phar-
however, high-density SNP arrays with paired transcriptional
macological revolution, with the approval of several new
profiles have identified 9p24.1 amplifications, which result in
targeted agents. B-cell receptor signaling, for example, has
increased expression of PD-1 ligands, PDL1 and PDL-2, and
emerged as a driving factor in CLL survival. Downstream of
subsequently increased immune evasion. Based on this dis-
the B-cell receptor is BTK, which is essential for activating
covery, checkpoint blockade has since emerged as an effec-
additional downstream targets such as Akt, extracellular
tive therapy for cHL, with excellent responses in patients with
signal–regulated kinase (ERK), and NF-κB. BTK has also
relapsed/refractory disease.
been implicated in B cell homing and adhesion. The targeted
inhibitor of BTK, ibrutinib, has resulted in high rates of
durable responses in patients with relapsed/refractory CLL
and those with high-risk features, such as deletion 17p, lead- Conclusions
ing to its Food and Drug Administration (FDA) approval
in both the frontline and relapsed/refractory settings. PI3K, Identification of the genes involved in lymphoma pathogen-
another key component of the breakpoint cluster region esis has allowed for better characterization of the disease.
(BCR) signaling pathway, has also been identified as a This has also led the discovery of novel, targeted agents and
therapeutic target, with idelalisib, a PI3Kδ, being FDA immunotherapies which have dramatically improved out-
approved for relapsed/refractory disease in combination comes for patients. While great progress has been made, there
with rituximab. are many questions remaining about the wealth of genetic
BCL-2, which expressed at high levels in more than information we now possess, and further investigation on
70% of CLL cases, is also a target in CLL. While BCL-2 how best to utilize this information is required.
Lymphoma genetics 109

Diffuse large B-cell lymphoma


Further reading
Davis, R.E., Ngo, V.N., Lenz, G. et al. (2010). Chronic active B-cell-
Introduction receptor signalling in diffuse large B-cell lymphoma. Nature 463
(7277): 88–92.
Sehn, L.H., Donaldson, J., Chhanabhai, M. et al. (2005). Introduction International Non-Hodgkin’s Lymphoma Prognostic Factors Project
of combined CHOP plus rituximab therapy dramatically improved (1993). A predictive model for aggressive non-Hodgkin’s lymphoma.
outcome of diffuse large B-cell lymphoma in British Columbia. J. Clin. N. Engl. J. Med. 329 (14): 987–994.
Oncol. 23 (22): 5027–5033. Morin, R.D., Johnson, N.A., Severson, T.M. et al. (2010). Somatic muta-
tions altering EZH2 (Tyr641) in follicular and diffuse large B-cell
lymphomas of germinal-center origin. Nat. Genet. 42 (2): 181–185.
Techniques Morin, R.D., Mendez-Lago, M., Mungall, A.J. et al. (2011). Frequent
Alizadeh, A.A., Eisen, M.B., Davis, R.E. et al. (2000). Distinct types of mutation of histone-modifying genes in non-Hodgkin lymphoma.
diffuse large B-cell lymphoma identified by gene expression profiling. Nature 476 (7360): 298–303.
Nature 403 (6769): 503–511. Scott, D.W., Mottok, A., Ennishi, D. et al. (2015). Prognostic significance
Barrans, S.L., Crouch, S., Care, M.A. et al. (2012). Whole genome of diffuse large B-cell lymphoma cell of origin determined by digital
expression profiling based on paraffin embedded tissue can be used to gene expression in formalin-fixed paraffin-embedded tissue biopsies.
classify diffuse large B-cell lymphoma and predict clinical outcome. J. Clin. Oncol. 33 (26): 2848–2856.
Br. J. Haematol. 159 (4): 441–453. Sehn, L.H., Berry, B., Chhanabhai, M. et al. (2007). The revised inter-
Dubois, S., Viailly, P.J., Mareschal, S. et al. (2016). Next-generation national prognostic index (R-IPI) is a better predictor of outcome
sequencing in diffuse large B-cell lymphoma highlights molecular than the standard IPI for patients with diffuse large B-cell lymphoma
divergence and therapeutic opportunities: a LYSA study. Clin. Cancer treated with R-CHOP. Blood 109 (5): 1857–1861.
Res. 22 (12): 2919–2928. Shipp, M.A., Ross, K.N., Tamayo, P. et al. (2002). Diffuse large B-
Lohr, J.G., Stojanov, P., Lawrence, M.S. et al. (2012). Discovery and pri- cell lymphoma outcome prediction by gene-expression profiling and
oritization of somatic mutations in diffuse large B-cell lymphoma supervised machine learning. Nat. Med. 8 (1): 68–74.
(DLBCL) by whole-exome sequencing. Proc. Natl. Acad. Sci. U S A Teras, L.R., DeSantis, C.E., Cerhan, J.R. et al. (2016). 2016 US lymphoid
109 (10): 3879–3884. malignancy statistics by World Health Organization subtypes. CA
Cancer J. Clin. 66 (6): 443–459.
Vose, J.M., Bierman, P.J., Anderson, J.R. et al. (1992). Progressive dis-
Commonly occurring translocations in ease after high-dose therapy and autologous transplantation for lym-
lymphoma phoid malignancy: clinical course and patient follow-up. Blood 80 (8):
2142–2148.
Treon, S.P., Xu, L., Yang, G. et al. (2012). MYD88 L265P somatic muta-
tion in Waldenstrom’s macroglobulinemia. N. Engl. J. Med. 367 (9):
826–833. Mantle cell lymphoma
Hoster, E., Dreyling, M., Klapper, W. et al. (2008). A new prognostic
Burkitt lymphoma index (MIPI) for patients with advanced-stage mantle cell lymphoma.
Blood 111 (2): 558–565.
Love, C., Sun, Z., Jima, D. et al. (2012). The genetic landscape of muta-
tions in Burkitt lymphoma. Nat. Genet. 44 (12): 1321–1325.
Magrath, I., Adde, M., Shad, A. et al. (1996). Adults and children with Follicular lymphoma
small non-cleaved-cell lymphoma have a similar excellent outcome
when treated with the same chemotherapy regimen. J. Clin. Oncol. 14 Bernell, P., Jacobsson, B., Liliemark, J. et al. (1998). Gain of chromo-
(3): 925–934. some 7 marks the progression from indolent to aggressive follicle
Richter, J., Schlesner, M., Hoffmann, S. et al. (2012). Recurrent muta- centre lymphoma and is a common finding in patients with diffuse
tion of the ID3 gene in Burkitt lymphoma identified by integrated large B-cell lymphoma: a study by FISH. Br. J. Haematol. 101 (3): 487–
genome, exome and transcriptome sequencing. Nat. Genet. 44 (12): 491.
1316–1320. Certo, M., Del Gaizo Moore, V., Nishino, M. et al. (2006). Mitochondria
Salaverria, I., Martin-Guerrero, I., Wagener, R. et al. (2014). A recur- primed by death signals determine cellular addiction to antiapoptotic
rent 11q aberration pattern characterizes a subset of MYC-negative BCL-2 family members. Cancer Cell 9 (5): 351–365.
high-grade B-cell lymphomas resembling Burkitt lymphoma. Blood Federico, M., Bellei, M., Marcheselli, L. et al. (2009). Follicular lym-
123 (8): 1187–1198. phoma international prognostic index 2: a new prognostic index
Schmitz, R., Young, R.M., Ceribelli, M. et al. (2012). Burkitt lymphoma for follicular lymphoma developed by the international follicular
pathogenesis and therapeutic targets from structural and functional lymphoma prognostic factor project. J. Clin. Oncol. 27 (27): 4555–
genomics. Nature 490 (7418): 116–120. 4562.
Swerdlow, S.H., Campo, E., Pileri, S.A. et al. (2016). The 2016 revision of Lo Coco, F., Gaidano, G., Louie, D.C. et al. (1993). p53 mutations are
the World Health Organization classification of lymphoid neoplasms. associated with histologic transformation of follicular lymphoma.
Blood 127 (20): 2375–2390. Blood 82 (8): 2289–2295.
110 Molecular Hematology

McDonnell, T.J., Deane, N., Platt, F.M. et al. (1989). bcl-2- Vose, J., Armitage, J., Weisenburger, D., and International TCLP (2008).
immunoglobulin transgenic mice demonstrate extended B cell International peripheral T-cell and natural killer/T-cell lymphoma
survival and follicular lymphoproliferation. Cell 57 (1): 79–88. study: pathology findings and clinical outcomes. J. Clin. Oncol. 26
Pastore, A., Jurinovic, V., Kridel, R. et al. (2015). Integration of gene (25): 4124–4130.
mutations in risk prognostication for patients receiving first-line
immunochemotherapy for follicular lymphoma: a retrospective anal-
Chronic lymphocytic leukemia
ysis of a prospective clinical trial and validation in a population-based
registry. Lancet Oncol. 16 (9): 1111–1122. Byrd, J.C., O’Brien, S., and James, D.F. (2013). Ibrutinib in relapsed
Solal-Celigny, P., Roy, P., Colombat, P. et al. (2004). Follicular lymphoma chronic lymphocytic leukemia. N. Engl. J. Med. 369 (13): 1278–1279.
international prognostic index. Blood 104 (5): 1258–1265. Chiaretti, S., Marinelli, M., Del Giudice, I. et al. (2014). NOTCH1,
Tsujimoto, Y., Yunis, J., Onorato-Showe, L. et al. (1984). Molecular SF3B1, BIRC3 and TP53 mutations in patients with chronic lympho-
cloning of the chromosomal breakpoint of B-cell lymphomas and cytic leukemia undergoing first-line treatment: correlation with bio-
leukemias with the t(11;14) chromosome translocation. Science 224 logical parameters and response to treatment. Leuk. Lymphoma 55
(4656): 1403–1406. (12): 2785–2792.
Cimmino, A., Calin, G.A., Fabbri, M. et al. (2005). miR-15 and miR-16
Lymphoplasmacytic lymphoma induce apoptosis by targeting BCL2. Proc. Natl. Acad. Sci. U S A 102
(39): 13944–13949.
Iida, S., Rao, P.H., Nallasivam, P. et al. (1996). The t(9;14)(p13;q32) chro- Crombie, J. and Davids, M.S. (2017). IGHV mutational status testing in
mosomal translocation associated with lymphoplasmacytoid lym- chronic lymphocytic leukemia. Am. J. Hematol. 92 (12): 1393–1397.
phoma involves the PAX-5 gene. Blood 88 (11): 4110–4117. Damle, R.N., Wasil, T., Fais, F. et al. (1999). Ig V gene mutation status
Treon, S.P., Tripsas, C.K., Meid, K. et al. (2015). Ibrutinib in previously and CD38 expression as novel prognostic indicators in chronic lym-
treated Waldenstrom’s macroglobulinemia. N. Engl. J. Med. 372 (15): phocytic leukemia. Blood 94 (6): 1840–1847.
1430–1440. Del Gaizo Moore, V., Brown, J.R., Certo, M. et al. (2007). Chronic
lymphocytic leukemia requires BCL2 to sequester prodeath BIM,
explaining sensitivity to BCL2 antagonist ABT-737. J. Clin. Invest. 117
Mucosa-associated lymphoid tissue
(1): 112–121.
lymphomas
Dohner, H., Stilgenbauer, S., Benner, A. et al. (2000). Genomic aberra-
Lucas, P.C., Yonezumi, M., Inohara, N. et al. (2001). Bcl10 and MALT1, tions and survival in chronic lymphocytic leukemia. N. Engl. J. Med.
independent targets of chromosomal translocation in MALT lym- 343 (26): 1910–1916.
phoma, cooperate in a novel NF-kappa B signaling pathway. Furman, R.R., Sharman, J.P., Coutre, S.E. et al. (2014). Idelalisib and rit-
J. Biolumin. Chemilumin. 276 (22): 19012–19019. uximab in relapsed chronic lymphocytic leukemia. N. Engl. J. Med.
Maes, B., Demunter, A., Peeters, B., and De Wolf-Peeters, C. (2002). 370 (11): 997–1007.
BCL10 mutation does not represent an important pathogenic mecha- Hamblin, T.J., Davis, Z., Gardiner, A. et al. (1999). Unmutated Ig V(H)
nism in gastric MALT-type lymphoma, and the presence of the API2- genes are associated with a more aggressive form of chronic lympho-
MLT fusion is associated with aberrant nuclear BCL10 expression. cytic leukemia. Blood 94 (6): 1848–1854.
Blood 99 (4): 1398–1404. Roberts, A.W., Davids, M.S., Pagel, J.M. et al. (2016). Targeting BCL2
Uren, A.G., O’Rourke, K., Aravind, L.A. et al. (2000). Identification of with venetoclax in relapsed chronic lymphocytic leukemia. N. Engl.
paracaspases and metacaspases: two ancient families of caspase-like J. Med. 374 (4): 311–322.
proteins, one of which plays a key role in MALT lymphoma. Mol. Cell Wang, L., Lawrence, M.S., Wan, Y. et al. (2011). SF3B1 and other novel
6 (4): 961–967. cancer genes in chronic lymphocytic leukemia. N. Engl. J. Med. 365
(26): 2497–2506.

Anaplastic large cell lymphoma


Hodgkin lymphoma
Gambacorti-Passerini, C., Messa, C., and Pogliani, E.M. (2011). Crizo-
tinib in anaplastic large-cell lymphoma. N. Engl. J. Med. 364 (8): 775– Ansell, S.M., Lesokhin, A.M., Borrello, I. et al. (2015). PD-1 block-
776. ade with nivolumab in relapsed or refractory Hodgkin’s lymphoma.
Kutok, J.L. and Aster, J.C. (2002). Molecular biology of anaplastic lym- N. Engl. J. Med. 372 (4): 311–319.
phoma kinase-positive anaplastic large-cell lymphoma. J. Clin. Oncol. Green, M.R., Monti, S., Rodig, S.J. et al. (2010). Integrative analysis
20 (17): 3691–3702. reveals selective 9p24.1 amplification, increased PD-1 ligand expres-
Morris, S.W., Kirstein, M.N., Valentine, M.B. et al. (1994). Fusion of sion, and further induction via JAK2 in nodular sclerosing Hodgkin
a kinase gene, ALK, to a nucleolar protein gene, NPM, in non- lymphoma and primary mediastinal large B-cell lymphoma. Blood
Hodgkin’s lymphoma. Science 263 (5151): 1281–1284. 116 (17): 3268–3277.
Chapter 9 The molecular biology of chronic
lymphocytic leukemia
John G. Gribben
Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK

Introduction and definition, 111 BCL2 family member expression in CLL, 116
CLL pathogenesis, 111 The CLL microenvironment, 116
Epidemiology, 111 CLL and impaired immune responses, 116
CLL cell of origin, 112 Prognostic factors and their clinical utility, 117
Stereotype receptors, 113 Richter transformation, 118
Molecular genetics of CLL, 113 Conclusions, 119
The B-cell receptor and B-cell signaling, 115 Further reading, 119

signaling and the BCL2 inhibitor venetoclax. The activity of


Introduction and definition agents targeting these pathways provides strong evidence for
their importance in the pathogenesis and maintenance of this
Chronic lymphocytic leukemia (CLL) is characterized by
disease.
the clonal expansion of small mature-appearing malignant B
cells with a distinct immunophenotype, with co-expression
of CD19, CD5, and CD23, coupled with low levels of CD20 CLL pathogenesis
and surface immunoglobulins. This immunophenotype is
different from any normal B cell, making it more difficult The molecular pathogenesis of CLL is complex and incom-
to determine the cell of origin of this malignancy. Although pletely understood. It appears to be a complex interaction of
CLL often has an indolent clinical course, there is great het- genetic predisposition, poorly understood cancer-initiating
erogeneity in its clinical behavior, with some patients rapidly events in mature B cells, and the subsequent acquisition of a
progressing to need for therapy, while other patients live long variety of genetic and epigenetic changes, shaped by interac-
term with the disease with few symptoms and might never tion with the host microenvironment (Figure 9.1).
require treatment.
Research in CLL has identified a large number of prognos-
Epidemiology
tic factors that can be used to help identify those patients who
are at high risk for disease progression. Because of its indo-
CLL is the most common leukemia in the Western world,
lent course, it was previously felt that CLL cells resembled
but its incidence varies between individuals in different geo-
and arose from long-lived resting lymphocytes, but this has
graphical regions, and ranges from 0.06% of individuals in
been challenged by heavy water labeling experiments demon-
Europe and the USA to less than 0.01% of individuals in
strating that CLL often contains fractions of cells with rapid
eastern Asia. The risk of developing CLL is almost twice
turnover. This disease has therefore become a paradigm for
as high for men than for women. The risk of developing
the concept of a cancer living in a near-equilibrium of cells
CLL increases with age, with the median age at diagnosis of
driven by proliferation through the B-cell receptor (BCR)
around 72 years.
and ongoing cell death, driven by the expression of both
pro- and anti-apoptotic members of the BCL2 family of pro-
Hereditary factors
teins. An understanding of the pathophysiology of this dis-
ease has led to great advances in its treatment by the approval The finding that strong geographical variability does not alter
of agents such as ibrutinib and acalabrutinib targeting BCR in patients of Asian origin who live in Europe or the USA

Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.

111
112 Molecular Hematology

Leukemia
initiating Microenvironment Secondary
event interactions lesion

Disease progression
Genetic Initiation Promotion/accumulation/ Chemorefractoriness
predisposition loss of immune control Richter transformation
Polygenic del13q Signaling pathways TP53
IRF4 +12 BCR NOTCH1
IRF8 NF-kB SF3B1
BCL2 TLR BIRC3
miR-15/16 CD38 ATM
VLA-4 Integrins MYC
NOTCH1 CDKN2A
CXCR4
Fig. 9.1 Chronic lymphocytic leukemia (CLL) pathogenesis and progression. CLL arises in the setting of polygenic predisposition and this
differs among geographical locations. Leukemia-initiating genetic events permit longer survival of B cells, which leads to subsequent accumulation,
resulting in subclonal genetic complexity.

suggests that genetic and not environmental factors are can increase the risk of CLL, and no evidence that dietary or
important. This is further supported by the familial prepon- lifestyle factors increase risk.
derance also seen in CLL. First-degree relatives of patients
with CLL have an 8.5-fold elevated risk of developing this dis-
ease, and the concordance of CLL is higher among monozy-
gotic than dizygotic twins. Genome-wide association studies
CLL cell of origin
in familial CLL have shown that this susceptibility is poly-
CLL can be divided into two major subsets, distinguished
genic, and single-nucleotide polymorphisms (SNPs) have
by whether CLL cells express immunoglobulin heavy chain
been identified in over 30 loci associated with familial CLL,
variable (IGHV) genes which have undergone somatic hyper-
suggesting that common genetic variations contribute to her-
mutation in their variable regions. Those cases that have less
itable risk and that these genetic variations differ in diverse
then 98% homology with germline are designated mutated
populations around the world. A number of these loci occur
chronic lymphocytic leukemia (M-CLL), and those that have
at sites of genes implicated in CLL pathogenesis. For exam-
98% or higher are designated unmutated chronic lympho-
ple, CLL-associated SNPs have been found at the BCL2 locus
cytic leukemia (UM-CLL). These are felt to reflect the stage
and also at IRF4, leading to reduced expression of interferon
of normal B cell differentiation from which the CLL cells
regulatory factor 4, implicated in Notch signaling. An SNP
originate.
associated with reduced expression of miR-15a and miR16-
At diagnosis, some 65% of CLL cases have mutated
1 is associated with familial CLL, and reduced expression of
IGHV, which are therefore proposed to arise from post–
these microRNA allows for increased expression of BCL2 and
germinal center B cells expressing immunoglobulin that has
ZAP70, proteins that confer increased resistance to cell death
undergone somatic hypermutation and, in some cases, also
or enhanced BCR-mediated signaling.
immunoglobulin isotype switching (Figure 9.2). The other 35
of cases express unmutated IGHV, suggesting origin from a
Environmental factors
B cell that has not undergone germinal center–driven differ-
Pesticide exposure might be a risk factor for CLL, and the US entiation. Patients with CLL cells with mutated IGHV typi-
Department of Veterans Affairs has accepted that exposure cally have a more indolent disease course than CLL patients
to Agent Orange is a risk factor for the disease. There is little whose cells express unmutated IGHV. Some CLL cases have
evidence to suggest that ionizing radiation or viral infections been described that are similar to unmutated IGHV CLL, but
The molecular biology of chronic lymphocytic leukemia 113

Germinal center

Selection
Low/lost affinity
Apoptosis

CD5+ Proliferation Class-switched


B Cell and SHM Improved affinity FDC Memory B cell
Class
switching

TH Cell
Dark Zone Light Zone IgM+IgD+
Constrained Memory
SHM ? B cell
IGHV-mutated
Ig+ CLL

IGHV-mutated
IgM+IgD+ CLL

CLL with limited SHM


IGHV-unmutated CLL
(e.g. IGHV3-21/IGLV3-21)
Fig. 9.2 Chronic lymphocytic leukemia (CLL) cell of origin. A CD5+ B cell enters the germinal center and undergoes a process of affinity
maturation and class switching. Leukemogenesis events arising in cells at different stages within this pathway explain the mutated and unmutated
subtypes of CLL seen. SHM, somatic hypermutation.

originate from B cells with limited somatic mutation, such as well-characterized genetic alterations, including chromoso-
those encoded by mutated IGHV3-21. mal alterations, mutations, alterations in miRNA expression,
and epigenetic modifications.

Stereotype receptors
Chromosomal alterations
The repertoire of immunoglobulin (Ig) molecules produced More than 80% of CLL patients carry at least one of four
by CLL cells is considerably more limited than the reper- common chromosomal alterations. The most common is a
toire of Ig molecules in normal B cells, reflecting biased deletion in chromosome 13q14.3 (del13q), occurring in more
use of IGHV genes that have restricted somatic mutation than 50% of patients and associated with a good progno-
and limited junctional and heavy–light chain combinatorial sis. Deletion in chromosome 11 (del11q) is found in 18%
diversity. As many as 30% of patients have CLL cells that of patients, often associated with alterations in ATM, which
express Ig “stereotypes,” and more than 1% of cases have encodes a protein involved in DNA repair. Del 11q is asso-
near-identity in their primary structure in the variable region ciated with more rapid disease progression, but has a good
among patients. This limited Ig diversity provides compelling response to modern therapy. Trisomy 12 is found in 16%
evidence that CLL B cells are selected by the binding activity of patients with CLL and is associated with an intermediate
of their expressed surface Ig, suggesting that B-cell receptor prognosis. Deletion in chromosome 17 (del17p) is found in
signaling plays a critical role in CLL pathogenesis. 5–7% of patients, is associated with loss of the tumor suppres-
sor gene TP53, and has the worst prognostic impact.

Molecular genetics of CLL


Somatic mutations
Unlike other B-cell malignancies, chromosomal translo- The application of whole-exome sequencing (WES) and
cations are rare in CLL, but there are a number of whole-genome sequencing (WGS) has transformed our
114 Molecular Hematology

* Q < 0.05 • Q < 0.10 Subclonal-low Subclonal-high Clonal % cases based on mutations:
25
* 36%
86 14% (unmutated)
20
18%
(clonal +
22% subclonal)
15 •
% cases

10%
51 *
43
10 36 •
*
27 27 27 *
22 20 *
19 18 17 17
5 15 15 14 *
11 *
7 6 6 6 5 5 3 3 2 2 2
0
1

AF

F1
53

M 4
M 5
R2

X1

AS
ZN E

AS
92

X
6

1
M

O1

3
7
T1

GA

ZM 2
OR

2
88

3
1
3B

F
I

S1
HL

AF

M
RC
XW

1
X3

ND

YM
KB
CH

AT

NX

IR
TP

F2

EG

BR
PO

DT

NR
KR

ED
YD
XP

BC

PI
SF

RP

TR
KL
BI

DD
NF

CC
FB
T
NO

Fig. 9.3 Frequency of genetic events seen in chronic lymphocytic leukemia and demonstrated in a word cloud.
Source: Modified from Landau, D.A., Tausch, E., Taylor-Weiner, A.N. et al. (2015). Mutations driving CLL and their evolution in progression and
relapse. Nature 526(7574): 525–530.

understanding of CLL pathophysiology, and sequences of or del17p, are found in only a fraction of the CLL cells and
more than 500 cases have been reported to date. CLL is one must therefore represent subclonal events which occur later
of the tumors with the lowest background mutational load in disease evolution. These subclonal driver mutations are
(0.6 per Mb). It is genetically heterogeneous and there are associated with more aggressive disease, particularly when
no unifying or diagnostic gene mutations (Figure 9.3). The two or more mutations are found concurrently. Single-cell
most frequently identified mutations were found in SF3B1 analysis is being used to determine whether these events
(21%), ATM (15%), TP53 (7%), NOTCH1 (6%), and BIRC3 are occurring in the same cells or whether there are dif-
(4%). Recurrent somatic mutations have been observed in ferent clonal subpopulations, and whether mutations occur-
genes that have a role in mRNA processing (SF3B1 and ring within the same cell are required to affect prognosis.
XPO1), DNA damage (TP53 and ATM), Notch signaling Of considerable interest are studies examining the impact of
(NOTCH1), B-cell signaling (EGR2 or BRAF), chromatin chemotherapy and how this can induce large clonal shifts due
modification (HIST1H1E, CHD2 and ZMYM3), Wnt signal- to increases in the proportions of CLL cells that have muta-
ing, and inflammatory pathways (MYD88). tions in genes conferring resistance to therapy, notable TP53
Next-generation sequencing has also revealed consider- mutation or del17p.
able intratumoral heterogeneity in CLL. Some somatic muta-
tions, such as those in MYD88, or chromosomal abnormal-
ities, such as trisomy 12 or del13q, are usually found in
MicroRNA alterations
almost all CLL cells in any one patient, suggesting that these CLL was the first human disease found to be associated
genetic alterations occurred early in disease evolution. Other with alterations in microRNA (miRNA), with the identifi-
mutations, notable those found in SF3B1 or NOTCH1, TP53, cation that miR-15a and miR-16-1 are deleted, altered, or
The molecular biology of chronic lymphocytic leukemia 115

downregulated in up to 60% of cases of CLL and are dysfunc- the data suggest that CLL cells derive from a continuum of B
tional in some cases of familial CLL. Reduced expression or cell maturation states.
loss of miR-15a and miR-16-1 can enhance the expression
of target genes such as BCL2 and MCL1, which encode
anti-apoptotic proteins of the BCL2 family. A number of
other miRNAs are dysregulated or differentially expressed The B-cell receptor and B-cell signaling
in subgroups with distinctive biological features, including
miR-29a/b, miR-29c, miR-34b, miR-181b, and miR-3676, CLL cells typically co-express low levels of expression of sur-
which target TCL1A. Notably, there is transgenic expression face IgM and IgD, and relatively few cases of CLL express sur-
of TCL1 in B cells which promotes the development of face IgG. The BCR in CLL comprises a heterodimer of the
CLL in mice. Increased expression of miR-155 is associated transmembrane Ig molecule of IgM or IgD and the signal-
with enhanced BCR signaling, B cell proliferation, and ing components CD79A (Igα) and CD79B (Igβ). Functional
lymphomagenesis. BCR is required for CLL cell survival. Crosslinking of surface
Ig phosphorylates the immunoreceptor tyrosine-based acti-
vation motifs of CD79A and CD79B molecules and triggers
BCR-mediated signaling. The Ig of CLL B cells most likely
Epigenetic changes
engages with auto-antigens, leading to constitutive BCR sig-
CLL cells exhibit global hypomethylation combined with naling in vivo (Figure 9.4).
local hypermethylation. Methylation profiling has demon- Enhanced BCR-mediated signaling is more commonly
strated substantial heterogeneity of intratumoral methyla- observed in UM-CLL, whereas an anergic response pre-
tion. Increasing methylation heterogeneity is associated with dominates in M-CLL, perhaps accounting for the more
increased genetic complexity, likely due to the acquisition indolent behavior of M-CLL cases. BCR signaling also has
of subclonal mutations. There is therefore a link between impacts on other signaling pathways, including integrins and
genetic and epigenetic evolution in CLL. UM-CLL and M- chemokines. The chemokine receptor CXCR4 is downmodu-
CLL cells have distinctive methylation patterns, similar to lated by BCR signaling, altering CLL cell adhesion and migra-
those of pre–germinal center versus post–germinal center tion. This intrinsic dependence on BCR-mediated signaling
memory B cells, providing further evidence for the cell of ori- explains the great sensitivity of CLL to BCR inhibitors such as
gin, as shown in Figure 9.2. However, when methylation sig- Bruton’s tyrosine kinase (BTK) or phosphoinositide 3-kinase
natures are used to classify distinct clinical CLL subgroups, (PI3K) inhibitors. Indeed, the success of treatment with the

Antigen
BCR

CD79 Extracellular

A B PIP 2 PIP 3 Intracellular


AKT
P P PI3K δ
P mTOR
P S P S P
Y Y BTK P
LYN K LYN K
P P
P PLC γ2

PKC β
Idelalisib
IKK

Ibrutinib NF- K B
Acalabrutinib

Fig. 9.4 Simplified B-cell receptor (BCR) signaling pathways and inhibitors of the pathway available to treat chronic lymphocytic leukemia.
116 Molecular Hematology

BTK inhibitor ibrutinib provides perhaps the strongest evi- venetoclax with or without anti-CD20 monoclonal antibody,
dence for the importance of BCR-mediated signaling in the and with or without ibrutinib, targeting both the BCR and
survival of CLL cells, and has activity in CLL cells with BCL2 pathways to provide perhaps the most effective treat-
TP53 abnormalities that do not respond well to conventional ments currently available.
chemotherapy.

The CLL microenvironment


BCL2 family member expression in CLL
CLL depends upon survival signals that the cells receive
The BCL2 protein family consists of at least 30 related pro- in the tumor microenvironment. Within lymph nodes,
teins characterized by the expression of BCL2 homology CLL form “proliferation centers,” where they interact with
domains. BCL2 family members are divided into three dif- non-malignant stromal cells, CLL-associated macrophages
ferent subclasses based on structural and functional fea- (Nurse-like cells), T cells, and mesenchymal-derived stromal
tures. Pro-survival or anti-apoptotic proteins include BCL2, cells, among others (Figure 9.6). Within proliferation centers,
BCL-XL, and MCL1 proteins, which are all overexpressed all CLL cells are exposed to chemokines, integrins, cytokines,
in CLL and are upregulated by contact of CLL cells with and survival factors including BAFF and APRIL, which acti-
the tumor microenvironment. The mechanism of expression vate canonical NF-κB, and induce the expression of miR-155,
of BCL2 in CLL likely involves miRNA-mediated changes thereby enhancing BCR-mediated signaling. Cytokines such
in the malignant cells, and the promoter region of CBCL2 as IL-4 upregulate surface IgM, potentially facilitating inter-
is hypomethylated in CLL, leading to increased expression. actions of the CLL cell with auto-antigens. Notch or Hedge-
The pro-apoptotic family members include BAX and BAD, hog signaling provides pro-survival stimulation, particularly
and the BH3-only protein members include BID, BIM, BAD, in those cases with trisomy 12.
NOVA, and PUMA. CLL cells express increased levels of both
pro- and anti-apoptotic proteins, so that these cells exist in a
complex relationship which is now able to be exploited in the CLL and impaired immune responses
clinic (Figure 9.5).
Venetoclax is a small-molecule BH3 mimetic that inhibits CLL is associated with significant immune suppression and
BCL-2. This drug is highly potent in inducing apoptosis in infectious complications are a major cause of morbidity
CLL cells, by diminishing the capacity of BCL-2 to sequester and mortality in this disease. An important aspect of CLL
BIM. Venetoclax is effective in patients with relapsed or is the development of hypogammaglobulinemia, with the
refractory disease and in patients with relapsed disease and consequent increased risk of infection and poor response to
TP53 mutations or loss. Studies are examining the use of vaccination. The mechanism involved is unclear, but likely

Pro-apoptotic Venetoclax
BCL-2 protein
BCL-2
Apoptosis
initiation
Mi
toc

Pro-apoptotic
h

BIM
on

BAX
protein
dri
on

BAK
BAX
Cancer cell survival Cancer cell death

Activation
of caspases
Cytochrome c

BCL-2 overexpression allows Venetoclax binds selectively to BCL-2,


cancer cells to evade apoptosis by freeing pro-apoptotic proteins that initiate
sequestering pro-apoptotic proteins. programmed cell death (apoptosis).
Fig. 9.5 BCL2 family member expression in chronic lymphocytic leukemia and impact of sequestering BCL2 by venetoclax.
The molecular biology of chronic lymphocytic leukemia 117

Antigen

IL-10 CCR1/3
CCL3/4

CCR4
IL-4 Nurse-like cell
T cell CD267 (TACI) LAM
CD269 (BCMA)

CC
CD268 (BAFF-R) FF)

L
(BA L)

17
CCL slg 7
5 I
CD2 6 (APR

/22
3/4
2 5 1
CCR1/3 CD CD3
CD40L
CD40 IL-4

CCL19 CCR7 Cell CD38


B-cell CXCL13
CCL21 5
CXCR
CD38
CD44 CXCL12

4
CR
HA VLA-4
CX
Frz
HEV ROR2 ROR1
12
CXCL
VCA Wnt
M-1 Wnt5a

Strom
al cell

Fig. 9.6 Interactions with chronic lymphocytic leukemia (CLL) cells within the microenvironment. HEV, high endothelial venules; LAM,
lymphoma-associated macrophage.

represents replacement of CLL cells for normal B cells within genetic, molecular, and biochemical characteristics of the
the immune microenvironment, as well as impairment of CLL cells. Multivariable models, prognostic indices, and
T cell help. CLL cells express high levels of programmed nomograms have been developed to consolidate the prog-
cell death 1 ligand 1 (PD-L1) and programmed cell death 1 nostic factors. Commonly used parameters associated with
ligand 2 (PD-L2), which suppress the effector responses of poorer outcome are late-stage disease at initial presen-
T cells expressing programmed cell death protein 1 (PD-1) tation, male sex, increased age, poor performance status
and other immune checkpoint inhibitors, leading to an (largely due to co-morbidities), and CLL cell characteristics,
“exhausted” T cell and NK cell phenotype and impaired including unmutated IGHV, increased expression of ZAP-70,
cellular immune function. CD49d, or CD38, high serum beta-2-microglobulin, and/or
the presence of complex karyotype, del11q, del17p, or TP53
mutations.
Prognostic factors and their The most reliable prognostic models are those developed
clinical utility to predict the interval from diagnosis to time of first treat-
ment. There is also increasing interest in predictive models
The clinical course of newly diagnosed CLL is extremely vari- to define outcome with specific types of therapy. Prognos-
able, with some patients being rapidly symptomatic or devel- tic markers that help predict response to specific treatments
oping high-risk disease, which requires treatment soon after are termed predictive biomarkers. TP53 and IGHV muta-
diagnosis, while others remain free of symptoms for decades. tional status are powerful predictive biomarkers for response
Prognostic factors that can be used to identify patients to chemoimmunotherapy. These markers are often specific
who require earlier treatment include clinical features, and for the type of treatment being investigated, and both of them
118 Molecular Hematology

Diagnosis Relapse Refractoriness

Chemotherapy Chemotherapy

TP53 mutated
CLL cell

Small TP53 mutated Chemotherapy Subsequent expansion Fig. 9.7 Molecular mechanism of chemoresistance in
subclone mixed with removes TP53 wt of the TP53 mutated clone chronic lymphocytic leukemia. Chemotherapy selects
TP53 wt clones clones and selects for expansion of subclones, such as TP53 mutant cells,
for the TP53 mutated with change in subclonal architecture over time and after
subclone chemotherapy.

lose their predictive value when treatment with ibrutinib which virtually always express IGHV with somatic mutations
or venetoclax is used. One of the most powerful predictive as a post–germinal center malignancy.
biomarkers has been the emergence of del17p to TP53 muta- RT cells have other distinctive genetic differences from de
tions (Figure 9.7). These changes are relatively rare at diagno- novo DLBCL. About 60% of RT lymphomas have inactivating
sis, but recent WES data has suggested that small subclones mutations and/or deletions in TP53, often with deregulation
harboring these changes might more frequently be present of MYC, which is observed in around 40% of cases. MYC
at low frequency. Subsequent treatment with chemotherapy deregulation is usually caused by translocations juxtaposing
removes the sensitive CLL cells, leaving behind and enriching MYC to Ig loci, gene amplification of MYC at 8q24, or
for cells with mutations conferring resistance to chemother- somatic mutations affecting MYC transregulatory factors,
apy, so that over time the disease becomes more refractory to such as NOTCH1, which is mutated in around 30% of cases.
treatment. NOTCH1 mutations are largely mutually exclusive with MYC
Treatment options are changing, and novel agents, notably oncogenic activation, consistent with the observation that
BCR-signaling inhibitors and BCL2 inhibitors, are highly NOTCH1 directly stimulates MYC transcription and sug-
effective agents that have activity among patients who would gesting that activation of oncogenic MYC may be a common
have been considered high risk when the only option was pathway in RS transformation. CDKN2A encodes the cell
conventional chemotherapy. These agents both have activity cycle progression inhibitor p16, and is mutated or deleted
in TP53 mutated cells. Ongoing clinical research is investigat- in roughly 30–50% of cases, but this rarely occurs in CLL or
ing whether earlier use of these agents will prevent the emer- de novo DLBCL. The high prevalence of TP53 disruption in
gence of chemorefractory clones. RS reflects the selection of a chemorefractory clone under
the pressure of previous CLL treatments, thus suggesting an
explanation for the poor outcome and the limited sensitivity
Richter transformation to conventional drugs. On the other hand, RT lymphomas
typically do not have mutations in a number of genes which
Richter transformation (RT) represents the clinicopatholog- are commonly seen in de novo DLBCL, including those
ical transformation of CLL into aggressive lymphoma, most encoding proteins involved in nuclear factor-κB signaling or
commonly diffuse large B-cell lymphoma (DLBCL), or more in transcriptional repressors such PRDM1/BLIMP1 or BCL6.
rarely Hodgkin lymphoma (Figure 9.8). About 7% of CLL Up to 20% of the DLBCL-type Richter transformation
patients develop RT, with an incidence rate of approximately and ∼50% of Hodgkin lymphoma–type Richter transforma-
0.5% per year. In over half the patients with RT, the lym- tion have IGHV rearrangements that differ from that of the
phoma cells with Richter transformation do not express CD5 original CLL clone, suggesting that these lymphomas repre-
or CD23, which are almost invariably expressed by CLL cells. sent a de novo secondary malignancy; some of these seem
The DLBCL-like lymphomas in RT most often share the same to be associated with Epstein–Barr virus infection, particu-
IGHV-D-J rearrangement as the original CLL clone and as larly in patients with severe disease-related immune dysfunc-
such may express unmutated IGHV, unlike de novo DLBCLs, tion and/or treatment-related immune suppression. Cases
The molecular biology of chronic lymphocytic leukemia 119

(a) DLBCL
NOTCH1
MYC
mutations
translocation
amplification
MGA
mutations
MYC
activation
CDKN2A
loss

Transformation

CLL

TP53 Richter
disruption transformation

(b)
Clonally related
Richter
80%
CLL V4-39 D6 J4

V4-39 D6 J4

Clonally unrelated
Richter
V4-34 D2-2J3

20%

Fig. 9.8 Molecular mechanisms of Richter transformation in chronic lymphocytic leukemia (CLL). (a) Changes in TP53, activation of MYC,
and loss of CDKN2A are among the most frequent changes seen with diffuse large B-cell lymphoma (DLBCL) transformation in CLL. (b)
Representation of clonally related and de novo clonally unrelated DLBCL arising in CLL.

with unrelated IGHV rearrangements have better response to response to conventional chemotherapy. These novel agents
chemotherapy and improved outcome. targeting these pathways are very well tolerated and suit-
able for more elderly patients with CLL. With increased sur-
vival in CLL with these new agents, Richter transformation is
Conclusions emerging as a major unmet need in CLL, and identification
of agents that will improve the outcome of these patients is
Advances in our understanding of CLL pathophysiology help the focus of much ongoing research.
explain the clinical heterogeneity that we see in this disease.
With the identification of pathways involved in the develop-
ment of CLL, an increasing array of prognostic and predic-
Further reading
tive biomarkers are increasingly used in clinical practice. The
Burger, J.A., Tedeschi, A., Barr, P.M. et al. (2015). Ibrutinib as initial
importance of BCR signaling in driving CLL cell prolifera-
therapy for patients with chronic lymphocytic leukemia. N. Engl. J.
tion and BCL2 overexpression to prevent apoptosis explains Med. 373 (25): 2425–2437.
the long survival of CLL cells. Identification of the impor- Byrd, J.C., Brown, J.R., O’Brien, S. et al. (2014). Ibrutinib versus ofatu-
tance of BCR signaling and BCL2 pathways to CLL survival mumab in previously treated chronic lymphoid leukemia. N. Engl. J.
has led to the development of the most effective agents that Med. 371 (3): 213–223.
we have in this disease, and these novel agents are able to Fischer, K., Bahlo, J., Fink, A.M. et al. (2016). Long-term remissions
overcome the poor prognostic markers that predict poor after FCR chemoimmunotherapy in previously untreated patients
120 Molecular Hematology

with CLL: updated results of the CLL8 trial. Blood 127 (2): 208– Landau, D.A., Tausch, E., Taylor-Weiner, A.N. et al. (2015). Mutations
215. driving CLL and their evolution in progression and relapse. Nature
Hallek, M., Cheson, B.D., Catovsky, D. et al. (2018). iwCLL guidelines 526 (7574): 525–530.
for diagnosis, indications for treatment, response assessment, and Stilgenbauer, S., Eichhorst, B., Schetelig, J. et al. (2018). Venetoclax for
supportive management of CLL. Blood 131 (25): 2745–2760. patients with chronic lymphocytic leukemia with 17p deletion: results
Landau, D.A., Sun, C., Rosebrock, D. et al. (2017). The evolutionary from the full population of a phase II pivotal trial. J. Clin. Oncol. 36
landscape of chronic lymphocytic leukemia treated with ibrutinib (19): 1973–1980.
targeted therapy. Nat. Commun. 8: 2185.
Chapter 10 The molecular biology of
multiple myeloma
Wee Joo Chng1,2 & P. Leif Bergsagel3
1
National University Cancer Institute, National University Health System of Singapore, Singapore
2
University of Singapore, National University Hospital, Singapore
3 Division of Hematology-Oncology, Comprehensive Cancer Center, Mayo Clinic Arizona, Scottsdale AZ, USA

Introduction, 121 Universal cyclin D dysregulation, 123


A plasmablast/plasma cell tumor of post–germinal center B cells, 121 Molecular classification of MM, 124
Stages of plasma cell neoplasms, 121 Prognostic and therapeutic implications of molecular classifications, 124
Immunoglobulin translocations present in the majority of MM Possible events mediating transformation of MGUS to MM, 125
tumors, 122 Model of molecular pathogenesis of MM, 126
Marked karyotypic complexity in MM, 122 Critical role of the bone marrow microenvironment and novel
Chromosome content associated with at least two different pathogenic therapeutic strategies targeting the bone marrow milieu, 127
pathways, 122 Conclusion, 128
Recurrent IgH translocations and recurrent trisomies representing Further reading, 128
primary oncogenic events, 123

long-lived plasma cells, including a strong dependence


Introduction on the bone marrow microenvironment for survival and
growth. In contrast to normal long-lived plasma cells, MGUS
Multiple myeloma (MM) is an incurable post–germinal cen-
and MM tumors retain some potential for an extremely low
ter B-cell malignancy. In 2018, it is estimated that in the
rate of proliferation, usually with no more than a few percent
USA 30 770 new cases will have been diagnosed, with 12 770
of cycling cells until advanced stages of MM.
patients succumbing to the disease. It is almost always pre-
ceded by a pre-malignant tumor called monoclonal gam-
mopathy of undetermined significance (MGUS), which is the
most common lymphoid tumor in humans, occurring in Stages of plasma cell neoplasms
approximately 4% of individuals over the age of 50. The
prevalence of both MM and MGUS increases with age, Based on the current diagnostic criteria proposed by the
and is about twofold higher in African Americans than in International Myeloma Working Group, four stages of
Caucasians, although the rate of progression from MGUS to plasma cell neoplasms can be identified based on the
MM is similar in these two populations. presence of myeloma-defining events (hypercalcemia, renal
impairment, anemia, bone lesions, >60% bone marrow
plasma cells, involved/uninvolved free light chain ratio
>100, more than one focal lesion on magnetic resonance
A plasmablast/plasma cell tumor of imaging [MRI]), level of bone marrow plasma cell infil-
post–germinal center B cells tration and serum or urine monoclonal immunoglobu-
lins, and extramedullary involvement. The stages include
Post–germinal center B cells that have undergone productive MGUS, smoldering multiple myeloma (SMM), symptomatic
somatic hypermutation, antigen selection, and immunoglob- myeloma, and plasmacytoma (PCT) (Table 10.1). MGUS can
ulin heavy chain (IgH) switching can generate plasmablasts, progress sporadically to MM expressing the same mono-
which typically migrate to the bone marrow, where the clonal immunoglobulins with a probability of about 0.6–3%
microenvironment enables differentiation into long-lived per year.
plasma cells. Importantly, MGUS and MM are monoclonal Through the analysis of several large prospective cohort
tumors that are phenotypically similar to plasmablasts/ studies, three predictive factors for progression of MGUS
to MM were identified, including M-protein greater than
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. 15 g l−1 , IgM or IgA M-protein, and presence of an abnormal
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. free light chain ratio. In the presence of all three risk factors,

121
122 Molecular Hematology

Table 10.1 Features used to diagnose monoclonal gammopathy of undetermined significance (MGUS), smoldering myeloma (SMM), and mul-
tiple myeloma (MM)

Serum Bone marrow plasma


M-protein cells (BMPC) Myeloma-defining events

MGUS <30 g l−1 <10% None


SMM ≥30 g l−1 10–60% None
MM Any ≥10% or PCT Hypercalcemia, anemia, renal impairment, bone lesions, >60% BMPC, involved/
uninvolved free light chain ratio >100, >1 focal lesion on magnetic resonance imaging
PCT Any <10% None, with biopsy-proven plasmacytoma (PCT)

the risk of progression at 20 years is 58%, compared with 5% MGUS or SMM, 55–70% for intramedullary MM, 85% in
in the absence of any of these risk factors. The risk of pro- plasma cell leukemia (PCL), and above 90% in HMCLs. Lim-
gression to MM is higher for SMM than for MGUS, with a ited studies indicate that IgL translocations are present in
median time to progression ranging from one to five years. about 10% of MGUS/SMM tumors, and in about 15–20%
In one study, M-protein in excess of 30 g l−1 , presence of IgA of intramedullary MM tumors and HMCLs. Translocations
subtype, and urinary M-protein excretion above 30 g l−1 were involving an Igκ locus are rare, occurring in only 1–2% of
factors associated with early progression to MM. MM tumors and HMCLs.
Extramedullary MM is a more aggressive tumor that
can present as secondary or primary plasma cell leukemia,
depending on whether or not preceding intramedullary MM Marked karyotypic complexity in MM
has been recognized. Human multiple myeloma cell lines
(HMCLs), which are presumed to include most oncogenic The karyotypes of MM are characterized by complex abnor-
events involved in tumor initiation and progression of the malities, both structural and numerical. Numerical chro-
corresponding tumor, have been generated mainly from a mosomal abnormalities are present in virtually all MM
subset of extramedullary MM tumors. tumors and most, if not all, MGUS tumors. There is non-
random involvement of different chromosomes in different
myeloma tumors, and often heterogeneity among cells within
Immunoglobulin translocations a tumor. The mechanism underlying this karyotypic instabil-
present in the majority of MM tumors ity is not fully understood. Centrosome abnormalities, one of
the mechanisms mediating chromosomal instability in solid
Like other post–germinal center B cell tumors, translocations tumors, have been identified in MGUS and about one-third
involving the IgH locus (14q32) or one of the immunoglob- of MM tumors. However, mutations of genes involved in the
ulin lambda (IgL) loci (κ, 2p12 or λ, 22q11) are common. mitotic spindle checkpoint, another mechanism leading to
In general, these events are mediated by errors in one of the genomic instability in solid tumors, have not been identified
three B-cell-specific DNA modification mechanisms: VDJ in MM. It is thought that karyotypic complexity increases
recombination, IgH switch recombination, or somatic hyper- during tumor progression, although karyotypic progression
mutation. With rare exceptions, these translocations result has not been well documented.
in dysregulated or increased expression of an oncogene that
is positioned near one or more of the strong immunoglob-
ulin enhancers on the translocated derivative chromosome Chromosome content associated with
14. However, translocations involving an IgH switch region at least two different pathogenic
uniquely dissociate the intronic from one or both 3′ IgH pathways
enhancers, so that an oncogene might be juxtaposed to an
IgH enhancer on either or both of the derivative chromo- There is a clear consensus that chromosome content reflects
somes, as first demonstrated for FGFR3 on der(14) and at least two pathways of pathogenesis. Approximately half of
WHSC1 on der(4) in MM. tumors are hyperdiploid (HRD; 48–75 chromosomes), and
These IgH translocations are efficiently detected by fluo- typically have multiple trisomies involving chromosomes 3,
rescence in situ hybridization (FISH) analyses. Large stud- 5, 7, 9, 11, 15, 19, and 21, but only infrequently (<10%) have
ies from several groups show that the prevalence of IgH one of the recurrent IgH translocations. Non-hyperdiploid
translocations increases with disease stage: about 50% in (NHRD) tumors (<48 and/or >75 chromosomes) usually
The molecular biology of multiple myeloma 123

(∼70%) have one of the recurrent IgH translocations. These


Table 10.2 Recurrent primary immunoglobulin heavy chain (IgH)
patterns of genetic aberration are already present in MGUS, translocation partners in MM
suggesting that the HRD/non-HRD dichotomy is estab-
lished early during disease pathogenesis, probably at dis- Genomic Class of gene
ease initiation, and is dictated by initial oncogenic events. Gene partner locus dysregulated Frequency
Tumors that have a t(11;14) translocation may represent
a distinct category of NHRD tumors, as they are often CCND1 11q13 CCND 15%
diploid or pseudodiploid, sometimes with this transloca- CCND2 12p13 CCND <1%
CCND3 6p21 CCND 2%
tion as the only karyotypic abnormality detected by conven-
MAF 16q23 MAF 5%
tional cytogenetics. This turns out to be true for a propor-
MAFB 21q12 MAF 2%
tion of t(11;14) MM even when assessed by high-resolution MAFC 8q24.3 MAF <1%
aCGH. MMSET/FGFR3 4p16 MMSET and 15%
In contrast to the selective occurrence of recurrent IgH usually FGFR3
translocations in NHRD tumors, other genetic events –
17p loss or TP53 (gene product p53) mutations, RAS
mutations, secondary immunoglobulin translocations, MYC have not been adequately confirmed as essential for mainte-
translocations – often occur with a similar prevalence in nance of the tumor and/or as therapeutic targets.
HRD and NHRD tumors. Extramedullary MM tumors and In HRD MM, the recurrent trisomies of chromosomes 3,
HMCLs nearly always have an NHRD phenotype, consistent 5, 7, 9, 11, 15, 19, and 21 appear to be the earliest events.
with the hypothesis that HRD tumors are more stromal In a karyotypic analysis of a large number of HRD myelo-
cell dependent than NHRD tumors. We have virtually no mas, the presence of these trisomies is associated with the
information about the timing, mechanism, or molecular simplest karyotypes, often occurring as the only abnormal-
consequences of hyperdiploidy. We do not know if the extra ities, suggesting that they are the earliest acquired genetic
chromosomes are accumulated one at a time in sequential abnormalities in these tumors. The pattern of acquisition of
steps, or as one catastrophic event. For tumors that are HRD these chromosomal trisomies seems to be random. Further-
but have one of the recurrent translocations – most often more, FISH studies have shown that this pattern of trisomies
t(4;14) – we do not know if hyperdiploidy occurred before already exists in MGUS, therefore representing early events.
or after the translocation.

Universal cyclin D dysregulation


Recurrent IgH translocations and
recurrent trisomies representing Analysis of gene expression data suggests that almost all
primary oncogenic events cases of plasma cell neoplasm starting from the MGUS stage
express one or more of the cyclin D genes in an aberrant fash-
There are seven recurrent chromosomal partners and onco- ion. Therefore, it has been proposed that dysregulation of a
genes that are involved in IgH translocations in approxi- cyclin D gene provides a unifying, early oncogenic event in
mately 40% of MM tumors. These translocations lead to the MGUS and MM. MGUS and MM appear closer to normal,
overexpression of three classes of genes: cyclin D, MAF, and non-proliferating plasma cells than to normal proliferating
MMSET/FGFR3 (Table 10.2). The recurrent translocation plasmablasts, for which 30% or more of the cells can be in
breakpoints usually occur within or near switch regions, but S phase, yet the expression level of cyclin D1, cyclin D2, or
sometimes within or near VDJ sequences, suggesting that cyclin D3 mRNA in MM and MGUS is distinctly higher than
these translocations are mediated by errors in IgH switch in normal plasma cells, in fact at a level comparable to that
recombination or somatic hypermutation. Since there is no of cyclin D2 mRNA expressed in normal proliferating plas-
evidence that IgH switch recombination or somatic hyper- mablasts. About 25% of MM tumors have an IgH translo-
mutation mechanisms are active in normal plasma cells or cation that directly dysregulates a cyclin D gene or a MAF
plasma cell tumors, it is presumed that these translocations gene encoding a transcription factor that markedly upregu-
usually represent primary, perhaps initiating, oncogenic lates cyclin D2.
events as normal B cells pass through germinal centers. In Although MM tumors with t(4;14) express moderately
addition, the most common of these translocations, t(4;14), high levels of cyclin D2, the cause of increased cyclin
t(11;14), and t(14;16), are already detected in MGUS, further D2 expression remains unknown. Whereas normal bone
highlighting their early role in disease pathogenesis. With the marrow plasma cells express little or no detectable cyclin D1,
exception of FGFR3 (especially with an activating mutation) the majority of HRD tumors express cyclin D1 biallelically,
and possibly MAF, the consequences of these translocations while most other tumors express increased levels of cyclin
124 Molecular Hematology

D2 compared with normal bone marrow plasma cells, may be somewhat misleading, since the HY group, which is
both by unknown mechanism. Only a few percent of MM about 28% of MM tumors, includes only about 60% of HRD
tumors do not express increased levels of a cyclin D gene tumors. The distribution of the remaining HRD tumors
compared with normal plasma cells, but many of these among the other six groups has not been clarified, although
tumors appear to represent samples that are substantially most are probably in the PR group, defined by increased
contaminated by normal cells, and another large fraction expression of proliferation-related genes; and the LB group,
of these tumors express little or no RB1, eliminating the defined by low bone disease and lower expression of genes
necessity of expressing a cyclin D gene. associated with bone disease in MM, such as FRZB and
DKK1.
There is significant overlap between the TC classification
Molecular classification of MM and the UAMS molecular classification. When TC classes are
assigned to the 414 MM cases assigned a UAMS molecular
The advent of genomic technologies has facilitated the global classification in the original dataset, the MS and MF groups
examination of copy number, translocation, expression, and correspond to the 4p16 and Maf groups, respectively, with
mutation in a highly parallel fashion. The use of such high- 100% concordance. The CD-1 and CD-2 groups together cor-
dimensional data has allowed the identification of novel respond to the 11q13 and 6p21 groups (88% concordance).
disease subtypes. In myeloma, the use of supervised and The HY group contains most of the TC D1 and D1 + D2 cases
unsupervised methods has led to the development of dif- (96% concordance). The bulk of the TC D2 cases (67%) fall
ferent molecular classifications. Interestingly, although the within the LB group. The PR group contains a mix of patients
two approaches are different, there are significant overlaps from the different TC classes: 13% 4p16, 4% Maf, 9% 11q13,
between them, with the subcategories anchored by known 21% D1, 19% D1 + D2, 28% D2, and 6% None. The PR group
recurrent primary genetic abnormalities, providing strong is therefore enriched for CCND2-expressing tumors, consis-
validation that the primary genetic events drive most of the tent with the hypothesis that these tumors are more aggres-
subsequent changes in cellular transcription in MM. sive. The additional insights gained from the incorporation of
One of the first of these classifications, the translo- next-generation sequencing has been primarily to fill in the
cation/cyclin D (TC) classification, takes the supervised details of the basic outline provided by the TC and UAMS
approach and is based on the spiked expression of genes classifications, and are outlined below.
deregulated by primary IgH translocations, and the univer-
sal overexpression of cyclin D genes either by these translo-
cations or via other mechanisms. The resultant classification Prognostic and therapeutic
identifies eight groups of tumors: those with primary translo- implications of molecular
cations (designated 4p16, 11q13, 6p21, Maf), those that over- classifications
express CCND1 and CCND2 either alone or in combination
(designated D1, D1 + D2, D2), and the rare cases that do not The intrinsic properties of the tumor cell are informative in
overexpress any cyclin D genes (designated “None”). Most of predicting the prognosis and the response to existing ther-
the patients with HRD MM fall within the D1 and D1 + D2 apies. In particular, studies over recent years have clearly
groups. This classification system therefore focuses on the established the importance of genetic abnormalities in prog-
different kinds of mechanism that dysregulate a cyclin D gene nosis. For example, it has been well documented that an
as an early and unifying event in pathogenesis. unfavorable outcome is associated with each of the follow-
There are several different unsupervised approaches, with ing: hypodiploidy compared with hyperdiploidy; 17p13 dele-
the best known developed at the University of Arkansas for tion, p53 mutation, t(4;14), and t(14;16). A recent large study
Medical Sciences (UAMS), which identified seven tumor from the Intergroupe Francophone du Myélome group has
groups characterized by the co-expression of unique gene confirmed that t(4;14) and 17p13 deletion are independent
clusters. Interestingly, these clusters also identify tumors prognostic factors in newly diagnosed MM patients under-
with t(4;14), MAF translocations, t(11;14), and t(6;14), going high-dose therapy with stem cell transplantation, and
corresponding to the MS, MF, and CD-1 and/or CD-2 can further dissect each of the International Staging System
groups, respectively. In this analysis, t(11;14) and t(6;14) can (ISS) categories. Furthermore, it established that most of the
belong to either the CD-1 or CD-2 group, depending on the prognostic impact of chromosome 13 deletion is due to the
expression of CD20 and other B-cell-related genes. This is concurrent presence of t(4;14), as patients with chromosome
consistent with the finding that t(11;14) and t(6;14) have very 13 deletion without t(4;14) do not have adverse prognosis.
similar expression profiles, clinical profiles, and outcome. In In patients treated with stem cell transplantation, the poor
contrast to the TC classification, the UAMS classification prognosis associated with t(4;14) appears to be due to early
identifies HRD MM as a distinct group, HY. However, this disease relapse. These patients derive little benefit from stem
The molecular biology of multiple myeloma 125

cell transplantation and alternative therapeutic strategies are RB pathway can also be dysregulated in MM. Inactivation of
needed. With the addition of sequencing information it has CDKN2C (p18), a critical gene for normal plasma cell devel-
now become clear that the poor prognosis associated with opment, is likely to contribute to increased proliferation.
del17p is only seen in patients who have concomitant inacti- There is biallelic deletion of p18 in 30% of HMCLs and in
vation of p53, either through mutation or homozygous dele- nearly 10% of tumors in the highest quintile of proliferation,
tion of the other allele. Biallelic p53 inactivation is seen at as determined by an expression-based proliferation index.
diagnosis in 4% of patients, and identifies an ultra-high-risk Forced expression of CDKN2C by retroviral infection of
population with a median overall survival of less than two HMCLs that express little or no endogenous p18 substan-
years. tially inhibits proliferation. Paradoxically, about 60% of
Recent studies have shown that treatment with bortezomib HMCLs and 60% of the more proliferative MM tumors have
overcomes the deleterious impact of chromosome 13 dele- increased expression of p18 compared with normal plasma
tion on prognosis. This has led to a proposal for risk-stratified cells. There is evidence that the E2F transcription factor,
management akin to the management of acute leukemias, which is upregulated in association with increased prolif-
where genetically defined high-risk patients will be treated eration, increases the expression of p18, presumably as a
with an upfront bortezomib- or lenalidomide-based regimen, feedback mechanism. Apart from the lack of a functional RB1
whereas standard-risk patients will receive upfront high-dose protein in approximately 10% of HMCLs, the mechanism(s)
therapy, with stem cell transplantation after induction ther- by which most HMCLs and proliferative tumors become
apy. In addition, the genes or pathways deregulated by the insensitive to increased p18 levels is not yet understood.
specific genetic events could be potential novel therapeutic
targets. This is best exemplified by the clinical development
Activating RAS pathway mutations
of FGFR3 inhibitor therapy for MM with t(4;14).
As the gene expression–based classifications have a strong An emerging genetic difference between MGUS and MM is
genetic basis, they could easily identify these high-risk activating RAS pathway mutation. There is a high prevalence
genetic groups, for example the 4p16 and MAF groups in of activating mutations of N- or K-RAS (40%), BRAF (7%),
the TC classification and the PR, MS, and MF groups in the and FGFR3 (2% of all patients, but 17% of t(4;14)) in newly
UAMS classification, and identify the clinically important diagnosed MM tumors, with only a small increase occur-
molecular subtypes of myeloma. In addition, new insights ring during tumor progression. Sequencing reveals that the
provided by these classifications could be exploited thera- mutations are often subclonal, and subclones with different
peutically. For example, the underlying cyclin D deregula- mutations of the pathway frequently coexist. The prevalence
tion potentially has an important therapeutic implication, as is similar in HMCLs. Importantly, less than 5% of MGUS
differential targeting of cyclin D may be very useful and add tumors have RAS mutations, consistent with the hypothesis
specificity to treatment. Indeed, some potential agents target- that RAS mutations may mark, if not mediate, the MGUS to
ing cyclin D2 have been identified in a drug library screen. MM transition.

MYC activation
Possible events mediating
transformation of MGUS to MM One of the major differences in gene expression that has been
identified between MGUS and MM is the increased expres-
Very few differences between MGUS and MM have been sion of MYC and its target genes in MM. Genomic stud-
identified at the genetic and molecular levels. All the pri- ies have shown that there are frequent chromosome translo-
mary genetic events, the recurrent IgH translocations and tri- cations involving the MYC locus, resulting in increased
somies of HRD MM, are already present in MGUS. By impli- expression of MYC, in about 40% of newly diagnosed MM.
cation, additional genetic events are therefore required for a There are diverse partner loci involved in the translocations,
transformation to MM. with one-third involving an immunoglobulin locus (most
frequently the heavy chain locus). The other two-thirds
involve plasma cell-specific super enhancers (e.g. FAM46C,
Loss of cell cycle control
TXNDC5, FOXO3) from elsewhere in the genome. Many
Chromosome 13 deletion is one of the most common genetic of the rearrangements are cryptic, explaining why they were
abnormalities in MM and is also found in MGUS at appar- not detected previously. They are also unevenly distributed
ently similar frequency. There does not appear to be any across primary genetic subtypes, with a low of 25% in t(11;14)
role for haploinsufficiency of RB1, and inactivation of the MM, and a high of 55% in HRD MM.
remaining allele is uncommon (1–2%), but when it develops The biological validity of MYC in the progression of
it is associated with poor outcome. Other components of the MGUS to MM is provided by an MM mouse model,
126 Molecular Hematology

which utilizes activation-induced cytidine deaminase (AID)- 40% of PCL and HMCLs. As noted above, the poor prognosis
dependent somatic hypermutation to activate the MYC onco- associated with del17 is entirely explained by a concomitant
gene sporadically. In mice with a genetic predisposition to inactivation of p53. There is no prognostic impact of del17p
developing monoclonal gammopathy (but not a control stain in patients with intact p53.
without this predisposition), the sporadic activation of MYC
led to the development of MM. MYC has been shown to be
Gain of chromosome 1q and loss of
directly involved in DNA replication, binding and activat-
chromosome 1p
ing DNA replicative origins and regulating S-phase transition
during the cell cycle through a non-transcriptional mech- A number of laboratories have determined, using a com-
anism. Therefore, in MM, activation of MYC may cooper- bination of FISH, array comparative genomic hybridization
ate with cyclin D/RB deregulation – which negates the early (aCGH), and gene expression profiling, that there is a gain
G1 S cell cycle checkpoint that allows the establishment of of sequences, and corresponding increased gene expression,
the initial limited clonal plasma cell expansion (MGUS) – to at 1q21 in 30–40% of tumors. These gains are concentrated
overcome remaining cell cycle constraints, leading to further substantially in those tumors that have t(4;14) or t(14;16),
clonal expansion and transformation to MM. or have a high proliferation expression index. It has been
proposed that the increased proliferation in tumors with a
gain of 1q21 sequences is due to the increased expression
NFkB pathway
of CKS1B as a result of increased copy number. One might
It has long been felt that activation of the NFκB pathway is expect to find other mechanisms, such as localized amplifi-
important in the pathogenesis of MM, but little is known cation or a translocation, if increased CKS1B expression is a
about the prevalence of NFκB activation or mechanisms that cause of increased proliferation, but there is no evidence for
cause it. An array of mutations that result in the constitu- other mechanisms to increase CKS1B expression. Further-
tive activation of the NFκB pathway have been identified in more, CKS1B expression correlates closely with the expres-
about 20% of patient samples and about 50% of HMCLs. sion of a number of proliferation genes, and therefore appears
The most common event is inactivating mutations of TRAF3 to be a consequence rather than a cause of the proliferation.
in 13% of patients. In addition, inactivating mutations of So, it seems prudent to remain skeptical that CKS1B is the
TRAF2, BIRC2/BIR3 (gene product cIAP1/2), and CYLD gene targeted by the gain of 1q21 sequences. Furthermore,
were identified. Chromosome translocations and amplifi- amplification of chromosome 1q tends to involve the whole q
cations resulting in activation of NFκB-inducing kinase arm, and on aCGH the minimally deleted areas do not always
(NIK), CD40, LTBR, TACI, NFκB1, and NFκB2 were also include CKS1B.
reported. Chromosome 1p loss is also common, occurring in about
Although activation of both the canonical and non- 46% of patients when analyzed by aCGH and in 7–36% in
canonical pathways is seen, the preponderance of mutations previous karyotype studies. Unlike 1q gain, 1p loss is usually
result most directly in increased processing of NFκB2 p100 to interstitial and involves fragments of varying length. Chro-
p52 (i.e. activation of the non-canonical pathway). Depletion mosome 1p loss has been shown to be a marker of poor prog-
of NIK with shRNAs directed against NIK results in inhibi- nosis in cytogenetic studies.
tion of both the classical and alternative NFκB pathways, and
also growth inhibition. Half of primary MM tumors have an
expression signature of NFκB target genes, with activating Model of molecular pathogenesis
mutations identified in less than half of these patients. Pre- of MM
sumably either other mutations or ligand-dependent interac-
tions in the bone marrow microenvironment are responsible Based on the results just summarized, a model for the molec-
for the NFκB activation in the remaining patients. ular pathogenesis of MM has been proposed. Chromosome
content appears to identify two different, but perhaps over-
lapping, pathways of pathogenesis: NHRD tumors and HRD
Abnormalities of TP53 gene and
tumors. In about 40% of tumors, a primary chromosome
chromosome 17p loss
translocation results in the dysregulated expression of an
Mutations of TP53 are relatively rare in newly diagnosed oncogene. These primary genetic events lead to cyclin D
MM, occurring in approximately 5% of tumors. However, dysregulation, either directly – cyclin D1 in t(11;14) and
the frequency of mutations appears to increase with disease cyclin D3 in t(6;14) – or indirectly – cyclin D2 in t(4;14) and
stage, and is about 30% in PCL and 65% in HMCLs. Deletion maf translocations – or by an as yet unknown mechanism –
(mainly monoallelic) of the TP53 locus, as detected by inter- cyclin D1 and/or D2 in HRD MM. The dysregulation of one
phase FISH, occurs in about 10% of MM and approximately of three cyclin D genes may render the cells more susceptible
The molecular biology of multiple myeloma 127

Table 10.3 Well-characterized genetic subtypes of multiple myeloma

Heavy Light RAS 13 TRAF3


Genetics Ploidy Prognosis chain chain Morphology CD20 mutation deletion mutation Bone CCND

t(11;14) NHRD/ Good G κ Lymphoplasmacytoid ++ ++ −/+ + ++ D1


t(6;14) diploid D3
t(4;14) NHRD Poor A λ Plasmablastic − − +++ ++ −/+ D2
t(14;16) NHRD Poor A λ Plasmablastic + − ++ −/+ −/+ D2
t(8;14)
t(14;20)
Trisomies HRD Good G κ Normal − ++ −/+ −/+ ++ D1

HRD, hyperdiploid; NHRD, non-hyperdiploid.

to proliferative stimuli, resulting in selective expansion as marrow microenvironment include mutation of TP53 or
a result of interaction with bone marrow stromal cells that deletion of the TP53 locus on chromosome 17p13, and com-
produce Interleukin 6 (IL-6) and other cytokines. These plex secondary translocations. In the future, it would be
primary genetic events, primary IgH translocations and important to further interrogate how these pathways inter-
trisomies of chromosomes 3, 5, 7, 9, 11, 15, 19, and 21, are act with each other, and differentiate the critical from non-
already present at the earliest identified stage of tumorigen- critical pathways during tumorigenesis that should be tar-
esis, and define subtypes of MM with unique clinical (bone geted therapeutically.
disease, heavy chain subtypes, prognosis), molecular (types
of cyclin D expressed, associated mutations), pathological
(morphology, CD expression), and cytogenetic (ploidy) Critical role of the bone marrow
features (Table 10.3). microenvironment and novel
A second genetic “hit” leading to subsequent transfor- therapeutic strategies targeting the
mation from MGUS to MM may be mediated by activation bone marrow milieu
of MYC or further deregulation of the RB pathway. The
MYC pathway may be activated by RAS mutation, IgH-MYC As already mentioned, different molecular pathways can be
translocations, or other as yet unidentified mechanisms. activated in the malignant plasma cells by intrinsic or extrin-
The RB pathway may be further deregulated by RB hap- sic mechanisms, either through microenvironmental sig-
loinsufficiency or p18 loss. These mechanisms are not nals derived from cytokines or via interaction between the
mutually exclusive and the pattern of cooperation is still not tumor cells and accessory cells in the bone marrow microen-
yet understood. There appears to be enrichment for some vironment, such as stromal cells, endothelial cells, osteo-
pathway deregulation, depending on primary genetic abnor- clasts, and osteoblasts. Direct contact between MM cells
malities. For example, tumors with cyclin D2 overexpression and cells of the bone marrow milieu induce paracrine or
are significantly enriched for chromosome 13 deletion and autocrine release of cytokines and growth factors such as IL-
hence RB haploinsufficiency, whereas RAS mutations are 6, insulin-like growth factor, hepatocyte growth factor, and
significantly associated with t(11;14) tumors. NFκB appears vascular endothelial growth factor, which provide further
to be an alternate pathway, probably important for providing proliferative and survival signals by activating intracellular
the growth and survival signals for maintenance of tumor pathways such as PI3K/AKT/mTOR/p70S6K, IKK-α/NFκB,
cell survival, growth, and proliferation. It can be activated RAS/RAF/MAPK, and JAK/STAT3.
through external signals from the tumor microenvironment, These interactions and the activated pathways not only
such as B cell activating factor (BAFF), or signaling triggered provide growth and survival signals, but also result in the
by contact between the malignant plasma cells and the uncoupling of new bone formation from bone resorption,
accessory cells in the bone marrow microenvironment. leading to the formation of lytic bone lesions and resis-
Alternative mechanisms intrinsic to the tumor cells such tance of the tumor cells to different chemotherapeutic agents.
as gene mutations may also activate these pathways (e.g. The importance of the bone marrow microenvironment
mutations in the NFκB pathway). in MM biology is highlighted by the spate of new drugs
Later events that may lead to more aggressive tumor char- being developed to target these cytokines/growth factors or
acteristics and emancipation from dependence on the bone contact-mediated pathways (Table 10.4). The Food and Drug
128 Molecular Hematology

by the multistage accumulation of genetic abnormalities


Table 10.4 Therapeutic compounds targeting the bone marrow
microenvironment
deregulating different pathways. Much of this knowledge
is already being utilized for diagnosis, prognosis, and risk
stratification of patients. Importantly, from a clinical stand-
Target pathway-activated therapy
point, this knowledge has led to the development of novel
therapeutic strategies, some of which are already in clinical
Targeting growth factors and cytokines
use, and many others showing promise in pre-clinical and
IL-6 Ras/Raf/Mek/Erk IL-6 antibodies
early clinical studies.
JAK2/STAT3 IL-6R antibodies
PI3K/AKT IL-6 superantagonist
IGF-1 Ras/Raf/Mek/Erk IGF1R kinase inhibitor
NFκB IGF1R antibodies Further reading
PI3K/AKT IGF-1 ligand antibodies
VEGF Ras/Raf/Mek/Erk VEGFR tyrosine kinase
General
inhibitor
β-Catenin Wnt PKF115-584 Chng, W.J., Glebov, O., Bergsagel, P.L. et al. (2007). Genetic events in the
DKK1 Wnt Anti-DKK1 neutralizing pathogenesis of multiple myeloma. Best Pract. Res. Clin. Haematol.
antibodies 20: 571–596.
Targeting interactions between multiple myeloma plasma cells Dispenzieri, A., Katzmann, J.A., Kyle, R.A. et al. (2010). Prevalence
and accessory cells and risk of progression of light-chain monoclonal gammopathy of
Adhesion molecules Proteasome inhibitors undetermined significance: a retrospective population-based cohort
Cell adherence Proteasome inhibitors, study. Lancet 375: 1721–1728.
immunomodulatory drugs Kawano, Y., Moschetta, M., Manier, S. et al. (2015). Targeting the bone
(IMiDs) marrow microenvironment in multiple myeloma. Immunol. Rev. 263:
Intracellular signaling molecules 160–172.
PI3K PI3 kinase inhibitor Kuehl, W.M. and Bergsagel, P.L. (2012). Molecular pathogenesis of mul-
AKT Perifosine tiple myeloma and its premalignant precursor. J. Clin. Invest. 122:
mTOR Rapamycin, CCI-779 3456–3463.
MEK/ERK MEK1/2 inhibitor Kyle, R.A., Larson, D.R., Therneau, T.M. et al. (2018). Long-term follow-
BCL2/ BCL2 inhibitor (venetoclax) up of monoclonal gammopathy of undetermined significance. N.
MCL1 In clinical trials Engl. J. Med. 378: 241–249.
Morgan, G.J., Walker, B.A., and Davies, F.E. (2012). The genetic archi-
Surface molecules
tecture of multiple myeloma. Nat. Rev. Cancer 12: 335–348.
CD38 Daratumumab, isatuximab,
Nair, S., Branagan, A.R., Liu, J. et al. (2016). Clonal immunoglobulin
CAR-T, bispecifics
against lysolipids in the origin of myeloma. N. Engl. J. Med. 374: 555–
SLAMF7 Elotuzumab
561.
BCMA GSK2857915, multiple
Siegel, R.L., Miller, K.D., and Jemal, A. (2018). Cancer statistics, 2018.
CAR-T, and bispecific
CA Cancer J. Clin. 68: 7–30.
antibodies
Weiss, B.M., Abadie, J., Verma, P. et al. (2009). A monoclonal gammopa-
FcRH5 Monoclonal antibodies,
thy precedes multiple myeloma in most patients. Blood 113: 5418–
bispecifics, CAR-T
5422.
IGF, insulin-like growth factor; PI3K, phosphatidylinositol 3-kinase;
VEGF, vascular endothelial growth factor.
Genetics
Bolli, N., Avet-Loiseau, H., Wedge, D.C. et al. (2014). Heterogeneity of
Administration–approved drugs thalidomide, bortezomib, genomic evolution and mutational profiles in multiple myeloma. Nat.
and lenalidomide are all thought to exert their therapeutic Commun. 5: 2997.
effect at least partly through targeting the microenvironment. Fonseca, R., Debes-Marun, C.S., Picken, E.B. et al. (2003). The recur-
rent IgH translocations are highly associated with nonhyperdiploid
variant multiple myeloma. Blood 102: 2562–2567.
Hurt, E.M., Wiestner, A., Rosenwald, A. et al. (2004). Overexpression
Conclusion of c-maf is a frequent oncogenic event in multiple myeloma that pro-
motes proliferation and pathological interactions with bone marrow
Significant progress has been made in understanding the stroma. Cancer Cell 5: 191–199.
molecular pathogenesis and biology of MM. Oncogenic Zingone, A., Cultraro, C.M., Shin, D.-M. et al. (2010). Ectopic expres-
pathways can be activated through cell-intrinsic or -extrinsic sion of wild-type FGFR3 cooperates with MYC to accelerate devel-
mechanisms. Similar to other cancers, MM is characterized opment of B-cell lineage neoplasms. Leukemia 24: 1171–1178.
The molecular biology of multiple myeloma 129

Secondary genetic events MYC


Annunziata, C.M., Davis, R.E., Demchenko, Y. et al. (2007). Frequent Affer, M., Chesi, M., Chen, W.D. et al. (2014). Promiscuous MYC locus
engagement of the classical and alternative NF-kappaB pathways by rearrangements hijack enhancers but mostly super-enhancers to dys-
diverse genetic abnormalities in multiple myeloma. Cancer Cell 12: regulate MYC expression in multiple myeloma. Leukemia 28: 1725–
115–130. 1735.
Boyd, K.D., Ross, F.M., Walker, B.A. et al. (2011). Mapping of chro- Chesi, M., Robbiani, D.F., Sebag, M. et al. (2008). AID-dependent acti-
mosome 1p deletions in myeloma identifies FAM46C at 1p12 and vation of a MYC transgene induces multiple myeloma in a conditional
CDKN2C at 1p32.3 as being genes in regions associated with adverse mouse model of post-germinal center malignancies. Cancer Cell 13:
survival. Clin. Cancer Res. 17: 7776–7784. 167–180.
Chiecchio, L., Dagrada, G.P., Ibrahim, A.H. et al. (2009). Timing of Chng, W.J., Huang, G.F., Chung, T.H. et al. (2011). Clinical and biolog-
acquisition of deletion 13 in plasma cell dyscrasias is dependent on ical implications of MYC activation: a common difference between
genetic context. Haematologica 94: 1708–1713. MGUS and newly diagnosed multiple myeloma. Leukemia 25: 1026–
Flores-Montero, J., de Tute, R., Paiva, B. et al. (2016). Immunophenotype 1035.
of normal vs. myeloma plasma cells: toward antibody panel specifi- Dib, A., Gabrea, A., Glebov, O.K. et al. (2008). Characterization of MYC
cations for MRD detection in multiple myeloma. Cytometry B Clin. translocations in multiple myeloma cell lines. J. Natl. Cancer Inst. 39:
Cytom. 90: 61–72. 25–31.
Halliley, J.L., Tipton, C.M., Liesveld, J. et al. (2015). Long-lived plasma Gabrea, A., Martelli, M.L., Qi, Y. et al. (2008). Secondary genomic rear-
cells are contained within the CD19(−)CD38(hi)CD138(+) subset in rangements involving immunoglobulin or MYC loci show similar
human bone marrow. Immunity 43: 132–145. prevalences in hyperdiploid and nonhyperdiploid myeloma tumors.
Jourdan, M., Cren, M., Robert, N. et al. (2014). IL-6 supports the Genes Chromosomes Cancer 47: 573–590.
generation of human long-lived plasma cells in combination with
either APRIL or stromal cell-soluble factors. Leukemia 28: 1647–
1656.
Molecular classification
Keats, J.J., Fonseca, R., Chesi, M. et al. (2007). Promiscuous mutations
activate the noncanonical NF-kappaB pathway in multiple myeloma. Bergsagel, P.L., Kuehl, W.M., Zhan, F. et al. (2005). Cyclin D dysregu-
Cancer Cell 12: 131–144. lation: an early and unifying pathogenic event in multiple myeloma.
Kuo, A.J., Cheung, P., Chen, K. et al. (2011). NSD2 links dimethylation Blood 106: 296–303.
of histone H3 at lysine 36 to oncogenic programming. Mol. Cell 44: Broyl, A., Hose, D., Lokhorst, H. et al. (2010). Gene expression profiling
609–620. for molecular classification of multiple myeloma in newly diagnosed
Lode, L., Eveillard, M., Trichet, V. et al. (2010). Mutations in TP53 are patients. Blood 116: 2543–2553.
exclusively associated with del(17p) in multiple myeloma. Haemato- Keats, J.J., Chesi, M., Egan, J.B. et al. (2012). Clonal competition with
logica 95: 1973–1976. alternating dominance in multiple myeloma. Blood 120: 1067–1076.
Martinez-Garcia, E., Popovic, R., Min, D.-J. et al. (2011). The MMSET Lohr, J.G., Stojanov, P., Carter, S.L. et al. (2014). Widespread genetic
histone methyl transferase switches global histone methylation and heterogeneity in multiple myeloma: implications for targeted therapy.
alters gene expression in t(4;14) multiple myeloma cells. Blood 117: Cancer Cell 25: 91–101.
211–220. Mikulasova, A., Wardell, C.P., Murison, A. et al. (2017). The spectrum
Pei, H., Wu, X., Liu, T. et al. (2013). The histone methyltransferase of somatic mutations in monoclonal gammopathy of undetermined
MMSET regulates class switch recombination. J. Immunol. 190: 756– significance indicates a less complex genomic landscape than that in
763. multiple myeloma. Haematologica 102: 1617–1625.
Pei, H., Zhang, L., Luo, K. et al. (2011). MMSET regulates histone Rasche, L., Chavan, S.S., Stephens, O.W. et al. (2017). Spatial
H4K20 methylation and 53BP1 accumulation at DNA damage sites. genomic heterogeneity in multiple myeloma revealed by multi-region
Nature 470: 124–128. sequencing. Nat. Commun. 8: 268.
Salhia, B., Baker, A., Ahmann, G. et al. (2010). DNA methylation analy- Walker, B.A., Boyle, E.M., Wardell, C.P. et al. (2015). Mutational spec-
sis determines the high frequency of genic hypomethylation and low trum, copy number changes, and outcome: results of a sequencing
frequency of hypermethylation events in plasma cell tumors. Cancer study of patients with newly diagnosed myeloma. J. Clin. Oncol. 33:
Res. 70: 6934–6944. 3911–3920.
Shah, M.Y., Martinez-Garcia, E., Phillip, J.M. et al. (2016). Walker, B.A., Mavrommatis, K., Wardell, C.P. et al. (2018). A high-risk,
MMSET/WHSC1 enhances DNA damage repair leading to an double-hit, group of newly diagnosed myeloma identified by genomic
increase in resistance to chemotherapeutic agents. Oncogene 35: analysis. Leukemia 2019; 33: 159–170.
5905–5915. Walker, B.A., Mavrommatis, K., Wardell, C.P. et al. (2018). Identifica-
Steinbrunn, T., Stuhmer, T., Gattenlohner, S. et al. (2011). Mutated RAS tion of novel mutational drivers reveals oncogene dependencies in
and constitutively activated Akt delineate distinct oncogenic path- multiple myeloma. Blood 132: 587–597.
ways, which independently contribute to multiple myeloma cell sur- Weinhold, N., Ashby, C., Rasche, L. et al. (2016). Clonal selection and
vival. Blood 117: 1998–2004. double-hit events involving tumor suppressor genes underlie relapse
Walker, B.A., Wardell, C.P., Chiecchio, L. et al. (2011). Aberrant global in myeloma. Blood 128: 1735–1744.
methylation patterns affect the molecular pathogenesis and prognosis Zhan, F., Huang, Y., Colla, S. et al. (2006). The molecular classification
of multiple myeloma. Blood 117: 553–562. of multiple myeloma. Blood 108: 2020–2028.
130 Molecular Hematology

Prognosis Trudel, S., Li, Z.H., Wei, E. et al. (2005). CHIR-258, a novel, multitar-
geted tyrosine kinase inhibitor for the potential treatment of t(4;14)
Avet-Loiseau, H., Attal, M., Campion, L. et al. (2012). Long-term anal- multiple myeloma. Blood 105: 2941–2948.
ysis of the IFM 99 trials for myeloma: cytogenetic abnormalities Weinhold, N., Heuck, C.J., Rosenthal, A. et al. (2016). Clinical value of
[t(4;14), del(17p), 1q gains] play a major role in defining long-term molecular subtyping multiple myeloma using gene expression profil-
survival. J. Clin. Oncol. 30: 1949–1952. ing. Leukemia 30: 423–430.
Brioli, A., Kaiser, M.F., Pawlyn, C. et al. (2013). Biologically defined risk
groups can be used to define the impact of thalidomide maintenance
therapy in newly diagnosed multiple myeloma. Leuk. Lymphoma 54: Current therapy
1975–1981. Attal, M., Lauwers-Cances, V., Hulin, C. et al. (2017). Lenalidomide,
Kumar, S., Fonseca, R., Ketterling, R.P. et al. (2012). Trisomies in mul- bortezomib, and dexamethasone with transplantation for myeloma.
tiple myeloma: impact on survival in patients with high-risk cytoge- N. Engl. J. Med. 376: 1311–1320.
netics. Blood 119: 2100–2105. Attal, M., Lauwers-Cances, V., Marit, G. et al. (2012). Lenalidomide
Nair, B., van Rhee, F., Shaughnessy, J.D. et al. (2010). Superior results of maintenance after stem-cell transplantation for multiple myeloma. N.
total therapy 3 (2003–33) in gene expression profiling-defined low- Engl. J. Med. 366: 1782–1791.
risk multiple myeloma confirmed in subsequent trial 2006–66 with Dimopoulos, M.A., Oriol, A., Nahi, H. et al. (2016). Daratumumab,
VRD maintenance. Blood 115: 4168–4173. lenalidomide, and dexamethasone for multiple myeloma. N. Engl. J.
Thanendrarajan, S., Tian, E., Qu, P. et al. (2017). The level of dele- Med. 375: 1319–1331.
tion 17p and bi-allelic inactivation of TP53 has a significant impact Lokhorst, H.M., Plesner, T., Laubach, J.P. et al. (2015). Targeting CD38
on clinical outcome in multiple myeloma. Haematologica 102: e364– with daratumumab monotherapy in multiple myeloma. N. Engl. J.
e367. Med. 373: 1207–1219.
Lonial, S., Dimopoulos, M., Palumbo, A. et al. (2015). Elotuzumab ther-
Targeted therapy apy for relapsed or refractory multiple myeloma. N. Engl. J. Med. 373:
621–631.
Andrulis, M., Lehners, N., Capper, D. et al. (2013). Targeting the BRAF Mateos, M.V., Dimopoulos, M.A., Cavo, M. et al. (2018). Daratumumab
V600E mutation in multiple myeloma. Cancer Discov. 3: 862–869. plus bortezomib, melphalan, and prednisone for untreated myeloma.
Bjorklund, C.C., Lu, L., Kang, J. et al. (2015). Rate of CRL4(CRBN) sub- N. Engl. J. Med. 378: 518–528.
strate Ikaros and Aiolos degradation underlies differential activity of Mateos, M.V., Hernandez, M.T., Giraldo, P. et al. (2013). Lenalidomide
lenalidomide and pomalidomide in multiple myeloma cells by regu- plus dexamethasone for high-risk smoldering multiple myeloma. N.
lation of c-Myc and IRF4. Blood Cancer J. 5: e354. Engl. J. Med. 369: 438–447.
Chesi, M., Matthews, G.M., Garbitt, V.M. et al. (2012). Drug response McCarthy, P.L., Owzar, K., Hofmeister, C.C. et al. (2012). Lenalidomide
in a genetically engineered mouse model of multiple myeloma is pre- after stem-cell transplantation for multiple myeloma. N. Engl. J. Med.
dictive of clinical efficacy. Blood 120: 376–385. 366: 1770–1781.
Delmore, J.E., Issa, G.C., Lemieux, M.E. et al. (2011). BET bromod- Mikhael, J.R., Dingli, D., Roy, V. et al. (2013). Management of newly
omain inhibition as a therapeutic strategy to target c-Myc. Cell 146: diagnosed symptomatic multiple myeloma: updated Mayo Stratifica-
904–917. tion of Myeloma and Risk-Adapted Therapy (mSMART) consensus
Demchenko, Y.N., Brents, L.A., Li, Z. et al. (2014). Novel inhibitors are guidelines 2013. Mayo Clin. Proc. 88: 360–376.
cytotoxic for myeloma cells with NFkB inducing kinase-dependent Moreau, P., Masszi, T., Grzasko, N. et al. (2016). Oral ixazomib, lenalido-
activation of NFkB. Oncotarget 5: 4554–4566. mide, and dexamethasone for multiple myeloma. N. Engl. J. Med. 374:
Heuck, C.J., Jethava, Y., Khan, R. et al. (2016). Inhibiting MEK in MAPK 1621–1634.
pathway-activated myeloma. Leukemia 30: 976–980. Palumbo, A., Cavallo, F., Gay, F. et al. (2014). Autologous transplanta-
Lee, L., Draper, B., Chaplin, N. et al. (2017, 2018). An APRIL based tion and maintenance therapy in multiple myeloma. N. Engl. J. Med.
chimeric antigen receptor for dual targeting of BCMA and TACI in 371: 895–905.
multiple myeloma. Blood 2018; 131: 746–758. Palumbo, A., Chanan-Khan, A., Weisel, K. et al. (2016). Daratumumab,
Mulligan, G., Lichter, D.I., Di Bacco, A. et al. (2014). Mutation of NRAS bortezomib, and dexamethasone for multiple myeloma. N. Engl. J.
but not KRAS significantly reduces myeloma sensitivity to single- Med. 375: 754–766.
agent bortezomib therapy. Blood 123: 632–639. San Miguel, J.F., Schlag, R., Khuageva, N.K. et al. (2008). Bortezomib
Qing, J., Du, X., Chen, Y. et al. (2009). Antibody-based targeting of plus melphalan and prednisone for initial treatment of multiple
FGFR3 in bladder carcinoma and t(4;14)-positive multiple myeloma myeloma. N. Engl. J. Med. 359: 906–917.
in mice. J. Clin. Invest. 119: 1216–1229. Stewart, A.K., Rajkumar, S.V., Dimopoulos, M.A. et al. (2015). Carfil-
Seckinger, A., Delgado, J.A., Moser, S. et al. (2017). Target expression, zomib, lenalidomide, and dexamethasone for relapsed multiple
generation, preclinical activity, and pharmacokinetics of the BCMA- myeloma. N. Engl. J. Med. 372: 142–152.
T cell bispecific antibody EM801 for multiple myeloma treatment. Weber, D.M., Chen, C., Niesvizky, R. et al. (2007). Lenalidomide plus
Cancer Cell 31: 396–410. dexamethasone for relapsed multiple myeloma in North America. N.
Tai, Y.T., Mayes, P.A., Acharya, C. et al. (2014). Novel anti-B-cell mat- Engl. J. Med. 357: 2133–2142.
uration antigen antibody-drug conjugate (GSK2857916) selectively
induces killing of multiple myeloma. Blood 123: 3128–3138.
Chapter 11 The molecular basis of bone
marrow failure syndromes and red cell
enzymopathies
Deena Iskander1 , Lucio Luzzatto2 & Anastasios Karadimitris2
1
Centre for Haematology, Imperial College London, Hammersmith Hospital, London, UK
2 Department of Haematology and Blood Transfusion, Muhimbili University College of Health Sciences, Dar-es-Salaam, Tanzania

Introduction, 131 Concluding remarks, 151


Conditions associated with bone marrow failure, 131 Further reading, 151
Red cell enzyme deficiencies, 144

Acquired BMF syndromes


Introduction
Idiopathic aplastic anemia
Since the last edition of this book the pace of advances in
IAA accounts for the majority (about 80–90%) of cases of
molecular hematology has been such that, after due consider-
acquired BMF syndromes, with an incidence estimated at
ation, we had to decide that, within the space allowed, it has
2 per million per year (two- to threefold higher in the Far
become impossible to do justice to the original title of this
East and possibly in Africa).
chapter, “The Molecular Basis of Anemia.” We have opted
instead to focus on anemias resulting from bone marrow fail-
ure (BMF) syndromes, and on anemias caused by heritable Immunopathogenesis The most direct evidence that IAA may
defects in the glycolytic pathway and in the pentose phos- be an autoimmune disorder came from the clinical observa-
phate pathways. In these areas, discoveries spurred in good tion that patients with IAA have complete or partial rever-
part by new technologies such as high-throughput sequenc- sion of their pancytopenia when they are treated with anti-
ing have led to new insights into pathophysiology and also lymphocyte globulin (ALG). Subsequently, it was shown that
to new therapeutic developments that we aim to highlight patients with IAA often have increased numbers of “acti-
here. vated” CD8+ CD25+ T cells in their blood and bone marrow.
In addition, T cells from IAA patients can inhibit the growth
of autologous in vitro hematopoietic colonies, and the growth
of colonies from human leukocyte antigen (HLA)-identical
Conditions associated with bone siblings. Based on these observations, a current model of the
marrow failure pathogenesis of IAA posits that auto-reactive T cells attack
hematopoietic stem cells (HSCs), causing their depletion –
The disease entities falling under this heading are quite hence the reduction of HSCs in severe IAA to about 1% of
diverse, but they have in common a crucial pathogenetic normal.
mechanism: the loss of hematopoietic stem cells and/or pro- The primary event that triggers this aberrant immune
genitor cells (HSPCs). The pace of stem cell depletion varies response remains elusive; a possible viral cause has long been
widely, from weeks (e.g. in idiopathic aplastic anemia, IAA) sought, but never proven. The identity of the putative auto-
to years (e.g. in Fanconi anemia, FA). These two condi- antigen on HSCs also remains unknown. There is evidence,
tions exemplify well two broad categories of BMF syndromes: however, that the inhibitory effect on HSCs of auto-reactive T
those that are acquired and those that are inherited (see cells is mediated, at least in part, through interferon-gamma
Table 11.1). (IFN-γ). In addition, IFN-γ upregulates the Fas receptor
on the surface of HSPCs, thus facilitating activation of the
Fas-dependent apoptotic pathways.
As in other autoimmune diseases, there is overrepresen-
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. tation of certain HLA alleles in IAA patients compared with
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. population controls: this is true for the HLA-DR2 in patients

131
132 Molecular Hematology

Table 11.1 Classification of the bone marrow failure (BMF) syndromes

Mode of Chromosomal
Disease inheritance locus Gene Clinical manifestations

Inherited
Fanconi anemia AR/X linked See Table 11.2 See Table 11.2 See text
Dyskeratosis congenita X-linked recessive Xq28 DKC1 See text
AD 3q26 TERC
AD or AR 5p15 TERT
AR 17p13 CTC1
AD 14q12 TINF2
AR 15q14 NOP10
AR 17p13 WRAP53/TCAB1
AD or AR 16q2 ACD/TPP1
AD or AR 20q13 RTEL1
AR 16p13 PARN
AR 5q35 NHP2
Diamond–Blackfan anemia AD See text See text See text
X-linked recessive Xp11 TSR2
X-linked recessive Xp11 GATA-1
Shwachman–Diamond syndrome AR 7q11 SBDS Neutropenia, exocrine pancreatic
insufficiency, metaphyseal dysostosis
Amegakaryocytic thrombocytopenia AR 1p34 c-mpl (thrombopoietin Absent megakaryocytes in bone marrow,
receptor) late BMF
Thrombocytopenia with absent radii AR 1q21 RBM8A Bilateral radial aplasia, lower limb
syndrome anomalies, cow’s milk intolerance, renal
anomalies, and cardiac anomalies
Congenital thrombocytopenia and AD 7p15–p14 HOXA11 Aplastic anemia, proximal radius–ulna
radius–ulna synostosis De novo 3q26 MECOM synostosis, clinodactyly, syndactyly, hip
dysplasia, and sensorineural hearing loss
Cartilage–hair hypoplasia AR 9p13 RMRP Short tubular bones, sparse hair, severe
immunodeficiency, macrocytic anemia
Pearson marrow–pancreas syndrome Mitochondrial Usually from nt Contiguous genes Pancreatic exocrine dysfunction,
8.469 to nt deleted sideroblastic anemia (see also text)
13447
Acquired
Idiopathic
Transient erythroblastopenia of childhood (TEC)
Radiation
Drugs and chemicals
Regular: cytotoxic, benzene
Idiosyncratic: chloramphenicol, NSAIDs, anti-epileptics, gold
Viruses
Epstein–Barr virus
Hepatitis
Parvovirus
Human immunodeficiency virus
Cytomegalovirus
Human T-lymphotropic virus-1
Human herpes virus-6
Immune diseases
Thymoma (5–10% develop pure red cell aplasia)
Myasthenia gravis
Systemic lupus erythematous
Pregnancy
Paroxysmal nocturnal hemoglobinuria

AD, autosomal dominant; AR, autosomal recessive; NSAID, BMF, bone marrow failure; non-steroidal anti-inflammatory drug; nt, nucleotide.
The molecular basis of bone marrow failure syndromes and red cell enzymopathies 133

with IAA of European ancestry, and for others in Japanese on the few residual HSCs in IAA. In one study, eltrombopag
patients with IAA. improved one or more cytopenias in 40% of patients after
Clinical findings have signaled an overlap in pathogene- three to four months of treatment. Furthermore, hematolog-
sis between IAA and myelodysplastic syndrome (MDS), in ical responses were sustained after drug discontinuation in
particular with the hypoplastic form of MDS; indeed, the five patients who had been on eltrombopag for months to
differential diagnosis between the two is often difficult. It is years.
now recognized that 20–30% of patients with MDS, especially
those conventionally classified as having refractory anemia Clonal hematopoiesis in IAA A significant proportion of
(RA) with hypocellular marrow, respond to immunosuppres- patients with IAA harbor PNH clones and approximately
sive therapy (IST) with alleviation of their cytopenias, sug- 20% of patients treated with IST have clinical manifestations
gesting that an immune process, similar to that operating in of PNH. IAA, especially after successful IST, also bears a sig-
IAA, is also involved in the pathogenesis of a subset of MDS nificant risk of late clonal disorders: the risk of MDS, AML,
patients. In addition, in 20% of patients with RA, small pop- and tumors of other organs is 18.8%, compared with 3.1%
ulations of paroxysmal nocturnal hemoglobinuria (PNH)- after bone marrow transplantation. Cytogenetic abnormal-
like blood cells are detected, as they are commonly seen in ities observed in IAA include +8, −7 (usually indicative of
IAA (see later). What is more, their presence is predictive of MDS if present at diagnosis), del(13q), and del(5q). Another
response to IST, providing further evidence of an immune common genetic lesion, identified by single-nucleotide poly-
process in the pathogenesis of MDS in this select group of morphism (SNP) array-based karyotyping, is acquired copy
patients. number and loss of heterozygosity of the short arm of chro-
The role of mutations in genes involved in telomere main- mosome 6 (6pLOH), which leads to the loss of the respec-
tenance in the pathogenesis of IAA is discussed later (see tive HLA-A haplotype. Given that in IAA CTLs may target
Dyskeratosis congenita). HSPCs that present autoantigens expressed by class I HLA
molecules, 6pLOH might result in immune escape and clonal
Clinical aspects and treatment The clinical picture of IAA dominance of a subset of HSPCs. SM have also been reported
generally reflects the extent of HSPC loss and the subsequent in PIGA (characteristic of PNH) and STAT3 (characteristic of
cytopenias. Typically, a patient with severe IAA presents with 40% of patients with T-LGL).
bruising and mucosal bleeding, anemia and septic episodes In the last few years, high-throughput sequencing has
(bacterial or fungal), but without hepatosplenomegaly. The expanded the repertoire of genetic events underlying clonal
differential diagnosis of IAA, as well as that of hypoplastic hematopoiesis in IAA and identified some of their clinical
MDS, includes inherited BMF syndromes, the aplastic form sequelae. These studies demonstrate that SMs in IAA occur
of childhood acute lymphoblastic leukemia, and infectious in approximately 50% of patients and are often the same
and malignant processes that may infiltrate the bone marrow. as observed in myeloid malignancies. For example, in one
Thus the diagnosis of IAA is made eventually by exclusion of cohort, 20% of patients had mutations in ASXL1, DNMT3,
bone marrow infiltration/fibrosis and of major granulocytic and BCOR. In those with IAA for more than six months, the
or megakaryocytic dysplasia. presence of these mutations was associated with a 40% risk of
The treatment of IAA involves supportive care for all progression to MDS. While ASXL1 and DNMT3A mutations
patients. Further management is dictated by its severity (as are predictive of poor response to immunosuppression and
determined by the degree of pancytopenia, reticulocytope- poor survival, PIGA and BCOR/BCOR1 mutations correlate
nia, and bone marrow cellularity) and by the patient’s age. positively with disease response and survival. Importantly,
Hematopoietic stem cell transplantation (HSCT) from an out of 43 patients successfully treated with eltrombopag, 8
HLA-identical sibling or from an alternative donor is the developed clonal cytogenetic abnormalities, most frequently
treatment of choice for younger patients <40 years with monosomy 7, which is known to predate transformation to
severe IAA, and offers better than 75% 6-year survival. In MDS and AML.
the absence of an appropriate donor, or when the patient is It will be pertinent to address when, in the natural history
older or the disease milder, immunosuppressive treatment, of the disease, these clones arise and how they are selected
in particular the combination of ALG or antithymocyte for. We can hypothesize that, analogous to PNH and LOH6p
globulin (ATG) with cyclosporin, results in a complete or clones, clones with SMs are propagated by immune destruc-
partial response in the majority of cases. More recently, the tion, both inherent to the disease and resulting from IST
thrombopoietin receptor agonist (TpoR) eltrombopag was (Figure 11.1). The interpretation of SMs in IAA must also
approved for the treatment of patients with severe aplastic take into account the recent observation of CHIP. In this
anemia refractory to IST. Eltrombopag is thought to context, the presence of SMs is associated with an increased
stimulate stem cell self-renewal and “awaken” HSCs from risk of hematological malignancy, in particular in cases with
their dormant state by non-competitive binding to the TpoR a high mean allele burden. Further clarification of these
134 Molecular Hematology

CD8+CD25+
CD4+THI CTLS TREGS

Recovery of normal
Immune-mediated BM hematopoiesis
destruction via IFNy
and TNFα
HSC pool
Intrinsic
to AA

OR
IST
Emergence of clones by
Aging Eltrambopag
immune escape and/or
proliferative advantage PNH
- occurs in patients with
PIGA mutant HSC and
GPI-specific T cells
CHIP whereby PIGA mutant
DNMT3A clone repopulates bone
TET2 Clone size marrow
ASXL1 Stable clone
TP53 size MDS/AML
Trisomy 8 Monosomy 7
Deletion 13q ASXL
LOH6P DNMT3A
BCOR/BCOR1
Fig. 11.1 Somatic mutations (SM) in idiopathic aplastic anemia (IAA). IAA results from immune destruction of hematopoietic stem cells
(HSCs) by CD8+ cytotoxic T lymphocytes (CTLs). Increased CD4+ T-helper-1 (TH1) cells offer assistance to CTLs and insufficient T-regulatory (Tregs)
cells are postulated to maintain autoreactive clones. Treatment with IST or eltrombopag enables some stem cells to survive/recover. In this scenario,
any clone with a somatic mutation (SM) that confers resistance to CTLs and/or has a proliferative advantage will either remain stable in size or will
expand. This entails a risk of transformation to myelodysplastic syndromes (MDS) and/or acute myeloid leukemia (AML). SMs also take place in the
context of normal aging and, in view of the presence of the resulting clones, the term “clonal hemopoiesis of indeterminate potential” (CHIP) has
been coined. These SMs are usually distinct from those that arise in IAA, but may also predispose to MDS/AML.

distinct physio- and pathological entities should allow for Molecular pathogenesis The initiating event in the pathogen-
more tailored treatment, for instance earlier consideration esis of PNH consists of SM(s) in the X-linked gene PIGA in a
of HSCT in patients with IAA and unfavorable SMs. At the multipotent HSC (sometimes more than one). PIGA encodes
same time, it is important to note that there is no evidence the enzymatically active subunit of an N-acetylglucosamine
that IAA originates from abnormal clones. The current transferase. This enzyme catalyzes an early step in the for-
phrase “clonal hematopoiesis in aplastic anemia” means mation of a complex glycolipid molecule called glycosylphos-
exactly what it says: namely, that in the bone marrow envi- phatidylinositol (GPI; Figure 11.2). The synthesis of GPI
ronment of IAA a variety of clones – which would otherwise takes place initially on the cytoplasmic surface of the endo-
remain unnoticed – may instead become detectable or plasmic reticulum, and is then completed on its luminal sur-
even prosper, whether because they have a relative selective face. Once formed, the GPI molecules (anchors) are cova-
advantage or through sheer opportunism as discussed in lently linked through a trans-peptidation reaction to the
detail elsewhere (Luzzatto & Risistano, 2018). carboxy-terminus of a variety of proteins. The GPI-linked
proteins, after post-translational modifications in the Golgi
apparatus, emerge eventually on the cell surface, to which
Paroxysmal nocturnal hemoglobinuria
they remain attached through the GPI anchor. As a result of
PNH is a rare acquired hematological disorder with three the impaired synthesis of GPI, blood cells are either com-
main clinical features: intravascular hemolysis, tendency to pletely (PNH III) or partially (PNH II) deficient in GPI-
thrombosis, and BMF of variable severity. As in IAA, the linked proteins. X-linkage of PIGA is highly relevant to the
precise cause of BMF remains unclear; in contrast, the molec- pathogenesis of PNH, because there is only one allele in
ular mechanism of hemolysis is well explained. For this rea- males, and only one active allele in female somatic cells, and
son, the space devoted here to this condition is out of propor- therefore just one inactivating mutation is sufficient to cause
tion to its prevalence. the consequences just outlined.
The molecular basis of bone marrow failure syndromes and red cell enzymopathies 135

PIGT* Complement activation and pathophysiology of red cell


GPI destruction For the majority of GPI-linked proteins the func-
EtNP protein maturation in tional consequences of their cell surface deficiency are not
Golgi
PIGO known; however, for CD55 and especially CD59, we do know
PGAP2 they are directly implicated in the destruction of red cells in
M PGAP3 PNH. CD59 is a critical negative regulator of complement
action at the surface of erythrocytes, as it interferes with
EtNP M
the formation of the membrane attack complex (MAC). The
PIGN PIGV MAC is formed first by the assembly of the terminal comple-
M
ment pathway components C6, C7, C8; then, the large multi-
EtNP
protein structure is completed through homopolymerization
PIGM
of C9, which produces physical pores in the cell membrane;
GlcN PIGL it is this final step that CD59 hinders. Activation of the ter-
PIGA minal pathway depends in turn on cleavage activation of C5
Inositol by upstream components of the alternative or classical prox-
imal pathways of complement activation. When, as a result
P
of a PIGA mutation, CD59 is low or absent, red cells become
susceptible to baseline or stress-induced complement activa-
tion, resulting in red cell lysis (intravascular hemolysis). This
Cell membrane
phenomenon is reproduced in vitro in the Ham test, whereby
the patient’s red cells are lysed by either autologous or ABO-
compatible donor acidified serum (pH about 6.5), as comple-
ment binds to red blood cell (RBC) at a weakly acidic pH.
Fig. 11.2 The glycosylphosphatidylinositol (GPI) anchor and its As for thrombosis, this may result in part from
genetic disorders. The structure of GPI–protein (protein–EtNP- complement-mediated activation of platelets deficient
6Manα1-2Manα1-6Manα1-4GlcNα1-6myoinositol-phospholipid) is
in CD55 and CD59. Other, as yet unidentified, genetic or
shown. GPI biosynthesis takes place mostly in the endoplasmic
acquired factors affecting coagulation and/or fibrinolysis
reticulum. The first step involves the addition of acetylglucosamine
(GlcN) to phosphatidylinositol (inositol-P). Synthesis of the GPI anchor
may have an additive or synergistic effect in producing
proceeds with the serial addition of a glycan moiety consisting of three thrombosis, which may be devastating, especially as it tends
mannose (M) molecules each modified by phosphoethanolamine to affect abdominal and cerebral veins. Given the central
(EtNP). Through a transpeptidation reaction, proteins with the role of C5 in complement activation, we could expect that
appropriate carboxy-terminal amino acid motif are attached covalently inhibiting its cleavage would prevent formation of the MAC
to the terminal EtNP. The GPI–protein complex subsequently travels to and thus destruction of red cells deficient in surface CD59.
the cell surface, where the protein becomes anchored to the lipid Indeed, eculizumab, a humanized anti-C5 monoclonal
bilayer through GPI. In PNH, PIG-A, a protein encoded by the X-linked antibody, is a very potent inhibitor of C5 cleavage with
gene PIGA and a member of a multi-subunit enzymatic complex that spectacular clinical effects (see later). By abrogating intravas-
catalyzes the first step (i.e. addition of GlcN to inositol-P), is
cular hemolysis, eculizumab also reduces the free plasma
somatically mutated in one or few HSCs. As a result, very little GPI is
hemoglobin that binds and depletes plasma nitric oxide,
synthesized, or none at all, with consequent severe deficiency of
GPI-anchored proteins on the surface of the mutated HSCs and their
a natural vasodilator; thus, it also reduces the incidence
progeny. In Inherited GPI deficiency, germline hypomorphic mutations of smooth muscle dystonias not uncommon in PNH (i.e.
in PIGL, PIGM, PIGN, PIGV, PIGO, PGAP2, or PGAP3 result in variable abdominal pain, retrosternal pain, and erectile dysfunction).
GPI deficiency. Germline mutations in PIGA are embryonically lethal in
males, with the exception of two families. Here, monoallelic PIGA Cellular pathogenesis Since PIGA-mutant PNH HSCs
mutations resulted in a truncated PIGA protein with sufficient residual lack GPI-linked proteins, one might have thought that
function to allow survival of the affected individuals, but causing they would compete poorly with normal HSCs. Instead,
severe congenital abnormalities and intellectual disability. For PIGT, a PNH HSCs can expand until they largely supplant normal
case of a germline mutation in one allele with a tissue-specific somatic hematopoiesis. In the first instance, this paradox may be
mutation expressed in blood cells has been reported. EtNP,
explained by invoking an intrinsic proliferative advantage of
phosphoethanolamine; GlcN, glucosamine; M, mannose; P,
PNH HSCs over normal HSCs, as is the case with leukemic
Phosphatidyl.
cells; however, the fact that patients with PNH can live for
decades with normal and PNH hematopoiesis coexisting
in their bone marrow goes against this notion. Lack of
a competitive growth advantage by PNH clones has also
136 Molecular Hematology

been demonstrated experimentally in pig-a null mouse product wherever they fall, whereas interestingly, missense
models. It remains possible that secondary mutations in the mutations are clustered mainly within exon 2, where it is pre-
PIG-A mutant clone may confer a proliferative advantage. sumed that amino acid residues critical for catalytic activity
In fact, in two patients molecular studies revealed that, as are located.
a result of the same (12;12) chromosomal translocation,
the transcription factor HMGA2 had a truncation of its 3′ Clinical aspects and treatment Although in the prototypi-
untranslated region. This may remove negative regulatory cal PNH patient hemoglobinuria is picturesquely qualified
elements, resulting in increased expression of HMGA2, but as paroxysmal, intravascular hemolysis is in fact continuous.
how increased HMGA2 could impart the cell with apparently Against this background of chronic hemolysis, the parox-
non-malignant growth advantage is not known. Recently, ysms are acute exacerbations experienced by PNH patients
mutations in several genes other than PIGA, essentially the during intercurrent illnesses such as infections (presumably
same as in IAA, have been found in the blood cells of patients because this is associated with activation of complement),
with PNH. However, the same study has shown that clonal other stressful events, or for no obvious reason.
expansion depended on the PIGA mutation itself. Hemolytic anemia with macrocytosis (partly due to reticu-
The most widely accepted pathogenetic model, the escape locytosis and partly due to BMF), different degrees of throm-
model, was suggested by the long-known association between bocytopenia and leukopenia, iron deficiency, and hemosider-
PNH and IAA. In this model, the focus is on the notion that inuria should raise the suspicion of PNH. The occurrence of
in PNH HSC-specific T cells would selectively target nor- venous thrombosis at an unusual site, such as the brain or the
mal HSCs, but not PNH HSCs. Under these circumstances, abdomen, ought to be an even more compelling pointer to
PNH HSCs would expand and sustain hematopoiesis, in the possible diagnosis of PNH. Venous thrombosis of small
some cases for years, to the tune of 90% or more. Several or large vessels is a serious and potentially life-threatening
lines of evidence support the escape model. First, small PNH complication of PNH, occurring in 40–50% of patients.
clones are present in as many as 50% of patients with bona Historically, the diagnosis was confirmed by the Ham
fide IAA; and minute PNH clones (∼1 in 105 granulocytes) test (which only detects the PNH abnormality in erythro-
are seen even in normal individuals. Second, expanded T cytes), but this assay has largely been supplanted by flow
cell clones have been demonstrated in the blood of PNH cytometric analysis. The latter is the most sensitive diagnos-
patients at a frequency three times higher than in appro- tic tool for PNH (capable of detecting <1 in 10 000 PNH
priate controls. Further characterization of T cells in PNH cells): it will demonstrate that there is a population of ery-
has revealed (i) increased frequency of CD8+ CD57+ T cells throcytes and leukocytes with absent or markedly reduced
expressing predominantly activating killer immunoglobulin expression of GPI-linked proteins such as CD55 and CD59.
and NKG2/CD94 receptors; (ii) the presence of exactly the Detection of GPI-deficient monocyte and granulocyte clones
same, or very similar, T-cell receptor (TCR)-β CDR3 clono- is also indicated by the absence of fluorescent aerolysin
typic sequences in the CD8+ CD57+ T cell subsets from differ- (FLAER), which binds directly to the cell surface GPI anchor
ent patients; and (iii) there is increased representation of the itself.
HLA-DR2 allele in PNH patients compared with population Since its introduction in 2006, eculizumab has been the
controls (as in IAA). main treatment for PNH. This humanized mAb inhibits
The molecular target of the postulated autoreactive T cells cleavage of the complement component C5, resulting in (i)
is not yet known. Potential targets are a surface GPI-linked almost complete abrogation of hemolysis in the majority of
protein or immunogenic peptides derived therefrom and but not all patients; (ii) reduction in transfusion require-
presented by the HLA complex; or the GPI molecule itself, ments, resulting in hemoglobin stabilization or transfusion
presented by CD1d. Experimental evidence has ruled out the independence in up to two-thirds of patients; (iii) significant
first mechanism, while the second mechanism is supported improvement in quality of life for most patients, in particular
by the identification of GPI-specific, CD1d-restricted T cells due to the mitigation of fatigue; and (iv) a reduction in the
in the peripheral blood of patients with PNH, and recently risk of thrombosis. Importantly, patients at risk of thrombo-
also with IAA. sis (i.e. those with large PNH clones or an additional throm-
bophilia) should still be considered for prophylactic long-
Molecular pathology All types of mutations have been term anticoagulation. Patients on eculizumab should also be
observed in the PIGA gene in patients with PNH. The vaccinated against encapsulated bacteria, especially Neisseria
majority (∼75%) are small insertions or deletions caus- meningitides, to which they are susceptible due to comple-
ing frameshifts, and the rest are nonsense and missense ment blockade.
point mutations. The nonsense and frameshift mutations are Unfortunately, worldwide use of eculizumab is precluded
spread throughout the coding sequence (exons 2–6), presum- by its high cost. Furthermore, the clinical response to
ably because they cause complete inactivation of the gene eculizumab is not uniform in all patients. There are several
The molecular basis of bone marrow failure syndromes and red cell enzymopathies 137

reasons for this variability. First of all, in some cases the autosomal recessive mutations in genes of the GPI biosyn-
anemia is due not only to hemolysis, but also to BMF, with thetic pathway. In the first example of IGD, in two unrelated
eculizumab acting on the former, but not on the latter. consanguineous families, affected children had spontaneous
Second, in all patients on eculizumab there is extravascular splanchnic vein thrombosis in the first year of life and
hemolysis, which occurs because, in the absence of the drug, absence seizures that became refractory to treatment later
C3 bound to PNH RBC membranes leads to C5 activation on. In contrast to PNH, there is no significant intravascular
and complement-mediated cell lysis. On eculizumab C5 is hemolysis nor BMF. In all three affected children, the same
blocked, and therefore cell lysis does not occur; however, promoter mutation was identified: a C>G substitution at
RBCs become opsonized by C3 molecules and are in turn position −270 from the start codon of the PIGM gene, which
phagocytosed in the reticuloendothelial system. As a result encodes a mannosyltransferase responsible for the first
of this opsonization process, the Coombs test becomes and essential mannosylation reaction in the biosynthesis
positive (typically it is negative in untreated PNH). of GPI.
Of note is that the extent of C3 binding to PNH RBCs The hypomorphic nature of the C>G mutation allows
has been shown to correlate with homozygosity for the rare for some transcriptional output, the extent of which varies
L allele at the complement 1 receptor locus. Thus, the L/L from tissue to tissue, hence the variable degree of GPI
(as opposed to L/H or hereditary hemochromatosis, HH) deficiency. For example, in red cells, GPI expression is
genotype is associated with a lesser response to treatment almost normal, explaining the lack of significant hemolysis;
with eculizumab in patients with PNH. In a small number of by contrast, granulocytes and B cells display almost com-
Japanese and Chinese patients, complete failure to respond plete, and fibroblasts partial, GPI deficiency. The precise
to eculizumab is caused by an SNP in C5 (c.2654G → A, molecular mechanism of this disease has recently been
p.Arg885His), which abrogates binding of eculizumab to its delineated: C>G substitution disrupts binding of the generic
target peptide in C5; the last two phenomena highlight a transcription factor Sp1 to a glucocorticoids (GC)-rich box
classical paradigm of pharmacogenomics. Thus, there is still in the core promoter of PIGM. Unlike in red cells, where
(extravascular) hemolysis on eculizumab and, as a result, PIGM transcription is independent of Sp1 binding to its core
some patients still require blood transfusion; because they promoter, in B cells PIGM transcription is dependent upon
no longer lose iron through hemoglobinuria, they are now Sp1 binding, hence the divergent GPI expression across cell
at risk of iron overload and may require iron chelation. How- lineages. Impaired Sp1 binding leads to reduced histone
ever, it is important to note that these patients still benefit acetylation in the promoter of PIGM and in turn transcrip-
from the abrogation of intravascular hemolysis and the con- tional repression by nucleosomal compaction, recruitment
sequent symptoms already mentioned. of the Polycomb repressor complex, and establishment of
Long-term therapeutic options for PNH include immuno- a bivalent chromatin state. As such, PIGM-associated IGD
suppressive agents (e.g. a combination of ALG/ATG and represents the first Mendelian disease whose pathogenesis
cyclosporin, especially for patients with moderate to severe is attributed to Polycomb-mediated bivalent chromatin
cytopenias). In the era of eculizumab, allogeneic HSCT may domain transcriptional repression. Importantly, histone
still be offered to patients as the only definitive treatment, but hypoacetylation is reversible. Specifically, in the presence
in practice it will be carried out only in select patients (such of histone deacetylase inhibitors (HDACis) such as sodium
as those developing MDS; see later). butyrate, histone acetylation, PIGM transcriptional activity,
GPI biosynthesis, and surface GPI-linked protein expression
can all be restored in vivo as well as in vitro. This remarkable
PNH and other clonal disorders
effect was highlighted by the complete abrogation of compli-
Patients with PNH have a small risk (<4%) of developing cated, life-long, and treatment-refractory epilepsy in a child
MDS (the reverse, i.e. patients with MDS developing PNH, is with IGD treated with sodium butyrate.
discussed earlier) and AML. Because a similar risk exists in In a small number of patients with variable clinical
IAA and in the inherited BMFs, it is probably the perturbed phenotypes, whole-exome sequencing analysis has identi-
inflammatory marrow environment in conjunction with the fied several other germline mutations in genes of the GPI
SMs described earlier, rather than the PIGA mutations per se, biosynthetic pathway (Figure 11.2). As with PIGM, these
that allows the emergence of premalignant (MDS) or malig- patients have homozygous or biallelic mutations that result
nant (AML) clones, predisposing to MDS and AML. in partial deficiency of the corresponding protein. Clinical
manifestations comprise epilepsy, learning difficulties,
and multiple organ abnormalities, but not BMF, with the
Inherited GPI deficiency
exception of one reported case of PNH caused by a germline
Inherited glycosylphosphatidylinositol deficiency (IGD) mutation in PIGT accompanied by an SM in neutrophils
encompasses a group of diseases caused by heritable (Figure 11.2).
138 Molecular Hematology

Inherited BMF syndromes commonly of the radius and thumb), skin lesions (hyperpig-
mentation, café-au-lait spots), renal and urinary tract mal-
Fanconi anemia
formations, and gonadal dysfunction. However, the clinical
The incidence of FA is estimated at 1 : 360 000 in the gen- spectrum is even wider, as it includes congenital defects of the
eral population. Early studies indicated that FA was a geneti- gastrointestinal system, heart, and central nervous system.
cally heterogeneous disease, a notion confirmed by the iden- Patients with FA have an unusually high risk of develop-
tification, through the use of somatic cell hybridization and ing treatment-resistant MDS and AML, estimated at 52% by
latterly genome sequencing, of 21 complementation groups. the age of 40 years. Furthermore, the risk of a variety of solid
All 21 FA genes have now been cloned and characterized tumors, especially squamous cell carcinomas of the skin,
(Table 11.2). The overall frequency of heterozygotes for FA head, neck, and anogenital region, is several times higher
mutant genes in the general population is estimated at 1 in than in the general population. Patients with mutations in
300. In Ashkenazi Jews and in the Afrikaans population of BRCA2 or PALB2 usually develop AML or embryonal tumors
South Africa it is much higher (1 in 100 and 1 in 89, respec- in the first few years of life. While FA results from bial-
tively), most likely as a result of founder effects. lelic mutations, heterozygous mutations in BRCA1, BRCA2,
PALB2, and RAD51C are associated with increased risk of
Clinical aspects The most common clinical manifestation is solid tumors, in particular breast and ovarian cancer. This has
gradual onset of BMF. BMF typically appears by the age of implications for first-degree relatives of an individual with FA
10 years (median age 7 years; range birth to 31 years) and caused by one of these specific complementation groups.
is often heralded by thrombocytopenia, macrocytosis, and
increased hemoglobin F levels (the latter indicative of stress Cellular phenotype and function of FA proteins FA pro-
erythropoiesis). Congenital abnormalities are present in two- teins constitute the “FA pathway,” a DNA damage-response
thirds of patients and comprise skeletal abnormalities (most pathway responsible for repair of inter- and intrastrand
DNA cross-links (ICLs), either occurring spontaneously or
induced by agents like mitomycin C or diepoxybutane (Fig-
ure 11.3). ICLs prevent the separation of the two DNA
Table 11.2 Identity of the Fanconi anemia (FA) genes and proteins strands, leading to replication fork arrest and cell cycle arrest
in the late S phase. Completion of the cell cycle is not possi-
Approximate ble unless ICLs are repaired. FA proteins, organized in several
FA genes Chromosome frequency of FA (%) groups functioning in a linear fashion, are essential for suc-
FANCA 16q24.3 65 cessful resolution of the stalled DNA replication at the site of
FANCB Xp22.2 2 the ILC. The Fanconi anemia core complex (FCC), compris-
FANCC 9q22.3 12 ing nine proteins (FANCA, B, C, E, F, G, L, M, and T), receives
FANCD1/BRCA2 13q12.3 2 signals of DNA damage through phosphorylation by ATR, a
FANCD2 3p25.3 4 kinase sensor of DNA damage. This promotes binding of the
FANCE 6p21.3 1 FCC to the damaged DNA through FANCM and FAAP24.
FANCF 11p15 2 FANCL, through its ubiquitin ligase activity, monoubiquiti-
FANCG 9p13 8 nates FANCD2 and FANCI, the two proteins constituting the
FANCI 15q26.1 1
ID complex. FANCL interacts with UBE2T, a ubiquitin con-
FANCJ/BRIP1 17q23.1 2
jugating enzyme that is required for monoubiquitination of
FANCL 2p16.1 <1
FANCM 14q21.3 <1
FANCD2. Ubiquitination of the ID proteins along with their
FANCN/PALB2 16p12.1 <1 phosphorylation by ATR are required for their specific local-
FANCO/RAD51Ca 17q23 <1 ization to the site of DNA damage, where they interact with
FANCP/SLX4 16p13.3 <1 a further family of proteins.
FANCQ/ERCC4/XPF 16p13.12 <1 The finding that these proteins, corresponding to
FANCR/RAD51a 15q15.1 <1 FANCD1, FANCN, and FANCJ complementation groups,
FANCS/BRCA1a 17q21.31 <1 were in fact identical to BRCA2 and its associated PALB2
FANCT/UBE2Ta 1q32.1 <1 and BRIP1 proteins, respectively, was an exciting discovery
FANCU/XRCC2a 7q36.1 <1 linking the FA pathway to an already established process
FANCV/REV7/MAD2L2 1p36.22 <1
of DNA repair. In concert with a number of other proteins
a Loss
of the DNA repair machinery (e.g. RAD51, RAD51C,
of function mutations in these genes have been associated
XRCC2), BRCA1 and BRCA2 are involved in different
with Fanconi-like congenital abnormalities, but not with bone mar-
row failure.
repair processes that include nucleotide excision repair,
translesional synthesis, and homologous recombination, all
The molecular basis of bone marrow failure syndromes and red cell enzymopathies 139

F C
B
FC complex
E
M G
Downstream effectors of DNA repair
T L A +Ub
ID complex
DNA Polζ
damage ATR Ub N
Ub P Q S
(ICLs) V J
+P I
D2 Ub Ub
P Ub Ub
P R
I I
D2 D2 D1 U
P P O
P P

Fig. 11.3 Molecular pathogenesis of Fanconi anemia (FA). The FA pathway of DNA repair is activated in response to intra- or interstrand DNA
cross-links. First the DNA sensor ATR phosphorylates the Fanconi core complex (FCC) and ID complexes. Subsequently, the FCC monoubiquitinates
FANCD2 and FANCI. The ID complex is recruited to the site of DNA damage and acts in concert with DNA repair proteins. Specifically, FANCP/SLX4
and FANCQ/ERCC4 catalyze unlooping of the interstrand cross-links (ICL), allowing translesional synthesis by Polζ, one component of which is
FANCV/REV7. The final phase of ICL repair is mediated by the remaining DNA repair proteins, including FANCD1/BRCA2, FANCN/PALB2,
FANCJ/BRIP2, and FANCS.BRCA1 along with FANCR/RAD51 and its paralogs: FANCO/RAD51c and FANCU/XRCC2. Ultimately the coordinated
functions of these proteins leads to the repair of the DNA cross-link through nucleotide excision repair, translesional synthesis, and homologous
recombination.

of which are required for removal and repair of ICLs and allele is found in Ashkenazi Jews at a polymorphic frequency
restarting of DNA replication. Nucleotide excision repair (1 in 80), and it is responsible for 85% of the FA cases in this
is also performed by two proteins recently found to be population. Patients with IVS4 or exon 14 mutations tend
mutated in dyskeratosis congenita (DC): SLX4 and ERCC4. to have earlier onset of hematological complications (BMF
These form a complex that facilitates nucleolytic incisions and MDS/AML), and to have a shorter survival compared
at the ICL by the endonuclease ERCC4, paving the way for with patients with exon 1 mutations or patients with FANCA
homologous recombination. Taken together, a mutation or FANCG-related FA. Mutations in the FANCG gene are
affecting any one of the FANC proteins impairs DNA repair, found in about 10% of FA cases. The stop codon mutation
resulting in genomic instability in HSCs and, in turn, BMF. E105X accounts for 44% of mutations in German FANCG
patients. It is interesting that although biallelic null muta-
Molecular genetics FA is an autosomal recessive disorder, tions are very frequent in FANC genes of FCC, only hypo-
with the exception of FANCB, which is X-linked, and morphic mutations have been identified in FANCD2 (3–6%
FANCO/FANCR, which are autosomal dominant. The most of all cases of FA), highlighting a more important role of the
characteristic cellular feature of FA cells is the formation of ID complex that is not restricted to ICL correction only, but
DNA double-strand breaks on exposure to DNA inter- and also encompasses the repair of ionizing irradiation-induced
intrastrand adducting agents (clastogens), such as mitomycin double-strand DNA damage. Consistent with this, FANCD2
C and diepoxybutane. The in vitro response to clastogens patients, despite the hypomorphic nature of the mutations,
has made it possible to test cells from different patients for appear to develop the most severe form of FA, with multiple
their ability to cross-correct each other’s defect by somatic developmental abnormalities and early development of BMF
cell fusion (hence the 21 complementation groups). and malignancy. The prevalence of mutations in other FANC
Mutations of FANCA account for about 60% of FA cases genes is very low (<2%).
and are spread throughout the gene. None of the mutant alle-
les is common and few have been encountered more than FA and cancer An important role of the FA pathway in
once. Thus, identifying mutations in newly diagnosed cases the biology of sporadic cancers is increasingly being recog-
of FA where the complementation group is unknown involves nized. Epigenetic silencing of FANC genes has been iden-
comprehensive sequencing of multiple FANC genes in par- tified in various solid tumors and in acute leukemias; in
allel, an approach that has been facilitated by the recent some cases this correlates with their sensitivity to chemother-
access to rapid and economical high-throughput sequencing, apeutic agents, causing DNA cross-links such as cisplatin,
which allows the simultaneous, targeted sequencing of tens of cyclophosphamide, and melphalan. In a small proportion of
genes. patients with AML, SMs in FANCA have been identified, but
Mutations in the FANCC gene account for about 10–15% their functional significance is unknown. Therefore, studying
of FA cases. The IVS4 + A → T and del322G mutations com- the functional status of the FA pathway in different tumors
prise more than 75% of FANCC mutations. The IVS4 + A → T may offer valuable leads to therapeutic choices.
140 Molecular Hematology

Diagnosis The clastogen test (outlined earlier) remains the The clinical hallmarks of DC are lacy reticulated skin, nail
gold standard clinical diagnostic test; however, screening for dystrophy, and mucosal leukoplakia in the first years of life;
pathogenic mutations in the FANC genes has now entered and later the development of BMF. Other clinical manifes-
routine clinical practice. As a deficiency in any of the eight tations include developmental delay, short stature, ocular,
proteins of the FCC impairs its ubiquitin ligase activity, dental, and skeletal abnormalities, hyperhidrosis, hyperkera-
immunoblotting for ubiquitinated FANCD2 can also be used tinization of the palms and soles, bullae on minimal trauma,
as a screening test to focus the search within the FCC genes or hair loss, nail loss, sometimes gonadal failure, and features
in those acting downstream of the FCC. With this approach, of premature aging. Patients with DC are also at risk of pul-
genetic counselling, pre-natal, and pre-implantation diagno- monary fibrosis and hepatic failure. The clinical spectrum of
sis are now feasible for most families with affected children. DC is variable among patients and dynamic with age: some
In areas with a large Ashkenazi Jewish population (e.g. New patients manifest only minimal physical features and normal
York City), it is realistic to carry out screening for polymor- hematopoiesis, while others have the classical triad of skin,
phic FANC alleles on a wider population basis, so that it can nail, and mucosal abnormalities coupled with early-onset
be offered to all couples at risk. BMF. Like FA, DC predisposes to malignancy, in particular
MDS/AML and squamous cell carcinomas of the head, neck,
Treatment The conventional management of FA focuses on and anogenital tract. The median age of onset is 35 years for
the consequences of BMF and includes hematopoietic growth MDS and 37 years for solid cancers.
factors, blood product support, and androgens. About half of
patients respond to androgens initially, but often suffer from Molecular genetics and pathogenesis Eleven genes have been
significant side effects, including androgen-induced hepatic identified to cause DC when mutated (Table 11.1). Mutations
adenomas. Eventually all patients become refractory. occur de novo, and the diseases they cause are inherited as X-
HSCT from an HLA-identical unaffected sibling or from linked recessive, autosomal dominant, or autosomal recessive
alternative sources is currently the only therapeutic approach traits.
that can successfully achieve long-term correction of BMF The X-linked form of DC was the first to be identified
and possible prevention of MDS and AML. The best results, and characterized: it results mostly from missense mutations
with reduced short- and long-term treatment-related mortal- in the housekeeping gene DKC1. Mutations in DKC1, which
ity, are obtained with fludarabine-based, reduced-intensity, maps to Xq28 (like G6PD and hemophilia A), account for
non-myeloablative conditioning HSCT regimens that do not about 40% of all DC cases. Hoyerall–Hreidarsson syndrome,
include irradiation (to reduce the risk of solid tumors). T a rare disorder characterized by severe growth failure, severe
cell depletion of the graft is employed to minimize graft- immunodeficiency, cerebellar abnormalities, and BMF, is in
versus-host disease. Unfortunately, even subsequently HSCT most cases allelic to DKC1.
patients remain at increased risk of developing solid tumors, Dyskerin, the protein product of DKC1, is homologous
and thus require close long-term follow-up as well as pre- to the yeast protein Cbf5p, which, in conjunction with the
ventative strategies such as human papilloma virus (HPV) H/ACA class of small nucleolar RNAs, is required for the
vaccination. pseudouridylation (i.e. conversion of uridine to pseudouri-
The cloning of FA genes has paved the way for gene ther- dine) of pre-ribosomal RNA (rRNA), a modification essential
apy for patients with FA. There is clinical and experimental for ribosome biogenesis. Although defective pseudouridyla-
evidence that HSCs with corrected phenotype have a survival tion and ribosome biogenesis have been observed in a hypo-
and growth advantage over uncorrected cells and can support morphic DKC1 mouse model, other studies failed to demon-
long-term hematopoiesis. Successful transfer of FA genes to strate any qualitative or quantitative defect of rRNA in DC
a small number of autologous HSCs should therefore be ade- patients. Instead, it was shown that dyskerin binds, via its
quate to ameliorate the severity of BMF. domain called PUA, to an H/ACA domain of the telom-
erase RNA component (TERC); that is, the template used by
its protein component, the telomerase reverse transcriptase
Dyskeratosis congenita (TERT). Thus, dyskerin is intimately linked to telomerase in
DC is another rare (incidence approximately 1 in 1 million), its crucial function of telomere maintenance during cell divi-
genetically heterogeneous, inherited disorder with BMF as a sion. Most DKC1 mutations map to the PUA domain, linking
major feature. DC is primarily a disorder of telomeres – it DC to abnormal RNA binding.
could be said to belong to the telomeropathies. Telomeres, Telomerase, as just outlined, is a ribonucleoprotein com-
the ends of chromosomes, are made of several megabases of plex, and dyskerin is one of the several proteins which, by
DNA consisting of identical nucleotide repeats (TTAGGG), interacting directly or indirectly with TERC, preserve its sta-
and of a protein complex essential for chromosome integrity bility and its ability to function as a template for TERT.
(Figure 11.4). Telomerase activity is highest in rapidly dividing cells (e.g.
The molecular basis of bone marrow failure syndromes and red cell enzymopathies 141

Shelterin complex Telomerase complex


protection of telomere ends telomere elongation

Telomere trafficking
PARN TERC processing
WRAP53
0 R
P1 GA
ACD NO
P2
TIN2 NH
POT1 TERT DKC1
G strand T T 3’
R R TTAGGG
F F AAUCCC
C strand 2 1
5’
5’
P1 TERC
RA STNI
RNA template
CTC1
TENI
Telomere replication
RTELI
+ stability
CST complex
C strand synthesis

Telomeric DNA

Telomere

Centromere

Telomere
Fig. 11.4 Schematic representation of human proteins involved in telomere maintenance. TERC encodes a non-coding RNA and no
protein. This RNA template interacts with TERT and other members of the telomerase complex to elongate telomeres by adding hexanucleotide
tandem repeats to the ends of chromosomes. The allied roles of the components of the CST and shelterin complexes are shown. Shaded proteins
are those that, when affected by germline mutations, cause dyskeratosis congenita (DC).

in hematopoietic progenitors, in tumor cells, during organo- TINF2 have been associated with the Revesz syndrome, a
genesis, and in early life). The critical evidence that DC is a form of DC characterized by bilateral exudative retinopathy.
disease of telomere dysfunction was provided by the find- Patients with monoallelic mutations are affected, and there-
ing that heterozygous mutations in the TERT and TERC fore the condition is designated as autosomal dominant; how-
genes themselves cause a disease virtually indistinguishable ever, those with biallelic mutations have far more severe dis-
from that caused by DKC1 mutations. Once again, not unlike ease. Thus, all types of DC are related to a common molecular
FA, the multitude of genes whose mutations can give rise pathogenetic pathway, namely the maintenance of telomere
to a dyskeratosis phenotype relates to the complexity of length. In keeping with this notion, all types of blood cells
the telomere maintenance/servicing machinery (see Table in DC patients have significantly shorter telomeres than age-
11.1 and Figure 11.4). NOP10 and NHP2, members of the matched controls. Shortening of telomeres to a critical length
H/ACA snoRNPs family, localize like DKC1 itself to nucleoli can result in either (i) growth arrest and apoptosis – hence
and to Cajal bodies in the nucleus. WRAP53 assists with the the development of BMF; or (ii) neoplastic transformation –
transportation of TERT to the Cajal bodies, and PARN facili- hence the increased propensity to epithelial tumors.
tates the interaction between TERC and the telomere. RTEL1 The study of the genetics of DC has revealed at least three
encodes a DNA helicase required to unwind the T loop struc- additional interesting features. First, in families with TERC,
ture, a prerequisite for telomere lengthening, whereas CTC1 TERT, and TINF2 mutations, DC has tended to show earlier
forms part of the CST complex that caps and protects telom- onset and more severe manifestations in successive genera-
eres. Finally, TINF2 (encoding TIN2) and ACD encode pro- tions, a phenomenon known as genetic anticipation. In keep-
teins that form part of the so-called shelterin complex, which ing with this, the rate of loss of telomere length was higher
binds to telomeric DNA and provides stability. Mutations in in successive generations within the same DC family (just as
142 Molecular Hematology

had been observed in Terc−/− mice). Second, heterozygous pathway (linked to the shelterin telomere capping complex)
germline mutations of TERT or TERC have been detected in was essential for intestinal stem cell renewal, and that treat-
a small minority (<5%) of patients who had been initially ment with Wnt agonists corrected the DC phenotype and
diagnosed with IAA. In many cases these mutations were restored telomere length to normal. Thus, the recent remark-
not obviously deleterious for the function of telomerase in able advances in genome editing approaches may lead to the
vitro; in some cases they were not detected in the parents of future development of gene therapy for DC and possibly for
index cases and in others they did not segregate with the dis- other BMF syndromes.
ease in the respective families. A possible interpretation of
these findings is that mild TERC and TERT mutations may Diamond–Blackfan anemia
predispose to BMF; however, this develops only upon expo-
sure to a trigger, such as an immunological attack. Third, Diamond–Blackfan anemia (DBA) is a rare (1–2 in 100 000
in patients with pathogenic mutations of TERT (and in one live births) inherited syndrome with isolated anemia and ery-
case of DKC1), somatic mosaicism has been demonstrated, throblastopenia as the main features, although with disease
whereby the mutation is found in DNA from skin fibroblasts progression patients may develop bi- or trilineage hypopla-
but not in DNA from peripheral blood cells. The presumed sia. Clinically DBA manifests itself in the first months of
mechanism is genetic reversion through back-mutation in a life with macrocytic anemia, reticulocytopenia, increased
single hematopoietic cell. The self-corrected cell would have hemoglobin F, and elevated erythrocyte adenosine deami-
a significant selective advantage, in some cases sufficient to nase (eADA) levels. It is not known why the anemia asso-
repopulate the bone marrow, an example of nature’s gene ciated with DBA rarely manifests during fetal life. The bone
therapy. marrow is normocellular, with a characteristic paucity of
erythroblasts (<5% of nucleated cells). Growth retardation
Diagnosis DC is characterized by very short telomeres, is seen in about 30% of patients, and other developmen-
defined as <1% of age-matched normal controls, in the tal abnormalities (including skeletal, cardiac, and urogenital
majority of leukocyte subsets. Measurements are performed defects) in 50% of patients. There is also an increased risk
by multiparameter flow cytometry fluorescence in situ (∼20% by age 46 years) of MDS/AML or solid tumors, mainly
hybridization (flow-FISH). In lymphocytes, sensitivity and osteogenic sarcoma.
specificity for DC are 97% and 91%, respectively. Genetic
testing is desirable to confirm diagnosis, but pathogenic Molecular genetics and pathogenesis In most cases, DBA is
germline mutations are detected in only about 70% of caused by a monoallelic, loss-of-function ribosomal protein
patients with a clinical diagnosis of DC, either because the (RP) gene mutation. DBA is therefore one of the “riboso-
panel of genes is incomplete or because some causative genes mopathies,” a group of inherited BMF syndromes also com-
are still unknown. TERT and TERC mutations correlate with prising Shwachman–Diamond syndrome, X-linked DC, and
IAA, pulmonary fibrosis, and adult-onset disease. cartilage hair hypoplasia (Table 11.1). In 55–60% of cases
mutations are de novo, whereas in the remainder they are
Treatment As for FA, there is no definitive treatment for DC, transmitted in an autosomal dominant fashion.
and allogeneic HSCT is the only curative option for severe The first pathogenic genetic lesion identified was a het-
BMF or MDS/leukemia. HSCT in DC has an increased risk erozygous mutation in RPS19, the most common (25% of
of graft failure, graft-versus-host disease, pulmonary fibro- cases) gene involved in DBA. The finding that the whole
sis, hepatic cirrhosis, and veno-occlusive disease. If a related RPS19 gene is deleted in some cases of DBA strongly argued
donor is unavailable, one can try therapy with androgens. that haploinsufficiency, rather than a dominant negative
The mechanism of action of these agents is thought to be effect, is the underlying pathogenetic mechanism in DBA.
direct upregulation of TERT gene expression with conse- As well as being a structural component of the mature small
quent increased telomerase activity. Most patients with DC ribosomal subunit (40S; the large subunit is 60S), RPS19 is
die before the age of 30 years, predominantly from complica- also required in the nucleolus for the correct processing and
tions of BMF, opportunistic infections, pulmonary complica- cleavage from the precursor rRNA of the 18S fragment, which
tions, and malignancy. is the main rRNA constituent of 40S. In the same process,
In a recent study, human induced pluripotent stem cells which is RPS19 independent, precursor rRNA is also cleaved
(iPSCs) were generated from skin fibroblasts of a DC patient to 5.8S and 28S rRNA, which, along with the independently
and differentiated to intestinal cells. Correction of the disease transcribed 5S rRNA, are the constituents of the large ribo-
allele was performed with clustered regularly interspaced somal subunit. Haploinsufficiency of RPS19 and of the other
short palindromic repeats (CRISPR)-Cas9-mediated homol- RP genes linked to DBA results in defective formation of
ogous recombination. Examination of the resultant isogenic the ribosomal subunits and eventually defective formation
DC and control cell lines showed that the Wnt signaling of the mature ribosome. How impaired ribosome function
The molecular basis of bone marrow failure syndromes and red cell enzymopathies 143

might lead to the relatively selective phenotype of erythroid underlying the development of DBA, we are yet to elucidate
hypoplasia is not clear. It is possible that this is related to the the molecular basis of the clinical heterogeneity observed in
extraordinary demand that hemoglobin production imposes this disease, including the remarkably variable penetrance
on the translational apparatus. In addition, many RPs, includ- within families.
ing RPS19, have been found to participate in extra-ribosomal
functions. For example, RPS19 might affect cell cycle progres- Diagnosis Diagnostic criteria for DBA include the presence
sion through its interaction with the serine/threonine kinase of classical hematological and clinical features and identifica-
Pim-1; whereas RPL11 and RPL5 stabilize and activate p53 tion of a causative gene mutation. The presence of congeni-
through an inhibitory physical association with the p53 reg- tal anomalies, age <1 year, raised eADA, and macroytosis all
ulator MDM2. Clarification of the mechanisms by which per- point toward a diagnosis of DBA, rather than transient ery-
turbed ribosome biogenesis leads to fewer erythroid progen- throblastopenia of childhood (TEC), the main differential.
itors with impaired function will be facilitated by the recent This is an important distinction, as the latter condition usu-
immunophenotypic characterization of early and late human ally resolves spontaneously within a few months.
erythroid progenitors. In specialized DBA centers, genetic diagnosis using
Since the identification of RPS19, several other RP genes targeted next-generation sequencing of RP genes has
have been implicated in DBA, including RPS24, RPS26, replaced traditional Sanger sequencing. This approach,
RPS10, RPS17, RPL5, RPL35A, and RPL11 (Table 11.3). A complemented by additional techniques such as multiplex
wide variety of mutations have been described, including ligation-dependent probe amplification for validation of
missense, nonsense, splicing, and whole allele deletions. Of computationally identified deletions, has led to an improve-
note is that DBA resulting from an RP gene deletion can ment in the pick-up rate of mutations from 40% to around
occur in the context of other syndromes, for example the 70% of patients, and to the identification of new candidate
microdeletion 3q29 syndrome, in which the deleted region RP genes (Table 11.3).
encompasses RPL35A. Mutations in the promoter region of Interestingly, a recent study discovered that 8 of 173
RPS19 have also been observed, and it is likely that more vari- patients with presumed DBA but no pathogenic RP gene
ants in regulatory areas will be implicated in DBA as knowl- mutation were in fact harboring mitochondrial DNA dele-
edge of the function of the non-coding genome expands. In tions similar to those seen in the Pearson marrow-pancreas
the last few years two X-linked genes associated with DBA syndrome. Therefore, while we have addressed each of the
have been discovered: GATA-1 (an erythroid-specific tran- inherited BMF syndromes separately, it is important to rec-
scription factor) and TSR2 (a RP chaperone). While great ognize that they share a triad of clinical features: anemia,
progress has been made in characterizing the genetic lesions physical abnormalities, and predisposition to cancer. Given
the overlap between these conditions and the accessibility of
genetic testing in routine clinical practice, a child presenting
with congenital anemia should be ideally tested for multi-
Table 11.3 Genes mutated in Diamond–Blackfan anemia (DBA)
ple conditions, using a panel encompassing multiple genes.
Gene name Genetic locus % of cases of DBA Furthermore, cases in which a mutation is not identified by
targeted multigene assays are candidates for whole-genome
RPS19 19q13.2 25 sequencing with the aim of novel gene discovery, as exempli-
RPL5 1p22.1 6.6 fied by the Genomics England 100 000 Genomes Project.
RPS26 12q13.2 6.4
RPL11 1p36.11 4.8
Treatment Blood transfusion, combined with iron chelation,
RPL35A 3q29 3
RPS10 6p21.31 2.6
is the mainstay of treatment for moderate to severe anemia
RPS24 10q22.3 2 in DBA. After regular immunizations with live vaccines
RPS17 15q25.2 1 are completed during infancy, most patients are started on
RPS7 2p25.3 <1 high-dose glucocorticoids (GC). Initially 80% of patients
RPS29 14q21.3 <1 respond, with a surge in reticulocytes followed by a rise
RPL26 17p13.1 <1 in hemoglobin, to the extent that the patient may become
RPL15 3p24.2 <1 transfusion independent. However, not infrequently GC
RPL31 2q11.2 <1 treatment is discontinued, either because the response is lost
RPS27 1q21.3 <1 (this can happen with puberty or pregnancy), or because at
RPS28 19p13.2 <1
the dose required to maintain therapeutic benefit the side
RPL27 17q21.1-21.2 <1
effects are unacceptable. In the long term only 20–40% of
GATA-1 Xp11.23 <1
TSR2 Xp11.22 <1
patients remain on a very low dose of GC; a further 10–20%
seem to go on to spontaneous remission by the age of 25.
144 Molecular Hematology

Patients who are steroid resistant, and those with severe iron kinase (PK), glucose-6-phosphate dehydrogenase (G6PD),
overload or progressive BMF, must be considered for allo- and pyrimidine 5′ -nucleotidase –representative of disorders
geneic HSCT. Ideally this should be performed before the age of glycolysis, the pentose phosphate pathway, and nucleotide
of 10 years. At present there is convincing evidence to recom- metabolism, respectively.
mend sibling donors, and promising results from unrelated
donors (either haploidentical or umbilical cord blood). Clinical features
Recent experimental evidence indicated that, through acti-
vation of the rapamycin-sensitive mTOR/RPS6 pathway, the In these disorders hemolytic anemia occurs with varying
amino acid L-leucine enhances ribosomal translational activ- degrees of severity. It is not unusual for the presentation to
ity in DBA cells with RPS19 mutations. As yet, a clinically be in the guise of severe neonatal jaundice that may require
effective treatment for patients with DBA has failed to evolve exchange transfusion; if the anemia is less severe it may
from these findings. One of the most clinically important present later in life, or it may even remain asymptomatic and
unsolved riddles in DBA is the mechanism of action of be detected incidentally when a blood count is performed for
GC. A recent study using chromatin immunoprecipitation unrelated reasons. The spleen is often enlarged. When other
sequencing in normal murine fetal liver erythroid progeni- systemic manifestations occur, they involve the central ner-
tors showed a physical interaction between PPARalpha and vous system, sometimes entailing severe mental retardation,
glucocorticoid receptor (GR), the transcription factor that or the neuromuscular system, or both.
mediates GC effects on gene transcription. It seems reason-
able to expect that as a follow-up to this and other studies, Diagnosis
the riddle of the GC effect will be resolved in the near future;
this will facilitate the development of novel therapies directly The diagnosis of hemolytic anemia is usually not diffi-
targeted to erythropoiesis, which will be effective in steroid- cult, thanks to the triad of normo-macrocytic anemia,
resistant patients and/or synergize with GC, so that their dose reticulocytosis, and hyperbilirubinemia. Enzymopathies
can be reduced. The experiment just mentioned suggests a should be considered in the differential diagnosis of any
possible therapeutic role for PPARα agonists in DBA. chronic Coombs-negative hemolytic anemia. A definitive
As supportive therapy improves and patients with DBA diagnosis can be made only by demonstrating the deficiency
can expect longer lives, it becomes increasingly important to of an individual enzyme by a quantitative assay. For the sake
understand the drivers of malignant transformation in a sub- of economy, it is sensible to carry out these rather laborious
set of patients. The relevance of ribosome biology to diseases tests in order of frequency of occurrence of the various
far more common than DBA has been highlighted recently by enzymopathies: first glucose-6 phosphate dehydrogenase
the discovery of RP gene mutations in hematological malig- (G6PD), then PK, then glucose 6-phosphate isomerase, and
nancies (specifically in T-cell acute lymphoblastic leukemia so on (Table 11.4). If a particular molecular abnormality is
and in chronic lymphocytic leukemia). In many cases these already known in the family, then of course one could test
somatically acquired mutations interfere with the function of directly for that at the DNA level, bypassing the need for
the tumor suppressor p53. enzyme assays. As for other multigenic heritable disorders,
the advent of high-throughput sequencing technologies
allows simultaneous analysis of multiple genes implicated in
Red cell enzyme deficiencies glycolytic enzyme deficiencies, thus simplifying the genetic
diagnosis of these disorders. Nevertheless, enzymatic assays
Inherited abnormalities of red cell enzymes, red cell enzy- should still be performed to validate candidate pathogenic
mopathies, are a distinct set of genetic disorders with one mutations.
important clinical manifestation in common, namely chronic
hemolytic anemia (acute haemolytic anaemia in the case
Molecular pathophysiology
of G6PD deficiency). This section deals with those enzy-
mopathies affecting red cell metabolism for which the molec- Most of the glycolytic enzymes involved are housekeeping
ular basis has been elucidated (Table 11.4). We do not discuss enzymes present, by definition, in all cells. Therefore, one
conditions in which an enzyme abnormality is also expressed might expect that a severe reduction in activity of any
in red cells but the main clinical manifestations are elsewhere of these might have generalized clinical manifestations.
(e.g. the porphyrias, galactosemia, Lesch–Nyhan syndrome). However, we can identify at least two reasons why red cells
All of these defects, except G6PD deficiency, are rare to very are more severely affected. First, red cells have a much more
rare. For the sake of brevity, we will give a brief overview limited metabolic machinery than most other somatic cells;
of enzymopathies, then focus on deficiencies of pyruvate if a particular enzyme is deficient, other cells may cope by the
The molecular basis of bone marrow failure syndromes and red cell enzymopathies 145

Table 11.4 Synopsis of red cell enzymopathiesa

Isoenzymeb Prevalence of Main clinical features


characteristic enzyme associated with Benefit from Chromosomal
Enzyme (abbreviation) of red cells deficiency enzyme deficiencyc splenectomyd localization

Adenylate kinase (AK) 1 Very rare HA, CNS Partial 9q34.1


Aldolase A Very rare HA, myopathy 16q22–q24
Cytochrome b5 reductase Rare Pseudocyanosis, CNS 22q13.31–qter
2, 3-Diphosphoglycerate Very rare Polycythemia 7q31–q34
mutase (DPGM)
Enolasee 1 (α) Very rare HA 1pter–p36.13
Glucose 6-phosphate B Common HA None Xq28
dehydrogenase (G6PD)
Glucose 6-phosphate isomerase Rare HA, NM, CNS Partial 19q13.1
(GPI)
γ-Glutamylcysteine synthetase Very rare HA, CNS 6p12
(GLCLC)f
Glutathione peroxidase Very rareg ?g 3q11–q12
(GSH-Px)
Glutathione reductase (GSR) Very rare HA 8p21
Glutathione synthetase (GSS) Very rare HA, CNS 20q11.2
Glyceraldehyde 3-phosphate Very rare HA 12p13.31–p13.1
dehydrogenase (GAPD)e
Hexokinase (HK) R (I) Very rare HA Partial 10q22
Lactate Dehydrogenase-A Very rare Myopathy 11p15
(LDHA)
Monophosphoglycerate mutase B Very rare Myopathy and 10q25.3
(PGAM-B) myoglobinuria
Phosphofructokinase (PFK)h M Very rare HA, myopathy 12q13
L 21q22.3
Phosphoglycerate kinase (PGK) 1 Very rare HA, CNS, NM Partial Xq13
Pyrimidine 5′ -nucleotidase Rare HA Partial 7p15–p14
(P5′ N1)
Pyruvate kinase (PK) Ri Rare HA Partial 1q21
Triosephosphate isomerase (TPI) Very rare HA, CNS, NM, None 12p13

a We have listed all enzymes in the intermediary metabolism of red cells for which, to the best of our knowledge, the corresponding cDNA/gene
has been cloned.
b No entry in this column means that there are no known isoenzymes; therefore it is assumed that the same enzyme type is present in all tissues.
c CNS, central nervous system involvement; HA, hemolytic anemia; NM, neuromuscular manifestations.
d Data available only on some patients.
e Since no mutations have yet been reported, there is no formal proof that HA associated with this enzyme deficiency is due to mutation of the

corresponding gene.
f γ-Glutamylcysteine synthetase consist of two subunits, a catalytic subunit and a regulatory subunit. The data concerning the catalytic subunit

are shown here.


g Enzyme reported to be low in some nutritional deficiencies, but clear evidence that inherited deficiency of glutathione peroxidase exists is

lacking.
h PFK in normal red cells consists of a mixture of the five tetrameric species that can be formed from random association of the M (muscle) and

L (liver) highly homologous subunits (i.e. M4, M3L, M2L2, ML3, L4).
i The red cell form of PK called R is produced by the gene encoding the L (liver) subunit. Because a different promoter is used, the size of liver PK

is 543 amino acids.


Source: Modified from Luzzatto L., Notaro R. (1998) Red cell enzymopathies. In Jameson J.L. (ed.), Principles of Molecular Medicine. Clifton, NJ:
Humana Press, pp. 197–207, with permission.
146 Molecular Hematology

use of alternative or surrogate metabolic pathways. Second, homozygotes or compound heterozygotes with biallelic
mature red cells lack nuclei and organelles, thus are not mutations. From the hematologic point of view, it is inter-
capable of protein synthesis; therefore, if a particular enzyme esting that PK-deficient patients, after splenectomy, have
is made highly unstable by a mutation, other cell lineages reticulocytosis to a degree (up to 50%) very seldom seen in
can compensate by increased enzyme synthesis, but red cells any other human condition; this has led to the suggestion
cannot. that there may be a specific mechanism for the spleen to
remove PK-deficient reticulocytes from the circulation.
Another interesting feature of PK deficiency is that 2,3-
Management
bisphosphoglycerate (BPG) levels are high (up to three times
There is no specific treatment for these conditions. Patients normal): this produces a right shift in the hemoglobin–
with moderate anemia may require occasional blood transfu- oxygen (O2 ) dissociation curve, thus increasing O2 delivery
sion when they experience exacerbations of the anemia due to tissues, which partially compensates for anemia.
to increased rate of hemolysis or to decreased red cell pro-
duction secondary to infection (the most extreme example
Molecular genetics
being aplastic crisis from parvovirus infection). Patients with
chronic severe anemia may require regular blood transfusion Over 200 mutations have been implicated in PK deficiency
therapy, and in some patients splenectomy has been benefi- and, as is the case for other glycolytic enzymes, most muta-
cial (Table 11.4). tions are of the missense type, causing single amino acid
replacements. This is important, because the low level of
residual enzyme activity can still support some metabolic
Pyruvate kinase deficiency
flow through the glycolytic pathway, which explains how red
PK deficiency is the most common defect of the glycolytic cells survive in the circulation, even though their lifespan is
pathway causing chronic non-spherocytic hemolytic anemia reduced. With respect to how precisely individual missense
(CNSHA): it has an estimated frequency of 1 in 20 000 in mutations reduce enzyme activity, we must consider at the
people of European ancestry, and a higher frequency in the protein level several possible mechanisms:
Old Order Amish population of Pennsylvania. The clinical 1 In the majority of cases, loss of activity is probably due to
phenotype is variable, from hydrops fetalis (non-immune) to decreased stability of the protein. In such cases we would pre-
mild anemia presenting in the second or third decade. There- dict that, compared to erythrocytes, other cells, e.g. hepato-
fore, underdiagnosis of PK deficiency occurs at both ends of cytes, might be much less affected, because they can compen-
the clinical spectrum. sate for decreased stability through increased synthesis of the
enzyme.
2 In some cases the amino acid replacement, being within
Pathophysiology
or near the active center of the enzyme, may affect substrate
PK catalyzes the second adenosine triphosphate (ATP)- binding (Km ), or catalytic rate (Kcat ), or both.
producing step of the glycolytic pathway; thus, it accounts 3 In some cases the allosteric regulation by FBP may be
for nearly 50% of the RBCs’ chemical energy in the form affected (altered Ka ).
of ATP (see Figure 11.5 for an overview of glycolysis). The In cases (2) and (3) not only red cells but other cells, in
activity of PK, the last reaction in the anaerobic glycolytic which the rate of glycolysis is critical, will be affected as
pathway, is allosterically regulated by the product of one well. Among severe missense mutations of this type, one
of the first reactions of the pathway, namely fructose-1,6- of the most common in Caucasians is 994A (Gly332Ser),
bisphosphate (FBP). There are four PK isoforms (M1, M2, which can, when homozygous, cause intrauterine death.
L, and R) encoded by two separate genes (PK-M and PK-LR). Cloning of the PKLR gene in an expression vector and
Alternative splicing of the PK-LR transcript encodes PKL and purification of recombinant normal and mutant human PKR
PKR in the liver and RBCs, respectively. enzyme has facilitated study of the biochemical impact of
In PK deficiency, like in other glycolytic enzymopathies, single amino acid substitutions. Of course, as with most
a shortage of energy supply leads to shortened RBC lifespan inherited disorders, the correlation between genotype and
and RBC destruction, mainly in the spleen. Glycolytic phenotype based on the molecular properties of the enzyme
enzymes are normally present in cells in considerable is only a first approximation, because other inherited and
excess, and therefore the 50% loss of enzyme activity seen acquired factors regularly come into play. Whereas most PK
in heterozygotes does not become limiting for red cell mutations are sporadic, it was found in sub-Saharan Africa
survival; thus, heterozygotes for PK deficiency do not have that one polymorphic mutation (E277K) has heterozygote
hemolytic anemia. Indeed, the disease shows an autosomal frequencies up to 7%, and it has been suggested it may have
recessive pattern of inheritance: affected patients are either been biologically selected by malaria.
The molecular basis of bone marrow failure syndromes and red cell enzymopathies 147

Glucose
ATP
HK
ADP G6PD
Glucose 6-phosphate 6-phosphogluconate

GPI NADP NADPH

Fructose 6-phosphate
ATP
GSH GSSG Pentose
PFK
phosphates
ADP ADP
GSS
Fructose 1,6-bisphosphate
ATP Gly
Aldolase γGluCys

TPI
Cytochrome b5
Dihydroxyacetone Glyceraldehyde
reductase
phosphate 3-phosphate
NAD + Pi HbFe2+
GAPD
NADH HbFe3+
1,3-diphosphoglycerate
ADP
PGK DPGM
2,3-DPG
ATP
Fig. 11.5 The Embden–Meyerhof pathway for
anaerobic glycolysis and the pentose phosphate 3-phosphoglycerate
pathway for oxidative glycolysis, as well as related
reactions (not the complete metabolic machinery PGAM
of the red cell). Enzymes are enclosed in rounded
2-phosphoglycerate
boxes. Abbreviations as in Table 11.4. Additional
abbreviations: DPG, diphosphoglycerate; GSH, Enolase
reduced glutathione; GSSG, glutathione; HbFe2+, H2O
hemoglobin; HbFe3+, methemoglobin; γGluCys, Phosphoenolpyruvate
γ-glutamylcysteine.
ATP
Source: Luzzatto L., Notaro R., (1998). Red cell PK
enzymopathies. In Jameson J.L. (ed.), Principles of
ADP
Molecular Medicine. Clifton, NJ: Humana Press, pp.
197–207. Reproduced with permission. Pyruvate Lactate

Diagnosis may be required in equivocal cases. Prenatal diagnosis of


at-risk fetuses is also feasible, by amniotic fluid or chorionic
PK deficiency is suggested by the constellation of clinical
villi sampling, followed by molecular testing of the PKLR
features and hematological indices; unlike in other glycolytic
gene.
enzymopathies, red cell morphology can be also suggestive,
with a multitude of cells displaying spiculae on their surface.
Management
As with all autosomal recessive disorders, there may be no
family history. Diagnosis is confirmed by finding in red cells The mainstay of therapy is supportive, with phototherapy
reduced PK enzymatic activity, which is usually 10–25% of and/or exchange transfusion in the newborn period, and
normal in patients with biallelic mutations. Of note is that, regular or intermittent red cell transfusions in children and
since routine enzyme tests measure Vmax without regard to adults. Iron chelation must be sometimes recommended
Km or Ka , enzymatic activity correlates poorly with the sever- even in patients who are not transfusion dependent, because
ity of hemolysis, and may even be normal. Mutation analysis with PK deficiency there is a propensity for iron overload.
148 Molecular Hematology

Splenectomy (invariably total) can help to reduce transfusion hemolysis is characteristically exacerbated by the same agents
requirement. In severe cases, provided there are no systemic that can cause AHA in people with the ordinary type of G6PD
manifestations other than hemolytic anemia, bone marrow deficiency.
transplantation can be considered; of course, it should be car-
ried out, if at all, before there is organ damage (e.g. from iron Diagnosis
overload).
In individuals with clinically apparent G6PD-deficiency
The development of more specific therapies has been
(type 3 in the list), the anemia is usually normocytic and
guided by structural analysis of the PKR enzyme. AG-348, a
normochromic, and its severity ranges from moderate to
novel first-in-class small molecule, has been designed to bind
extremely severe. The morphology of red cells is not char-
to the allosteric FBP site of PKR so as to simulate activation by
acteristic (non-spherocytic). AHA (in G6PD deficiency of
FBP, and thus functionally restoring the activity of a mutant
type 1) is due to intravascular as well as extravascular hemol-
enzyme to a normal or near-normal level. In an early clini-
ysis, and hence is associated with hemoglobinemia and
cal trial, this orally administered small molecule resulted in
hemoglobinuria. Here the blood film shows instead rather
clinically meaningful improvement in nearly 50% of patients
spectacular evidence of hemolysis in the guise of anisocy-
with PK deficiency, in some cases with normalization of the
tosis, polychromasia, spherocytes, bite cells, blister cells, and
hemoglobin level.
hemighosts. Supravital staining reveals the presence of Heinz
bodies, consisting of precipitates of denatured hemoglobin.
Glucose 6-phosphate dehydrogenase Definitive diagnosis relies on the direct demonstration of
deficiency decreased activity of G6PD in red cells by an appropriate
enzyme assay. Of note is that during an acute crisis the level
G6PD deficiency is by far the most common abnormality
of enzyme may be overestimated due to reticulocytosis, while
occurring in the pentose phosphate pathway. It affects 200–
at the same time the oldest (most G6PD-deficient) red cells
300 million individuals worldwide, with a high prevalence
have been destroyed.
in populations of Africa, southern Europe, the Middle East,
Southeast Asia, and parts of Oceania, as well as in regions
to which migrations from these areas have taken place. The Molecular genetics
overall geographical distribution of G6PD deficiency and its G6PD is a homodimeric or homotetrameric molecule, and its
heterogeneity, together with clinical field studies and in vitro single subunit is encoded by an X-linked gene that maps to
culture experiments, strongly support the view that this com- Xq28. The tetramer, a dimer or the dimer, is in equilibrium
mon genetic trait has been selected by Plasmodium falci- with the dimer itself. Of the 187 mutations reported in the
parum malaria, by virtue of the fact that it confers relative G6PD gene, most are point mutations causing single amino
resistance against this highly lethal infection. Delineation of acid substitutions. It is therefore likely that complete absence
the molecular mechanism underlying this resistance could of G6PD would be lethal; this has been proven in the mouse
be used to develop new approaches to protect G6PD-normal (see later). As a result of the phenomenon of X-chromosome
individuals against malaria. inactivation in somatic cells (lyonization), female heterozy-
gotes are genetic mosaics, in whom approximately one-half
Clinical features of the red cells are normal and approximately one-half are
G6PD deficient. However, there is a wide distribution around
Three types of clinical presentation are well characterized: this mean, and in some cases the skewing is extreme. There-
1 The vast majority of G6PD-deficient people are asymp- fore clinical manifestations, such as favism, can occur in both
tomatic most of the time, but they are at risk of developing hemizygous males and heterozygous females, but they tend
acute hemolytic anemia (AHA), which may be triggered by to be milder in the latter, roughly in proportion to the frac-
drugs (e.g. primaquine, rasburicase), infections, or ingestion tion of red cells that are G6PD deficient. This correlation
of fava beans. between enzyme activity and clinical phenotype has been for-
2 The risk of developing neonatal jaundice is much greater mally demonstrated in a study of 200 heterozygous children
in G6PD-deficient than in G6PD-normal newborns. This exposed to dapsone.
is of great public health importance, because untreated
severe neonatal jaundice can lead to permanent neurological
Function of G6PD
damage.
3 CNSHA: in contrast to the first two, this clinical pre- In intermediary metabolism G6PD is aptly depicted as
sentation is very rare. The clinical picture is rather simi- the first step in the pentose phosphate pathway. However,
lar to CNSHA associated with glycolytic enzymopathies (see several lines of evidence indicate that its most essential role
earlier) and again it is of variable severity. However, the is not to produce pentose, but rather to provide reductive
The molecular basis of bone marrow failure syndromes and red cell enzymopathies 149

potential in the form of NADPH. NADPH is necessary for Molecular pathophysiology


the replenishment of GSH when it is oxidized by reactive
AHA is seen with most of the variants of G6PD whereby
oxygen species (ROS). These are generated endogenously,
red cells retain some 10% of normal G6PD activity,
all the time in the course of deoxy hemoglobin cycling, at a
resulting in a limited capacity of these cells to withstand
low rate; and at a much higher rate from the metabolism of
the oxidative action of an exogenous factor. In contrast,
pro-oxidant compounds, such as primaquine or substituted
in a subset of variants (referred to as class I variants), the
pyrimidines present in fava beans. In G6PD-normal red cells
steady-state level of G6PD is so low that it becomes limiting
NADPH production is stepped up in response to ROS; in
for red cell survival, even in the absence of any oxidant
G6PD-deficient red cells ROS result in GSH depletion and
challenge; the result is CNSHA. Numerous point mutations
subsequent red cell damage (oxidative hemolyis). G6PD-null
in the G6PD gene causing CNSHA have been identified
mouse embryos have been obtained through targeted inac-
(Figure 11.6); interestingly, some individual mutations
tivation of G6PD in embryonic stem cells. From a detailed
appear to have arisen independently more than once, since
analysis of hemizygous mutant embryos, who die by day
they have been observed in cases from distinct geographical
10.5, it was inferred that the cause of death is precisely the
areas. With the majority of mutations, G6PD deficiency is
onset of aerobic metabolism, confirming the non-redundant
caused, at the biochemical level, by decreased stability of the
role of G6PD in protection against oxidative stress. Interest-
enzyme; that is, a marked acceleration of the physiological
ingly, heterozygous embryos also die, somewhat later, only if
decay of G6PD and of many other cytoplasmic proteins as
their G6PD-null gene is of maternal origin; in this case the
erythrocytes age. With a few mutations, G6PD deficiency
cause of death is a defective placenta, as a consequence of
is caused instead by decreased catalytic activity or altered
the selective inactivation of the paternal X chromosome in
substrate affinity. Although we cannot explain the reason
extra-embryonic tissues.

1 kb

1 2 3 4 5 6 7 8 9 10 11 12 13
(a)

100bp
m U
A S A h k
a t i S v C
M z
2 3 4 5 6 7 8 9 10 11 12 13
X

(b)
U Union; C Canton; M Mediterranean; A A-(202A); k Kaiping; t Taipei; v Viangchan;

m Mahidol; h Chatham; i Coimbra; S Seattle; s Santamaria; a Aures; z Cosenza; A A-(968C).


Fig. 11.6 Distribution of mutations along the human G6PD gene (Xq28). (a) Genomic structure of the human G6PD gene. Exons 1–13
are shown as numbered rectangles (black rectangles represent coding sequences; shaded rectangles represent non-coding sequences). (b) The
locations of amino acid substitutions are shown along the coding sequence of the gene, in which the exons are shown as open boxes.
Substitutions giving rise to the more severe sporadic (class I) variants are shown as filled circles below. Small deletions, a nonsense mutation, and a
splice site mutation are shown as filled rectangles, a cross and ∫, respectively. The milder class II and class III variants are shown as open circles
above; proven polymorphic variants are shown as a letter in a colored circle. Class IV variants are shown as open ellipses. All variants shown result
in G6PD deficiency, except G6PDA.
Source: Luzzatto L., Mehta A., Vulliamy T. (2001). Glucose-6-phosphate dehydrogenase deficiency. In Scriver C.R., Beaudet A.L., Sly W.S., Valle D.
(eds.), The Metabolic and Molecular Basis of Inherited Disease, 8th edn. New York: McGraw-Hill, pp. 4517–4553, with permission.
150 Molecular Hematology

for a severe clinical phenotype in every case, a cluster of Recent advances in molecular biology offer the possibility
mutations causing CNSHA in exons 10 and 11 corresponds of novel therapies for this common disease. First, lifelong
closely to the region of the molecule where the two subunits expression of human G6PD at therapeutic levels has been
interface. Dimerization is essential for enzyme activity, and obtained in RBCs and in white blood cells of mice through
it is not surprising that amino acid replacements in this retroviral-mediated transfer into HSCs. Second, in human
region will interfere with dimer formation or cause marked G6PD-deficient cells (class I–III variants) restoration of
instability of the dimer (Figure 11.6). G6PD activity has been achieved in vitro using HDACi. This
effect is mediated by histone hyperacetylation at the G6PD
Enzyme testing versus DNA testing promoter, resulting in increased transcription, protein level,
and enzymatic activity of the mutant protein. Interestingly,
Given that nowadays many laboratories are more geared to
HDACi do not exert this effect on all genes within the
polymerase chain reaction (PCR)-based technologies than to
glycolytic pathway; rather, transcription of G6PD is selec-
enzyme assays, it is tempting to diagnose G6PD deficiency
tively upregulated. Further understanding of the mechanism
by simply testing for underlying mutations. This is perfectly
of action of HDACi and other epigenetic modifiers could
feasible, especially in areas where prevalent mutations are
therefore open up therapeutic opportunities in other dis-
known. However, when no mutation is found this approach is
eases in which missense mutations result in an enzymatic
not definitive, because a known mutation unexpected in that
deficiency.
area, or a novel mutation, would be missed. DNA testing is
certainly the best way to detect all heterozygotes; however, it
will tell us nothing about skewing of X-chromosome inacti- Pyrimidine 5′ -nucleotidase deficiency
vation, and therefore it will not predict the risk and severity
Nucleotide metabolism in RBCs is necessary for the main-
of AHA in a heterozygote. For these reasons, measurement
tenance of ATP and guanosine triphosphate (GTP) levels.
of red cell G6PD activity is still necessary to inform clin-
Deficiency of pyrimidine 5′ -nucleotidase, involved in pyrim-
ical decision-making. The need that has emerged in many
idine metabolism, is of interest for several reasons. First, it
parts of the world to test for G6PD deficiency before admin-
is probably the third most common red cell enzymopathy
istering primaquine for definitive treatment of Plasmodium
(trailing G6PD and PK deficiencies). Second, the diagnosis
vivax malaria, has prompted very recently the development
can be suspected from red cell morphology, because it is
of “point of care” G6PD assays requiring practically no labo-
associated with basophilic stippling (accounted for by the
ratory facilities.
accumulation of RNA in mature red cells). Third, although
we do not really understand the precise mechanism, 5′ -
Management
nucleotidase deficiency is a good example of how the red
The commonest manifestations of G6PD deficiency, neonatal cell has virtually only one way to manifest it is suffering –
jaundice and AHA, are largely preventable or controllable by almost any metabolic abnormality will lead to accelerated
screening, surveillance, and avoidance of triggering factors, destruction (i.e. hemolysis). Fourth, the anemia of chronic
particularly fava beans, by G6PD-deficient subjects. One of lead poisoning (which had been known for a long time to
the current challenges is to develop a good animal model be associated with basophilic stippling) turns out to be the
that will allow pre-clinical testing of new drugs to assess consequence of the fact that lead is a powerful inhibitor of
whether they are likely to cause AHA in G6PD-deficient 5′ -nucleotidase; thus in terms of its hematological effects,
individuals. When a patient presents with AHA, and once lead poisoning is a phenocopy of 5′ -nucleotidase deficiency.
the cause is diagnosed, no specific treatment may be needed Since mapping of the pyrimidine-5′ -nucleotidase gene
if the episode is mild. At the other end of the spectrum, and (NT5C3) to chromosome 7 in 2001, 24 different mutations,
especially in children, AHA may be a medical emergency most commonly in homozygosity, have been identified in just
requiring immediate blood transfusion. The management over 30 unrelated families with 5′ -nucleotidase deficiency.
of neonatal jaundice does not differ from that of neonatal Mutations include missense, nonsense, alterations of splicing
jaundice due to causes other than G6PD deficiency and, sites, insertions, and deletions, and diagnosis is by molecular
in order to prevent neurological damage, treatment with testing. 5′ -nucleotidase deficiency has also been associated
phototherapy and/or exchange blood transfusion may be with hemoglobin E, hemoglobin D Punjab, and spectrin defi-
required. The management of CNSHA is similar to that of ciency. In the first case, concurrent 5′ -nucleotidase deficiency
CNSHA due to glycolytic enzymopathies, but in addition made the clinical expression of HbE disease more severe.
it is important to avoid exposure to potentially hemolytic Investigation of a father and daughter pair affected by
drugs. Again, although there is no evidence of selective red 5′ -nucleotidase deficiency, in which the father had a more
cell destruction in the spleen (as seen in hereditary sphero- severe disease phenotype, revealed that the father was
cytosis), splenectomy has proven beneficial in severe cases. homozygous for a polymorphism in the TATAA element of
The molecular basis of bone marrow failure syndromes and red cell enzymopathies 151

the promoter region in the uridine diphosphoglucurono- be greatly facilitated by the advent of a plethora of epigenetic
syl transferase 1A (UGT1A) gene (TA7/TA7), while the drugs, beyond HDAC inhibitors.
daughter was heterozygous (TA6/TA7). This polymor- An emerging and highly promising field of therapeutics
phism, usually associated with benign Gilbert syndrome, for glycolytic enzymopathies is the development of small
contributed to more severe hemolysis in the father. Such a molecules acting as allosteric activators.
case illustrates the value of molecular characterization of Therefore, it is realistic to expect that when this chapter is
multiplex families affected by a disease primarily regarded as due to be updated again, more patients with BMF syndromes
monogenic, with the aim of identifying disease-modifying and hemolytic anemias will have benefited from novel and
epistatic factors that contribute to clinical heterogeneity and effective treatments based on a deeper understanding of the
that could be the target of therapeutic intervention. molecular pathogenesis of the respective diseases.

Concluding remarks Further reading

Since the last edition of this volume, advances in different Aplastic anemia
areas of biology have already had impacts on the diagnosis
Desmond, R., Townsley, D.M., Dumitriu, B. et al. (2014). Eltrom-
and treatment of BMF and hemolytic anemias. In IAA the bopag restores trilineage haematopoiesis in refractory severe aplastic
impressive therapeutic success of the TpoR agonist eltrom- anaemia that can be sustained on discontinuation of drug. Blood 123
bopag promises to offer a simple and effective therapeutic (12): 1818–1825.
option for patients who are not candidates for allogeneic stem Luzzatto L. and Risitano A.M. (2018). Advances in understanding the
cell transplantation. pathogenesis of acquired aplastic anaemia. Br J Haematol. 182(6):
In inherited BMF syndromes, the ease of genetic diag- 758–776.
nosis by next-generation sequencing of multigene panels Marsh, J.C. and Mufti, G.J. (2016). Clinical significance of acquired
already offers accurate and swifter diagnosis for patients somatic mutations in aplastic anaemia. Int. J. Hematol. 104 (2): 159–
and their families, allowing for appropriate management 167.
Nishimura, J., Yamamoto, M., Hayashi, S. et al. (2014). Genetic variants
decisions, such as selection of disease-free familial donors
in C5 and poor response to eculizumab. N. Engl. J. Med. 370 (7): 632–
for hematopoietic stem cell transplantation and in many
639.
instances pre-natal diagnosis and counseling. Young, N.S. (2002). Acquired aplastic anaemia. Ann. Intern. Med. 136:
In PNH, the success of eculizumab has spurred the devel- 534–546.
opment of a new generation of complement inhibitors. Young, N.S. and Barrett, A.J. (1995). The treatment of severe acquired
Because these molecules target the C3 component of com- aplastic anaemia. Blood 85: 3367–3377.
plement, they are expected to be more effective in con- Young, N.S. and Maciejewski, J. (1997). The pathophysiology of
trolling extravascular hemolysis (regularly seen in patients acquired aplastic anaemia. N. Engl. J. Med. 336: 1365–1372.
on eculizumab) as well as intravascular hemolysis; and Young, N.S., Scheinberg, P., and Calado, R.T. (2008). Aplastic anaemia.
some could be administered orally. In the meantime, an Curr. Opin. Hematol. 15: 162–168.
eculizumab analog, ravulizumab, with a much longer half-
life, has become available. Paroxysmal nocturnal hemoglobinuria
Gene editing technologies, in particular CRISPR/Cas9- Almeida, A.M., Murakami, Y., Layton, D.M. et al. (2006). Hypomorphic
based gene correction, offer a more realistic hope than ever promoter mutation in PIGM causes inherited glycosylphosphatidyli-
before for gene therapy, in principle for any monogenic dis- nositol deficiency. Nat. Med. 12: 846–851.
order, such as those covered in this chapter. Araten, D.J., Nafa, K., Pakdeesuwan, K., and Luzzatto, L. (1999).
Increasing knowledge of epigenetic mechanisms of how Clonal populations of haematopoietic cells with paroxysmal noctur-
gene transcription is regulated and relevant mechanistic nal hemoglobinuria genotype and phenotype are present in normal
studies focused on genes associated with heritable BMF and individuals. Proc. Natl. Acad. Sci. U.S.A. 96: 5209–5214.
hemolytic anemias also promise to lead to new therapies. Belet, S., Fieremans, N., Yuan, X. et al. (2014). Early frameshift mutation
The case of restoring G6PD activity in G6PD-deficient cells in PIGA identified in a large XLID family without neonatal lethality.
Hum. Mutat. 35 (3): 350–355.
by simply enhancing transcription of the mutant allele using
Gargiulo, L., Papaioannou, M., Sica, M. et al. (2013).
HDAC inhibitors provides the proof of concept for a novel Glycosylphosphatidylinositol-specific, CD1d-restricted T cells
and simple notion: transcriptional upregulation of mutant in paroxysmal nocturnal hemoglobinuria. Blood 121 (14): 2753–
gene alleles has significant therapeutic potential, both in the 2761.
case of missense mutation-associated enzymopathies with Hillmen, P., Young, N.S., Schubert, J. et al. (2006). The complement
respect to the wild-type allele, and in the case of diseases inhibitor eculizumab in paroxysmal nocturnal hemoglobinuria. N.
due to haploinsufficiency. Exploration of this approach will Engl. J. Med. 355: 1233–1243.
152 Molecular Hematology

Krawitz, P.M., Höchsmann, B., Murakami, Y. et al. (2013). A case of Wang, X., Andreassen, P.R., and D’Andrea, A.D. (2004). Functional
paroxysmal nocturnal hemoglobinuria caused by a germline muta- interaction of monoubiquitinated FANCD2 and BRCA2/FANCD1 in
tion and a somatic mutation in PIGT. Blood 122 (7): 1312–1315. chromatin. Mol. Cell. Biol. 24: 5850–5862.
Luzzatto, L., Bessler, M., and Rotoli, B. (1997). Somatic mutations in Wang, A.T. and Smogorzewska, A. (2015). SnapShot: Fanconi anaemia
paroxysmal nocturnal hemoglobinuria: a blessing in disguise? Cell 88: and associated proteins. Cell 160 (1–2): 354–354.e1. 186.
1–4. Xia, B., Dorsman, J.C., Ameziane, N. et al. (2007). Fanconi anaemia is
Takeda, J., Miyata, T., Kawagoe, K. et al. (1993). Deficiency of the GPI associated with a defect in the BRCA2 partner PALB2. Nat. Genet. 39:
anchor caused by a somatic mutation of the PIG-A gene in paroxys- 159–161.
mal nocturnal hemoglobinuria. Cell 73: 703–711. Yamamoto, K., Ishiai, M., Matsushita, N. et al. (2003). Fanconi anaemia
FANCG protein in mitigating radiation- and enzyme-induced DNA
double-strand breaks by homologous recombination in vertebrate
Fanconi anemia cells. Mol. Cell. Biol. 23: 5421–5430.

Alter, B. and Young, N.S. (1998). The bone marrow failure syndromes.
In: Nathan and Oski’s Hematology of Infancy and Childhood, 5e (eds. Dyskeratosis congenita
D.G. Nathan and S.H. Orkin), 237–335. Philadelphia: WB Saunders.
Ameziane, N., May, P., Haitjema, A. et al. (2015). A novel Fanconi Autexier, C. and Lue, N.F. (2006). The structure and function of
anaemia subtype associated with a dominant-negative mutation in telomerase reverse transcriptase. Annu. Rev. Biochem. 75: 493–
RAD51. Nat. Commun. 6: 8829. 517.
Andreassen, P.R., D’Andrea, A.D., and Taniguchi, T. (2004). ATR cou- de la Fuente, J. and Dokal, I. (2007). Dyskeratosis congenita: advances
ples FANCD2 monoubiquitination to the DNA-damage response. in the understanding of the telomerase defect and the role of stem cell
Genes Dev. 18: 1958–1963. transplantation. Pediatr. Transplant. 11 (6): 584–594.
Cohn, M.A. and D’Andrea, A.D. (2008). Chromatin recruitment of DNA Heiss, N.S., Knight, S.W., Vulliamy, J.T. et al. (1998). X-linked dysker-
repair proteins: lessons from the Fanconi anaemia and double-strand atosis congenita is caused by mutations in a highly conserved gene
break repair pathways. Mol. Cell 32: 306–312. with putative nucleolar functions. Nat. Genet. 19: 32–38.
Garcia-Higuera, I., Taniguchi, T., Ganesan, S. et al. (2001). Interaction Jones, M., Bisht, K., Savage, S.A. et al. (2009). The shelterin complex and
of the Fanconi anaemia proteins and BRCA1 in a common pathway. haematopoiesis. J. Clin. Invest. 126 (5): 1621–1629.
Mol. Cell 7: 249–262. Jongmans, M.C., Verwiel, E.T., Heijdra, Y. et al. (2012). Revertant
Howlett, N.G., Taniguchi, T., Olson, S. et al. (2002). Bi-allelic inactiva- somatic mosaicism by mitotic recombination in dyskeratosis con-
tion of BRCA2 in Fanconi anaemia. Science 297: 606–609. genita. Am. J. Hum. Genet. 90: 426–433.
Joenje, H. and Patel, K.J. (2001). The emerging genetic and molecular Kirwan, M. and Dokal, I. (2008). Dyskeratosis congenita: a genetic dis-
basis of Fanconi anaemia. Nat. Rev. Genet. 2: 446–459. order of many faces. Clin. Genet. 73: 103–112.
Levitus, M., Waisfisz, Q., Godthelp, B.C. et al. (2005). The DNA heli- Mitchell, J.R., Wood, E., and Collins, K. (1999). A telomerase compo-
case BRIP1 is defective in Fanconi anaemia complementation group nent is defective in the human disease dyskeratosis congenita. Nature
J. Nat. Genet. 37: 934–935. 402: 551–555.
Litman, R., Peng, M., Jin, Z. et al. (2005). BACH1 is critical for homol- Vulliamy, T., Marrone, A., Goldman, F. et al. (2001). The RNA compo-
ogous recombination and appears to be the Fanconi anaemia gene nent of telomerase is mutated in autosomal dominant dyskeratosis
product FANCJ. Cancer Cell 8: 255–265. congenita. Nature 413: 432–435.
Meetei, A.R., Medhurst, A.L., Ling, C. et al. (2005). A human ortholog Woo, D.H., Chen, Q., Yang, T.L. et al. (2016). Enhancing a Wnt-telomere
of archaeal DNA repair protein HEF is defective in Fanconi anaemia feedback loop restores intestinal stem cell function in a human organ-
complementation group M. Nat. Genet. 37: 958–963. otypic model of dyskeratosis congenita. Cell Stem Cell 19 (3): 397–
Mosedale, G., Niedzwiedz, W., Alpi, A. et al. (2005). The vertebrate Hef 405.
ortholog is a component of the Fanconi anaemia tumour-suppressor
pathway. Nat. Struct. Mol. Biol. 12: 763–771. Diamond–Blackfan anaemia
Niedzwiedz, W., Mosedale, G., Johnson, M. et al. (2004). The Fan-
coni anaemia gene FANCC promotes homologous recombination Clinton, C. and Gazda, H.T. (2009). Diamond-Blackfan anaemia. In:
and error-prone DNA repair. Mol. Cell 15: 607–620. GeneReviews (eds. R.A. Pagon, M.P. Adam, H.H. Ardinger, et al.),
Pichierri, P. and Rosselli, F. (2004). The DNA crosslink-induced S-phase 1993–2016. Seattle, WA: University of Washington, Seattle.
checkpoint depends on ATR–CHK1 and ATR–NBS1–FANCD2 De Keersmaecker, K., Atak, Z.K., Li, N. et al. (2013). Exome sequencing
pathways. EMBO J. 23: 1178–1187. identifies mutation in CNOT3 and ribosomal genes RPL5 and RPL10
Strathdee, C.A., Gavish, H., Shannon, W.R., and Buchwald, M. (1992). in T-cell acute lymphoblastic leukaemia. Nat. Genet. 45 (2): 186–190.
Cloning of cDNAs for Fanconi’s anaemia by functional complemen- Draptchinskaia, N., Gustavsson, P., Andersson, B. et al. (1999). The gene
tation. Nature 356: 763–767. encoding ribosomal protein S19 is mutated in Diamond–Blackfan
Venkitaraman, A.R. (2004). Tracing the network connecting BRCA and anaemia. Nat. Genet. 21: 169–175.
Fanconi anaemia proteins. Nat. Rev. Cancer 4: 266–276. Gerrard, G., Valgañón, M., Foong, H.E. et al. (2013). Target enrichment
Wang, W. (2007). Emergence of a DNA-damage response network con- and high-throughput sequencing of 80 ribosomal protein genes to
sisting of Fanconi anaemia and BRCA proteins. Nat. Rev. Genet. 8: identify mutations associated with Diamond-Blackfan anaemia. Br. J.
735–748. Haematol. 162 (4): 530–536.
The molecular basis of bone marrow failure syndromes and red cell enzymopathies 153

Iskander, D., Psaila, B., Gerrard, G. et al. (2015). Elucidation of the and Molecular Basis of Inherited Disease, 8e (eds. C.R. Scriver, A.L.
EP defect in Diamond-Blackfan anaemia by characterisation and Beaudet, W.S. Sly and D. Valle), 4637–4664. New York: McGraw-Hill.
prospective isolation of human EPs. Blood 125 (16): 2553–2557. Luzzatto, L. (2015). G6PD deficiency: a polymorphism balanced by
Sankaran, V.G., Ghazvinian, R., Do, R. et al. (2012). Exome sequenc- heterozygote advantage against malaria. Lancet Haematol. 2: e400–
ing identifies GATA1 mutations resulting in Diamond-Blackfan e401.
anaemia. J. Clin. Invest. 122: 2439–2443. Luzzatto, L., Mehta, A., and Vulliamy, T. (2001). Glucose-6-phosphate
Vlachos, A., Ball, S., Dahl, N. et al. (2008). Diagnosing and treating Dia- dehydrogenase deficiency. In: The Metabolic and Molecular Basis of
mond Blackfan anaemia: results of an international clinical consensus Inherited Disease, 8e (eds. C.R. Scriver, A.L. Beaudet, W.S. Sly and D.
conference. Br. J. Haematol. 142: 859–876. Valle), 4517–4553. New York: McGraw-Hill.
Luzzatto, L., Nannelli, C., and Notaro, R. (2016). Glucose-6-phosphate
dehydrogenase deficiency. Hematol. Oncol. Clin. North Am. 30 (2):
Red cell enzyme deficiencies
373–393.
Beutler, E. (1994). Glucose-6-phosphate dehydrogenase deficiency. N. Makarona, K., Caputo, V.S., Costa, J.R. et al. (2014). Transcriptional and
Engl. J. Med. 324: 169–174. epigenetic basis for restoration of G6PD enzymatic activity in human
Chiarelli, L.R., Morera, S.M., Galizzi, A. et al. (2008). Molecular basis G6PD-deficient cells. Blood 124 (1): 134–141.
of pyrimidine 5′ -nucleotidase deficiency caused by 3 newly identi- Mentzer, W.C. (1998). Pyruvate kinase deficiency and disorders of gly-
fied missense mutations (c.187T>C, c.469G>C and c.740T>C) and colysis. In: Nathan and Oski’s Hematology of Infancy and Childhood,
a tabulation of known mutations. Blood Cells Mol. Dis. 40 (3): 295– 5e (eds. D.G. Nathan and S.H. Orkin), 665–703. Philadelphia: WB
301. Saunders.
Grace, R.F., Zanella, A., Neufeld, E.J. et al. (2015). Erythrocyte pyruvate Pamba, A., Richardson, N.D., Carter, N. et al. (2012). Clinical spec-
kinase deficiency: 2015 status report. Am. J. Hematol. 90 (9): 825–830. trum and severity of hemolytic anaemia in glucose 6-phosphate
Hirono, A., Kanno, H., Miwa, S., and Beutler, E. (2001). Pyruvate defi- dehydrogenase-deficient children receiving dapsone. Blood 120 (20):
ciency and other enzymopathies of the erythrocyte. In: The Metabolic 4123–4133.
Chapter 12 Anemia of chronic disease
Tomas Ganz
Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles CA, USA

Introduction and definition, 155 Principles of treatment, 158


General principles, 155 Future treatments, 159
Diagnosis, 155 Further reading, 159
Pathogenesis, 156

compounded by direct suppression of the erythrocyte pre-


Introduction and definition cursor population by inflammatory substances, blood loss,
decreased erythrocyte survival, decreased production of ery-
Anemia of chronic disease (more specifically referred to as
thropoietin, and true iron deficiency. Several of these factors
anemia of inflammation) is the second most common form
may contribute to the pathogenesis of the extreme variant of
of anemia (after iron deficiency) and is seen in the set-
the anemia that develops acutely in the critical care setting.
ting of chronic infections, inflammatory disorders includ-
ing rheumatological conditions and inflammatory bowel dis-
eases, certain malignancies, and chronic kidney diseases. A
related form of anemia develops within days in critically ill
Diagnosis
patients with sepsis or other acute conditions of systemic
inflammation. While the prevalence of iron deficiency in Iron parameters
the industrialized countries is now decreasing, anemia of
Hypoferremia, a decrease in serum iron concentration, is
inflammation would be expected to become more prevalent
the defining feature of anemia of inflammation. It develops
as the number of elderly with chronic inflammatory con-
within hours of the onset of infection or severe inflamma-
ditions continues to rise. The disorder usually presents as
tion. Synthesis of the iron-binding protein transferrin (mea-
a mild-to-moderate anemia, with erythrocyte morphology
sured directly or as total iron-binding capacity) is decreased,
ranging from normocytic and normochromic to microcytic
unlike iron-deficiency anemia where it is often increased.
and hypochromic, depending on the severity and duration of
The decrease in transferrin concentrations develops more
the underlying disease. Anemia of inflammation is defined
slowly than the decrease in serum iron levels because of the
by inadequate erythrocyte production in the setting of low
longer half-life of transferrin (8–12 days) compared with that
serum iron, despite preserved or even increased macrophage
of iron (about 90 minutes).
iron stores.

Serum ferritin
General principles Ferritin is found in serum as a large multimer consisting
mostly of glycosylated L-ferritin subunits. Ferritin is secreted
The essential pathogenic feature of this anemia is the iron by macrophages and hepatocytes, and its serum levels are
restriction of hemoglobin synthesis. Unlike iron-deficiency increased in response to iron loading of these cells. Systemic
anemia, the restriction is due to the slow release of iron from inflammation also increases ferritin secretion. Serum ferritin
stores rather than the absolute deficiency of total body iron. concentrations, reflecting inflammation and iron stores,
Depending on the underlying disease, the anemia is variably are increased in anemia of inflammation and decreased
in iron deficiency. Serum ferritin is thus very useful in
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. the differential diagnosis of patients with low serum iron
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. concentrations. However, depleted iron stores in patients

155
156 Molecular Hematology

with coexisting inflammation may yield intermediate ferritin restrict the availability of iron to extracellular microbes,
values, and this test becomes less useful. In this situation, limiting their proliferation during early phases of infec-
iron deficiency should be suspected if ferritin unexpectedly tion. Recent studies indicate that non-transferrin bound iron
decreases below the patient’s usual baseline. (NTBI) is a particularly potent simulator of pathogenesis by
It has been suggested that serum transferrin receptor gram-negative bacteria, and may be the most important tar-
assay may be helpful in differentiating iron-deficiency ane- get of the hypoferremic response.
mia from anemia of inflammation. Soluble transferrin recep- Hypoferremia of inflammation is mediated by inflamma-
tor is increased in iron deficiency, reflecting the post- tory cytokines, including prominently interleukin (IL)-6,
transcriptional stimulation of the synthesis of transferrin which increase rapidly in the first few hours of infection
receptor by iron-regulatory proteins 1 and 2 during cellular and induce the synthesis of the iron-regulatory hormone
iron deficiency, but it can also be increased whenever ery- hepcidin. Hepcidin binds to the iron efflux channel ferro-
throblast numbers increase as well as in inflammatory dis- portin on iron-exporting cells (macrophages, enterocytes,
orders, where it may reflect iron restriction affecting ery- and hepatocytes), causing the internalization and degra-
throcyte precursors. It is possible that the quantitative effects dation of ferroportin. Iron efflux is diminished and the
of inflammation and iron deficiency on serum transferrin continuing consumption of iron depletes the extracellular
receptor assay differ due to the differential effects of the iron pool (Figure 12.1). By recycling senescent erythrocytes,
two processes on the erythroblasts, but this remains to be macrophages in the liver and the spleen normally supply
explored experimentally and in larger human studies. most of the extracellular iron. During infection and inflam-
mation, macrophages retain iron, restricting its delivery to
plasma and to erythrocyte precursors (Figure 12.2).
Bone marrow studies
Bone marrow aspiration or biopsy is now rarely necessary
Iron restriction of erythropoiesis
for the diagnosis of anemia of inflammation. In general, the
bone marrow appears normal, unless the underlying disease Although hypoferremia develops within hours of the onset
directly affects it. The most important information obtained of infection or inflammation, the effect of decreased iron
from marrow examination is the content and distribution of and decreased hemoglobin synthesis on circulating ery-
iron, found as storage iron in the cytoplasm of macrophages throcytes is not immediately reflected in average erythro-
or as functional iron in nucleated red cells. In normal indi- cyte hemoglobin content or size. This is because the iron
viduals, several Prussian blue-staining particles are seen in restriction affects only erythrocyte precursors engaged in
many macrophages, and about one-third of nucleated red hemoglobin synthesis and not mature erythrocytes. New ery-
cells, called sideroblasts, contain blue inclusions. In iron defi- throcytes normally replace less than 1% of the circulating ery-
ciency, both sideroblasts and macrophage iron are absent. In throcytes per day, so the average erythrocyte indices (mean
contrast, in anemia of inflammation, sideroblasts are fewer corpuscular hemoglobin concentration, mean corpuscular
but macrophage iron is increased. The increase in storage volume) change slowly. However, some newer blood analyz-
iron in the face of decreased plasma iron and fewer siderob- ers, which can measure the hemoglobin content of reticu-
lasts is characteristic of anemia of inflammation. Although locytes, detect the effect of iron restriction as a decrease in
the bone marrow iron stain could be considered the gold reticulocyte hemoglobin before there is a significant change
standard for the differential diagnosis of anemia of inflam- in total hemoglobin.
mation and iron deficiency, the procedure causes patient dis- Iron restriction in anemia of inflammation is also detected
comfort, can be confounded by differences in interpreter as an increase in zinc protoporphyrin. Normally, when iron
skills and by the presence of poorly mobilizable iron deposits is sufficient during an intermediate step in the synthesis of
from intravenous iron preparations, and the serum ferritin heme, iron becomes incorporated into protoporphyrin IX. In
assay has largely replaced it. iron deficiency, zinc partially replaces iron and the amount
of zinc incorporated into protoporphyrin IX is increased.
In anemia of inflammation, zinc protoporphyrin is also
Pathogenesis increased, indicating that insufficient iron is reaching the
sites of heme synthesis in developing erythrocytes. Over the
course of several weeks, iron restriction results in anemia
Hypoferremia
that can eventually become microcytic and hypochromic and
Within hours of the onset of systemic infection or inflam- resemble iron-deficiency anemia. The development of ane-
mation, plasma iron concentrations markedly decrease in mia of inflammation is accelerated by other effects of inflam-
both humans and experimental animal models. This defen- mation or the underlying disease that may act to shorten ery-
sive response, hypoferremia of inflammation, is thought to throcyte lifespan or cause blood loss.
Anemia of chronic disease 157

RBC
Ferroportin
Ferroportin RBC Hepcidin
RBC
RBC

Increased
hepcidin
DMT1 DMT1
Ferroportin
RBC
RBC
Hepcidin

Ferroportin Macrophage Ferroportin Macrophage

Hepcidin
Hepcidin Hepcidin

Hepcidin

Fig. 12.1 Hepcidin–ferroportin interaction causes iron sequestration in macrophages. (Left) Macrophages in the spleen and liver ingest
senescent erythrocytes and extract their iron. When hepcidin is low, ferroportin molecules on macrophage cell membranes rapidly export the
recycled iron (gray tint). (Right) During inflammation, hepcidin synthesis is increased; hepcidin binds to macrophage ferroportin and induces its
internalization and degradation. Export of iron from macrophages slows, and iron remains in macrophage cytoplasm bound to ferritin. DMT,
divalent metal transporter; RBC, red blood cells.

Inflammation

Spleen

↓Fpn
↑hepcidin
↑hepcidin
e
↓F
↑hepc

↓Fpn
Liver ↓Fe
idin

Plasma
↓Fe-Tf
↓Fpn ↓Fe-
Tf
e
↓F

↓Erythropoiesis

Duodenum
Fig. 12.2 Iron restriction in anemia of inflammation. Under the influence of increased hepcidin concentrations, the release of stored and
recycled iron (Fe) from splenic and hepatic macrophages and hepatocytes is decreased, as is the absorption of dietary iron in the duodenum.
Continued iron utilization depletes plasma iron, causing hypoferremia and limiting iron delivery to erythropoietic tissue. Fpn, ferroportin; Tf,
transferrin.
158 Molecular Hematology

Effects of inflammation on iron absorption were not as high as in iron-deficiency anemia, although other
and release from stores studies have not observed a significant difference in erythro-
poietin concentrations between iron deficiency and anemia
Inflammation-induced hepcidin also decreases the absorp-
of inflammation. Erythropoietin suppression by cytokines
tion of dietary iron by acting on ferroportin displayed on
or lipopolysaccharide has been seen in some animal mod-
the basolateral membranes of enterocytes. Decreased iron
els of sepsis as well as in erythropoietin-producing cell lines.
absorption usually does not contribute much to the develop-
However, such suppression of erythropoietin production is
ment of anemia, because iron stores are generally sufficient
not a major pathogenic mechanism of anemia of inflamma-
to supply erythropoiesis for many months. However, the
tion. If it were, the administration of relatively small amounts
inflammatory blockade of iron absorption may contribute to
of erythropoietin should be sufficient to reverse the anemia
anemia in situations where iron demand is high and inflam-
of inflammation. Clinical experience and studies in animal
mation is long-lasting, as is the case in growing children with
models suggest the opposite: severe inflammation decreases
inflammatory bowel diseases or juvenile rheumatological
the effectiveness of erythropoietin, a common clinical situa-
disorders. Hepcidin would also be expected to inhibit the
tion described as erythropoietin resistance. Both anemia and
release of iron from hepatocyte stores, but the contribution
erythropoietin resistance have been seen in mice moderately
of this effect to anemia of inflammation has not yet been
overexpressing hepcidin, supporting the importance of the
established.
hepcidin–ferroportin axis and inflammatory iron restriction
in the pathogenesis of anemia of inflammation, as well as in
Cytokines involved in anemia of erythropoietin resistance.
inflammation
The contribution of specific cytokines to hypoferremia and Effects of inflammation on erythrocyte
anemia of inflammation has been difficult to delineate, lifespan
mainly because cytokines interact in complex networks reg- In early studies, a small decrease in erythrocyte lifespan was
ulating each other’s synthesis. In humans, depending on the noted in some animal models of inflammation, and it was
disease, therapies targeting IL-1, IL-6, and tumor necro- suggested that inflammation activates reticuloendothelial
sis factor (TNF)-α have all been shown to improve anemia macrophages to increase the removal of damaged or senes-
of inflammation, along with other inflammatory manifesta- cent erythrocytes. Other potential disease-specific mecha-
tions. The association of IL-6 with anemia of inflammation nisms that could contribute to the decreased lifespan include
appears to be the strongest. In both humans and mouse mod- the deposition of opsonic antibodies on erythrocytes, and
els, IL-6 induces hepcidin synthesis within hours by a tran- mechanical damage to erythrocytes from microvascular fib-
scriptional mechanism causing hypoferremia. rin strands or injured endothelia. In most cases, the effects on
Cytokines also have direct effects on erythropoiesis. Stud- erythrocyte lifespan are relatively small, and would be com-
ies in erythropoietic culture systems have demonstrated that pensated for by a minor increase in erythropoiesis.
TNF-α, IL-1, and interferon (IFN)-β and IFN-γ can inter-
fere with erythropoietin stimulation of erythroid progeni-
tors. This effect is seen in mice engineered to lack hepcidin, Principles of treatment
where treatment with potent inflammatory stimuli does not
cause hypoferremia but anemia still develops, albeit in a sub- Most often, the presenting symptoms of anemia of inflam-
stantially milder form than in the presence of hepcidin and mation, fatigue, and exercise intolerance are difficult to
hypoferremia. distinguish from those of the underlying disease, be it an
infection, inflammatory disease, or malignancy. Whenever
possible, the treatment should be aimed at the underlying
Inflammation and erythropoietin
disease. Effective treatment of the underlying disease will
resistance
also lead to the resolution of anemia. Unfortunately, this is
The normal response to anemia is an increase in erythro- not always feasible.
poietin production and subsequent compensatory increase in Infrequently, anemia of inflammation can be severe
erythropoiesis. It has been suggested that for the same sever- enough to exacerbate underlying cardiac or pulmonary
ity of anemia, erythropoietin production in anemia of inflam- disease and cause cardiac ischemia. In this acute setting,
mation is lower than it would be in other types of anemia. erythrocyte transfusion can rapidly improve oxygen delivery
Indeed, in some studies of patients with anemia associated and the manifestations of ischemia. More commonly, anemia
with rheumatoid arthritis or cancer, erythropoietin levels is moderate and its treatment is less urgent. Erythropoietin
Anemia of chronic disease 159

with or without high-dose parenteral iron can be useful Mechanisms of hypoferremia


for anemia symptoms that cannot be reversed by treating
Kawabata, H., Tomosugi, N., Kanda, J. et al. (2007). Anti-interleukin 6
the causative disease. High doses of parenteral iron may be
receptor antibody tocilizumab reduces the level of serum hepcidin
required to overwhelm the hepcidin-induced retention of in patients with multicentric Castleman’s disease. Haematologica 92:
iron in macrophages, and possibly to expand erythrocyte 857–858.
precursor populations that produce a hepcidin-suppressing Kemna, E., Pickkers, P., Nemeth, E. et al. (2005). Time-course analysis of
factor. In each patient, the potential side effects of erythro- hepcidin, serum iron, and plasma cytokine levels in humans injected
poietin and iron must be weighed against the anticipated with LPS. Blood 106: 1864–1866.
benefits, and the clinical effectiveness of the drugs. The con- Nemeth, E., Rivera, S., Gabayan, V. et al. (2004). IL-6 mediates hypofer-
tinued need for therapy should be evaluated regularly and remia of inflammation by inducing the synthesis of the iron regula-
critically. tory hormone hepcidin. J. Clin. Invest. 113: 1271–1276.
Rivera, S., Nemeth, E., Gabayan, V. et al. (2005). Synthetic hepcidin
causes rapid dose-dependent hypoferremia and is concentrated in
ferroportin-containing organs. Blood 106: 2196–2199.
Future treatments
Hepcidin and anemia
Improved understanding of the pathogenesis of anemia of
Gardenghi, S., Renaud, T.M., Meloni, A. et al. (2014). Distinct roles
inflammation should lead to the development of therapies for hepcidin and interleukin-6 in the recovery from anemia in mice
targeting the cytokine pathways that mediate increased injected with heat-killed Brucella abortus. Blood 123: 137–145.
hepcidin production, hepcidin itself, its interaction with Kim, A., Fung, E., Parikh, S.G. et al. (2014). A mouse model of anemia
ferroportin, or the ferroportin internalization pathway. If of inflammation: complex pathogenesis with partial dependence on
successful, such interventions should reverse the iron block hepcidin. Blood 123: 1129–1136.
and provide sufficient iron for normal erythropoiesis. Nicolas, G., Bennoun, M., Porteu, A. et al. (2002). Severe iron deficiency
anemia in transgenic mice expressing liver hepcidin. Proc. Natl. Acad.
Sci. U.S.A. 99: 4596–4601.
Roy, C.N., Mak, H.H., Akpan, I. et al. (2007). Hepcidin antimicrobial
Further reading peptide transgenic mice exhibit features of the anemia of inflamma-
tion. Blood 109: 4038–4044.
General
Clinical studies
Nemeth, E. and Ganz, T. (2014). Anemia of inflammation. Hematol.
Oncol. Clin. North Am. 28: 671–681. Cash, J.M. and Sears, D.A. (1989). The anemia of chronic disease: spec-
trum of associated diseases in a series of unselected hospitalized
Diagnosis patients. Am. J. Med. 87: 638–644.
Cazzola, M., Ponchio, L., de Benedetti, F. et al. (1996). Defective iron
Braga, F., Infusino, I., Dolci, A., and Panteghini, M. (2014). Soluble supply for erythropoiesis and adequate endogenous erythropoietin
transferrin receptor in complicated anemia. Clin. Chim. Acta 431: production in the anemia associated with systemic-onset juvenile
143–147. chronic arthritis. Blood 87: 4824–4830.
Hastka, J., Lasserre, J.J., Schwarzbeck, A. et al. (1993). Zinc protopor- Corwin, H.L. and Krantz, S.B. (2000). Anemia of the critically ill: “acute”
phyrin in anemia of chronic disorders. Blood 81: 1200–1204. anemia of chronic disease. Crit. Care Med. 28: 3098–3099.
Chapter 13 The molecular basis of
iron metabolism
Nancy C. Andrews1 & Tomas Ganz2
1
Duke University School of Medicine, Durham NC, USA
2
Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles CA, USA

Introduction, 161 Iron disorders, 166


Mechanisms of iron transport, 161 Conclusions, 170
Regulation of iron homeostasis, 164 Further reading, 171

Our understanding of the molecular processes of iron


Introduction metabolism has advanced considerably over the last two
decades as new techniques in genetics and molecular biology
Iron is one of several metals that are essential for nor-
have been applied to problems in this field. Much of what we
mal cellular processes. Its primary role in mammalian biol-
have learned has come from the study of patients with dis-
ogy is to bind oxygen in hemoglobin and myoglobin, and
eases of iron metabolism, and from investigation of animals
to catalyze the enzymatic transfer of electrons by iron-
with spontaneous and induced mutations in genes important
dependent enzymes, including cytochromes, peroxidases,
for the transport and storage of iron. Most features of iron
ribonucleotide reductases, and catalases. When iron is defi-
metabolism are very similar among mammalian species, val-
cient, the synthesis of iron-containing proteins is impaired,
idating the use of rodent models. More recently, important
with adverse consequences for oxygen delivery and cellular
insights into iron disorders in humans have also come from
metabolism. However, the same properties that make iron
the study of iron metabolism in zebrafish and even yeast.
useful for these functions can also lead to cellular damage
when iron is present in excess. Normally, several binding
proteins constrain the activity of iron, but when their iron-
binding capacity is exceeded, iron promotes the formation
Mechanisms of iron transport
of reactive oxygen species that attack cellular lipids, proteins,
and nucleic acids. Thus, iron balance must be carefully main- General principles
tained to avoid the deleterious effects of iron deficiency and
Iron is a large charged ion that cannot freely diffuse across
iron overload. All known disorders of iron metabolism can
cellular membranes. Transmembrane transfer requires
be considered abnormalities of iron balance.
specific carrier proteins. There are two general ways in
There is no physiological excretion mechanism for iron:
which cells transport iron. Some cells, such as intestinal
iron losses result only from bleeding and exfoliation of skin
epithelial cells, hepatocytes, macrophages, and placental
and mucosal cells. Under normal conditions, iron enters the
trophoblast, are equipped both to take in (import) iron and
body exclusively by dietary absorption, and absorption is
to release (export) it. These cell types are involved in the
closely regulated to balance the small losses. Iron balance is
acquisition, storage, and mobilization of iron for the entire
disrupted when intake and losses are not matched. Iron defi-
organism. Many other cells import iron but do not release it
ciency occurs when the dietary iron supply is inadequate,
unless the cells are destroyed. An intermediate group of cells,
when losses are increased (primarily because of bleeding), or
including erythrocyte precursors and cardiac myocytes,
when both of these circumstances are present. Iron overload
export iron primarily to maintain cellular homeostasis, but
results when iron absorption is inappropriately increased
the released iron contributes little to systemic iron needs.
due to genetic defects in iron-regulatory proteins, or when
Approximately 25 mg of iron are needed every day to sup-
repeated blood transfusions create a substantial iron burden.
port hemoglobin production in maturing erythrocytes. This
amount is much greater than the 1–2 mg entering the body
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. each day through the intestine. The iron for erythropoiesis is
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. largely provided by splenic and hepatic (reticuloendothelial)

161
162 Molecular Hematology

macrophages recycling iron from old erythrocytes and and some is exported through the basolateral membrane
making it available to developing erythroid precursors. through the action of a distinct transporter, ferroportin. A
Cells use at least three mechanisms to take up iron. Intesti- multicopper oxidase protein, hephaestin, facilitates basolat-
nal absorptive cells have cell surface transporters that carry eral transport, perhaps by oxidizing Fe2+ to Fe3+ to allow it
ferrous (Fe2+ ) ions directly across the membrane. Erythroid to exit from ferroportin and to bind to the plasma iron carrier
precursors use membrane receptors to take up iron bound to protein, transferrin.
a protein carrier, concentrate the iron in a subcellular com- The expression of ferroportin on the basolateral mem-
partment, and then transfer it across the membrane of that brane of enterocytes is the principal mechanism control-
compartment into the cytoplasm. Hepatocytes use both of ling the rate of iron flux through this transport system.
these mechanisms. Finally, recycling macrophages acquire The more ferroportin on the basolateral membranes, the
iron through the phagocytosis of aged or damaged erythro- more iron from enterocytes is passed into the plasma. Iron
cytes, lysing the cells and extracting the iron from their retained within the enterocytes is lost from the body when
hemoglobin. these cells finish their short lifespan and slough into the gut
lumen. The partitioning of iron (i.e. the process that gov-
erns how much enters the plasma and how much is retained
Intestinal iron transport within cells) is regulated by the hormone hepcidin through
post-translational degradation of ferroportin. The hepcidin–
Our current understanding of intestinal non-heme iron
ferroportin interaction plays an important role in determin-
transport is illustrated in Figure 13.1. Iron absorption takes
ing the overall efficiency of iron absorption.
place in an acidic environment in the proximal small intes-
In humans and other meat-eating organisms, direct
tine, just distal to the gastric outlet. Most non-heme dietary
absorption of heme contributes importantly to total iron
iron is in the ferric (Fe3+ ) form, whose very limited solubility
absorption. The pathways involved in heme absorption are
is enhanced by the low pH. It is reduced to Fe2+ by a brush
not well understood and may not be present in mice.
border ferrireductases, including the duodenal cytochrome b
(DCYTB). The Fe2+ ions pass through divalent metal trans-
porter 1 (DMT1, formerly called Nramp2, DCT1), a mem-
Other iron uptake mechanisms
brane protein that allows iron to traverse the apical bilayer.
DMT1 requires an acidic environment for its activity, because Upon entry into the plasma, iron attaches to transferrin,
it cotransports protons with iron atoms. The requisite pro- an abundant plasma protein that binds iron with extremely
tons are generated by apical Na+ /H+ exchangers, predomi- high affinity. Transferrin serves three important functions.
nantly NHE3. Once it crosses the membrane, some of the iron First, it keeps iron in solution. In an aqueous, neutral
is retained within the absorptive intestinal cells (enterocytes), pH environment, iron exists as Fe3+ ion, which is almost

? e– Fe3+
Fe Hephaestin Fe2Tf
Lumen
Heme
Blood
Ferroportin
Fe Fe Fe2+

HO
Fe3+ Fe2+ Fe3+
Ferrireductases
Ferritin
Fe2+
H+ DMT1

H+ NHE3
+
Na Enterocyte

Fig. 13.1 Intestinal non-heme iron absorption. The cartoon shows an absorptive enterocyte from the duodenal epithelium, joined to adjacent
cells by iron-impermeable tight junctions. The apical brush border is on the luminal side (left) and the basolateral surface is on the blood side
(right). Dietary Fe3+ iron is reduced by the ferrireductase DCYTB and other mechanisms to produce Fe2+ ion for transport. Fe2+ crosses the apical
membrane through the action of DMT1 to enter the cell. There, iron is partitioned between storage and export; stored iron is ultimately lost from
the body when the epithelial cells senesce and exfoliate into the gut lumen. Meanwhile, a fraction of the iron is exported across the basolateral
membrane by ferroportin. The iron hormone hepcidin binds to ferroportin and causes its degradation, thereby inhibiting iron efflux, proportionally
to hepcidin concentration. Hephaestin is a ferroxidase-like protein that aids in iron export, probably by oxidizing the Fe2+ iron leaving ferroportin to
the Fe3+ form, which binds to plasma transferrin (TF).
The molecular basis of iron metabolism 163

insoluble. Second, it renders iron non-reactive, and allows into the endosomes to lower their internal pH, leading to the
it to circulate in a safe, non-toxic form. Third, transferrin release of iron from transferrin. The liberated iron then leaves
facilitates the delivery of iron to cells bearing transferrin the endosome to enter the cytoplasm. This also requires a
receptors (TFRs) on their surfaces. transmembrane transport step, which is probably mediated
Most differentiated cell types express few, if any, trans- by DMT1 and facilitated by the low endosomal pH. The pro-
ferrin receptors, but there are three important exceptions: cess is assisted by one or more essential ferrireductases, in
tumor cells, activated lymphocytes, and erythroid precursors. erythroid cells principally Steap3 (six transmembrane epithe-
Tumor cells presumably use transferrin receptors to opti- lial antigen of the prostate 3), which convert ferric to ferrous
mize iron uptake to support rapid proliferation. The same iron. Within the cytoplasm, ferrous iron is shuttled to sites
may be true for activated lymphocytes. However, the great- of use and storage. Two iron chaperones, poly(rC)-binding
est demand for iron is by erythroid precursors, to support proteins (PCRB) 1 and 2, have been shown to escort iron to
the large-scale production of hemoglobin. In normal adults, ferritin and several other proteins that require or transport
about two-thirds of the total body iron endowment is found iron. Meanwhile, transferrin and transferrin receptor pro-
in hemoglobin, distributed among erythroid precursor cells teins return to the cell surface, where transferrin is released
and circulating erythrocytes. and the receptors become available for further cycles of iron
Iron-loaded transferrin binds with high affinity to trans- delivery.
ferrin receptors. Of the two forms of the receptor, transfer- Why should cells have evolved the complicated trans-
rin receptor 1 (TFR1) appears to be critical for cellular iron ferrin cycle when it is possible to take up iron directly?
uptake, but both TFR1 and transferrin receptor 2 (TFR2) are There are at least two likely answers. First, tight binding
involved in systemic iron homeostasis. Transferrin receptors, of iron to transferrin is advantageous while iron is in the
carrying their iron–transferrin cargo, undergo endocytosis circulation, but it complicates the transport of iron into
(Figure 13.2). Portions of the cell membrane bearing trans- cells. The pH-dependent release of iron, occurring in a con-
ferrin receptors invaginate into the cytoplasm and bud off trolled intracellular environment, solves the problem of lib-
as intracellular vesicles (endosomes). Protons are pumped erating the iron. Second, binding of iron-loaded transfer-
rin to transferrin receptors serves to concentrate iron in the
vicinity of DMT1, probably achieving much higher local
Transferrin receptor iron concentrations than would be possible without such a
mechanism. This allows more efficient iron uptake by cells
with large needs (erythroid precursors, tumor cells, activated
lymphocytes), without exposing other cells to unnecessary
iron.
Undoubtedly, other cell types use these and other schemes
for assimilating iron. Hepatocytes and macrophages are par-
DMTI ticularly important in iron homeostasis and their iron uptake
mechanisms, though not well understood, deserve men-
H+ tion. Hepatocytes express both types of transferrin recep-
tor and likely take up iron via the transferrin cycle. Hepato-
cytes also avidly take up non-transferrin-bound iron (NTBI)
H+ when the plasma iron concentration exceeds the binding
capacity of transferrin. This is an abnormal situation, except
perhaps in the portal circulation after an iron-rich meal,
Erythroid precursor
because there are usually about three times as many trans-
Fe2.transferrin ferrin iron-binding sites as are needed (i.e. transferrin is
–Apo-transferrin normally about 30% saturated with iron). However, patients
with iron overload may have more iron than transferrin can
Fig. 13.2 The transferrin cycle. The transferrin cycle of
accommodate, and this excess iron appears to be rapidly
receptor-mediated endocytosis is initiated by binding of diferric
removed from the circulation by hepatocytes. The molecu-
(Fe2 )-transferrin to a cell surface transferrin receptor. The
ligand–receptor complex is internalized by invagination of
lar mechanism for hepatic NTBI uptake uses the SLC39A14
clathrin-coated pits to form specialized endosomes. Influx of protons (also called ZIP14) transporter, and in the absence of this
into the endosome decreases its pH to approximately 5.5, facilitating transporter in mice, NTBI is no longer taken up by the
release of iron from transferrin. The iron is then transferred to the liver. Hepatocytes express ferroportin and release iron at
cytoplasm by DMT1. Apo-transferrin and transferrin receptor return to least in part by a hepcidin-controlled ferroportin-dependent
the cell surface for further cycles of iron uptake. mechanism.
164 Molecular Hematology

As discussed earlier, reticuloendothelial macrophages to form hemosiderin, a heterogeneous iron-containing sub-


obtain iron by phagocytosing and breaking down ery- stance that probably serves little purpose but to keep iron
throcytes. This process takes place in discrete phagocytic from causing harm. Both ferritin and hemosiderin accumu-
vesicles within the cells, and likely involves the action of late in iron-overloaded tissues.
heme oxygenase, an enzyme that catalyzes the degrada- The liver serves as the primary depot for iron in excess of
tion of heme. After crossing the phagosomal membranes, immediate needs. It has a very large capacity for storing iron,
cytoplasmic iron is stored in ferritin. Macrophages export though this capacity is ultimately exceeded in iron overload
ferrous iron through their abundant ferroportin, assisted disorders. While other tissues (myocardium, pancreas) also
by the plasma ferroxidase ceruloplasmin so that ferric iron fill up with iron in iron overload, the liver is frequently the
is delivered to plasma transferrin. Similar to intestinal cells first site where damage from iron overload becomes apparent.
and hepatocytes, reticuloendothelial macrophages partition Hepatocytes avidly take up NTBI from the plasma.
their iron content into retained and released portions. This Reticuloendothelial macrophages are also important for
process is regulated in response to the iron needs of the iron storage, but their iron comes from degraded erythro-
body by the hepatic hormone hepcidin, which induces the cytes. Patients treated with frequent transfusions typically
internalization and degradation of macrophage ferroportin. accumulate excess iron in macrophages first, and only later
While there is currently no direct way to measure how in other tissues. This pattern of iron accumulation has been
much iron is retained and how much is released, com- referred to as “siderosis” to distinguish it from hemochro-
monly used laboratory tests provide some information. matosis, which is primary iron loading of parenchymal cells.
The concentration of serum iron (and hence transferrin
saturation) is determined by two factors: macrophage iron
release and erythroid iron utilization. When erythropoiesis Regulation of iron homeostasis
occurs at a steady rate, transferrin saturation is determined
primarily by the rate of macrophage iron release, increasing Iron homeostasis requires the coordinated regulation of iron
when there is increased iron export and decreasing when transport and iron storage so that tissues will have adequate
iron is retained or when less iron is being recycled from amounts to meet their needs, but will not become overloaded
erythrocytes. In contrast, the concentration of serum ferritin with iron. Regulation must involve the control of cellular iron
roughly correlates with the amount of storage iron in the import, export, and partitioning. Over the last decade, a bet-
body. Serum ferritin appears to be derived primarily from ter understanding of regulation at each of these steps has
reticuloendothelial macrophages and hepatocytes. However, emerged.
serum ferritin is not a very accurate indicator, because levels There are at least four known regulators of intestinal iron
are increased by inflammation, tissue damage, and rare absorption (Figure 13.3): iron stores, erythropoietic demand,
congenital hyperferritinemia disorders. Nonetheless, a low hypoxia, and inflammation. The stores regulator modulates
serum ferritin value invariably indicates depleted iron stores. absorption several-fold, increasing absorption in iron defi-
ciency and decreasing absorption in iron overload. The ery-
throid regulator is more potent: it can increase iron absorp-
Sites of iron storage
tion many-fold when erythropoiesis becomes iron restricted.
Iron is stored within cells in the cavities of ferritin protein The hypoxia regulator is not well characterized, but its effects
multimers. There are two types of ferritin subunit, L and H, may be at least partly distinct from those of the erythroid reg-
both approximately 20 kDa in size. These subunits assemble ulator. This regulator increases iron absorption in response
in varying proportions into 24-subunit cage-like structures. to hypoxia. Finally, an inflammation regulator decreases iron
Up to several thousand iron atoms can be stored in each fer- absorption in response to inflammation from a variety of
ritin multimer. Like transferrin, ferritin’s function is to pre- causes.
vent iron from reacting with other cellular constituents, and Similar influences also regulate the release of recycled
allows controlled iron release in response to increased cellu- iron from reticuloendothelial macrophages. During inflam-
lar needs. The molecular details of iron incorporation into mation or iron sufficiency, iron is retained in macrophages,
and release from ferritin are not fully understood, but it now but is released in response to iron deficiency or erythroid
appears that iron is released by ferritinophagy, a regulated demand. The regulation of iron storage in hepatocytes is less
process in which nuclear receptor coactivator 4 (NCOA4) well understood, but it is clearly responsive to systemic iron
binds to ferritin under low iron conditions and delivers fer- requirements.
ritin to the autophagy pathway, culminating in the degrada- The iron-regulatory hormone hepcidin is likely to be
tion of ferritin in lysosomes and the release of iron into the the common effector of the stores, erythroid, hypoxia, and
cytoplasm. Under some circumstances, ferritin and other cel- inflammation regulators (Figure 13.3). Hepcidin (also called
lular components are partially degraded and conglomerated LEAP, HAMP) is produced by hepatocytes as a 25-amino acid
The molecular basis of iron metabolism 165

he
pc
idi Spleen
n
Fpn
Liver
Hypoxia Inflammation hepcidin

Fe
Fpn
Fe
Liver Erythropoietic signal Plasma
he Fe-Tf
pc Fe-T
idi f
n
Fpn
he

Bone marrow
pc

Plasma Fe
id

Fe-Tf and other sites


in

of iron usage

(a) Iron signal Duodenum (b)


Fig. 13.3 Regulators of iron absorption, storage, and tissue distribution. (a) Iron stores, erythropoietic demand, hypoxia, and inflammation
modulate the hepatic production of hepcidin, an iron-regulatory hormone. Increased iron stores and inflammation both increase hepcidin
expression (↑), whereas increased erythropoietic demand and hypoxia decrease hepcidin expression (⊥). (b) Hepcidin controls the major flows of
iron in the body, including the efflux of iron into plasma from absorptive enterocytes, from splenic and hepatic macrophages involved in recycling
of iron from erythrocytes, and from hepatocytes that store iron. Plasma iron is destined for hemoglobin synthesis by erythrocyte precursors in the
bone marrow, and for other sites of iron utilization. Fe, iron; Fpn, ferroportin; Tf, transferrin.

peptide with four disulfide bonds from an 84-amino acid pre- The production of hepcidin is increased in response to
cursor molecule, with the ultimate cleavage mediated by the dietary or parenteral iron loading, presumably as part of
prohormone convertase furin. Administration of synthetic a compensatory mechanism to decrease iron absorption
hepcidin to mice results in profound and prolonged hypofer- and decrease plasma iron (primarily derived from recycling
remia. The hypoferremic effect of hepcidin is due to its ability macrophages). Hepcidin thus acts as the stores regulator. The
to inhibit the major mechanisms that deliver iron to plasma: production of hepcidin is decreased by iron deficiency, allow-
intestinal iron absorption and the release of iron from recy- ing more iron to enter the body through the intestine and
cling compartments and stores in the spleen and the liver. more iron to enter the plasma from recycling macrophages.
Continuing consumption of iron by erythropoiesis and other In addition, hepcidin is suppressed by increased erythro-
iron-requiring processes then rapidly depletes the relatively poietic activity, sometimes even in the face of systemic iron
small plasma and extracellular iron compartment. overload, confirming that hepcidin is the ultimate erythroid
On the cellular and molecular levels, hepcidin inhibits regulator. How enhanced erythropoiesis suppresses hepcidin
the efflux of cellular iron into extracellular fluid or plasma, production is not yet fully understood. Recent studies indi-
exerting its effect on the major cell types involved in iron cate that erythropoietin-stimulated erythroblasts secrete ery-
transport: enterocytes, macrophages, hepatocytes, and pla- throferrone (also called Fam132b), a member of the TNFα
cental syncytiotrophoblast. Unlike other cell types, iron- superfamily, which acts on hepatocytes through an unknown
exporting cells express ferroportin, the sole known cellu- pathway to suppress hepcidin transcription. The production
lar iron exporter. Hepcidin acts by binding to ferroportin, of hepcidin is also decreased in hypoxia, suggesting that
inducing its internalization and lysosomal degradation (Fig- hepcidin is an ultimate effector of the hypoxia regulator. The
ure 13.4). The ferroportin degradation pathway is similar effect of hypoxia on hepcidin could be partly mediated by
to that of other receptors undergoing ligand-induced endo- hypoxia-induced erythropoietin acting on erythroblasts to
cytosis, and is triggered by ubiquitination of a string of increase erythroferrone secretion. Finally, hepcidin expres-
lysine residues in the cytoplasmic loop connecting the two sion is induced by inflammation, probably through a direct
halves of the molecule. The loss of ferroportin from cell action of the cytokine interleukin (IL)-6 and other inflamma-
membranes proportionately reduces the efflux of iron into tory mediators on hepatocytes, where hepcidin transcription
extracellular fluid. Alternatively, at higher concentrations, is then modulated by the JAK2-STAT3 regulatory pathway.
hepcidin may directly occlude the channel through which In this case, induced hepcidin expression leads to decreased
iron moves. intestinal iron absorption and decreased macrophage iron
166 Molecular Hematology

DMT1 DMT1

Ferritin
Hepcidin

Ferritin

Ferroportin

Ferroportin

Hepcidin

Fig. 13.4 Regulation of cellular iron transport by hepcidin. Hepcidin regulates the release of iron from enterocytes (shown in the figure) and
macrophages by binding to ferroportin and inducing its internalization and degradation in lysosomes. Under the influence of hepcidin, iron export
is decreased and iron accumulates in cytoplasmic ferritin. In enterocytes, regulatory effects of increased cellular iron may result in decreased iron
uptake.

release, acting as an inflammation regulator. There is grow- iron-regulatory pathway. Although multiple BMPs, including
ing evidence that, in response to the inflammatory regulator, BMP-2, BMP-4, BMP-6, and BMP-9, can stimulate hepcidin
increased hepcidin expression contributes to the abnormal synthesis in hepatocytes and hepatocyte-derived cell lines,
iron homeostasis observed in the anemia of chronic disease BMP-6 and BMP-2 appear to be uniquely specialized for sig-
(also known as the anemia of inflammation). Useful clinical naling in iron homeostasis. Remarkably, the BMPs impor-
assays for hepcidin levels in serum are available. tant for iron homeostasis are not produced by hepatocytes
Based on recent studies in cellular models, genetically but by adjacent sinusoidal endothelial cells, yet their secre-
altered mice, and patients with genetic iron disorders, hep- tion reflects closely the amounts of iron stored in hepatocytes.
cidin synthesis in hepatocytes is regulated by iron sensors How all these cells and molecules interact to regulate hep-
and signaling molecules assembled around the scaffold of the cidin is still an unfinished story.
bone morphogenetic protein receptor (BMPR) and its sig-
naling pathway. Two molecules that may function as iron
sensors are TFR1 and TFR2. The transmembrane molecule Iron disorders
HFE can bind to both transferrin receptors, but is displaced
from TFR1 when the receptor binds its main ligand, iron- In the most general terms, iron disorders represent a failure of
transferrin. The glycosylphosphatidylinositol (GPI)-linked homeostatic mechanisms that match intestinal iron absorp-
membrane protein hemojuvelin binds to BMPR and its bone tion and systemic iron distribution to the iron requirements
morphogenetic protein (BMP) ligands, and may act as a co- of erythropoiesis and other iron-consuming processes. Iron
receptor that converts the generic BMPR pathway into an homeostasis results in the maintenance of normal plasma
The molecular basis of iron metabolism 167

and extracellular iron concentrations (10–30 μmol l−1 in heart. Macrophages of the reticuloendothelial system are iron
humans) and normal iron stores in macrophages and hep- depleted or at least relatively spared. This disorder results
atocytes that help buffer transient disturbances in the supply from a small but chronic increase in intestinal iron absorp-
or demand for iron. Either genetic or environmental causes, tion, averaging about two- to threefold the normal level.
and often both, can contribute to iron disorders. Over time, the presence of iron causes damage by promot-
ing the formation of toxic oxygen radicals, which attack
cellular structures and thereby cause reactive fibrosis. The
Iron-deficiency disorders earliest manifestation of HFE-associated hemochromatosis
Iron deficiency is a major public health problem and the most is increased transferrin saturation, often approaching 100%
common nutritional cause of anemia. Very rarely, iron defi- before tissue iron deposition is noted. The treatment for
ciency is genetic and is manifested as hypoferremia, hypo- hemochromatosis is phlebotomy, and this has been used
ferritinemia, and iron-restricted microcytic anemia refrac- effectively for more than half a century. Initially, blood is
tory to oral iron administration. Recent studies have shown removed frequently to rapidly decrease storage iron. Later,
that homozygous or compound mutations in a gene encod- iron balance is maintained by periodic phlebotomy, titrated
ing TMPRSS6 (Transmembrane protease, serine 6, also called to meet the needs of the individual patient. This treat-
matriptase 2) are responsible for most cases of iron-refractory ment apparently produces no significant morbidity and has
iron-deficiency anemia. TMPRSS6 negatively regulates hep- been shown to normalize the life expectancy of affected
cidin, by cleaving membrane hemojuvelin. patients.
Most genetic disorders of iron metabolism result in iron Hemochromatosis has been recognized as an inborn error
overload rather than iron deficiency. They are attributable to of iron metabolism since the 1930s. In 1976 a French physi-
mutations that affect the regulation of intestinal iron absorp- cian, Marcel Simon, made the important observation that the
tion and body iron distribution. genetic predisposition to hemochromatosis was linked to the
human major histocompatibility complex on chromosome
6p, and was most frequently associated with an HLA-A3 hap-
Iron overload disorders lotype. This insight laid the groundwork for the discovery of
causative mutations in the HFE gene 20 years later. It is now
It is now clear that there are many genetic iron overload dis- known that most patients with classical hemochromatosis are
orders, affecting distinct components of the iron-regulatory homozygous for a unique mutation (cysteine 282 to tyrosine,
circuitry. They can be generally classified as hemochro- or C282Y) in HFE, perhaps originating in a northern Euro-
matosis disorders (iron deposition in parenchymal cells) or pean ancestor.
siderosis disorders (deposition of iron in reticuloendothelial HFE is an atypical HLA class I molecule, similar to its chro-
macrophages). The known disorders and their genetic causes mosomal neighbors. Although most members of this family
are listed in Table 13.1. Over the last few years, it has become are involved in immune regulation, HFE has no known func-
clear that the final common mechanism for most forms of tion in the immune system. It interacts with TFR1 on the cell
hereditary hemochromatosis is relative or absolute hepcidin surface, where its binding site overlaps with that of transfer-
deficiency or, rarely, resistance to hepcidin. The lack of hep- rin. Recent studies indicate that the binding of transferrin to
cidin or the loss of its effect on ferroportin results in exces- TFR1 displaces HFE, making it active as a stimulus for hep-
sive or unrestrained absorption of dietary iron, as well as the cidin synthesis.
depletion of the macrophage iron storage compartment. The The C282Y mutation is highly prevalent. In typical popula-
genetically defective proteins in hereditary iron overload dis- tions of northern European descent, the carrier frequency has
ease are implicated as regulators of hepcidin (HFE, TFR2, been estimated to be between about 1 in 8 and 1 in 10. This
hemojuvelin), targets of hepcidin (ferroportin), or molecules indicates that about 1 in 200 individuals are homozygous,
that convey iron to the iron-sensing mechanism in hepato- and at risk of iron loading. However, not all C282Y homozy-
cytes (transferrin). gotes will develop clinical hemochromatosis. There is a wide
range in iron loading and its complications. Some individ-
uals will have severe manifestations by the third decade
HFE-associated hemochromatosis
of life, whereas others may never have signs or symptoms
The most common form of hemochromatosis was classi- of hemochromatosis. This variability is probably explained
cally described as the triad of cirrhosis, diabetes, and skin by gender (iron loss through menstruation), other genetic
melanosis (“bronze diabetes”), but fortunately such advanced factors (modifying genes), and environmental factors (e.g.
disease is now rare. It is a late-onset disorder, inherited in alcohol intake, dietary iron consumption). Although several
an autosomal recessive pattern and characterized by iron modifying genes have been proposed, their identification and
deposition in parenchymal cells of the liver, pancreas, and characterization are still incomplete.
168 Molecular Hematology

Table 13.1 Iron overload disorders

Chromosomal
location of Gene (types of
Disorder defective genes mutations) Phenotype Hepcidin

Hemochromatosis disorders
HFE-associated 6p, near the HLA HFE (missense and Iron accumulation in the parenchymal Low or
hemochromatosis complex splicing mutations; cells of the liver, heart, pancreas; inappropriately
(also called type 1 C282Y is by far the elevated transferrin saturation; relative normal for iron
hemochromatosis) most important) paucity of iron in macrophages. overload
Clinical manifestations include liver
fibrosis, cirrhosis, markedly increased
incidence of hepatocellular carcinoma,
cardiomyopathy, and diabetes
Juvenile 1q Hemojuvelin hepcidin Similar to HFE-associated Very low to
hemochromatosis 19q (all known mutations hemochromatosis, but greatly absent
(also called type 2 prevent production accelerated, leading to severe cardiac
hemochromatosis) of any hepcidin and endocrine complications in the
protein) second decade of life
TFR2-associated 7q TFR2 (missense and Similar to HFE-associated Low
hemochromatosis nonsense mutations) hemochromatosis
(also called type 3
hemochromatosis)
Autosomal dominant 2q Ferroportin (rare Iron accumulation in parenchymal cells ?
hemochromatosis missense mutations
(also called type 4 causing resistance to
hemochromatosis) hepcidin). Most
other missense
ferroportin
mutations cause loss
of function and
siderosis
Siderosis disorders
Autosomal dominant 2q Ferroportin (only Macrophage-predominant iron loading; Normal or high
siderosis (also called missense mutations) in some patients parenchymal iron (few patients
type 4 loading can occur later. Some patients studied)
hemochromatosis) have anemia early in their course
(particularly women). Ferritin levels are
markedly elevated, but serum
transferrin saturation generally is not
African siderosis ? Unknown Similar to autosomal dominant siderosis, ?
but ferroportin mutations have not
been reported. Thought to be a
combination of genetic and
environmental factors
Disorders of iron balance
Atransferrinemia 3q Transferrin (missense Deficiency in serum transferrin leading to Low
mutations) tissue iron overload and severe
iron-deficiency anemia
Aceruloplasminemia 3q Ceruloplasmin Deposition of iron in the brain, liver, and ?
(missense and null pancreas. Late-onset
mutations) neurodegenerative disease, dementia.
and diabetes
The molecular basis of iron metabolism 169

In addition to C282Y, other mutations and polymorphisms chromosome 1q. Hemojuvelin is a GPI-linked membrane
have been identified in the HFE gene. The most common of protein that functions as an essential regulator of hepcidin.
these is a histidine-to-aspartic acid substitution at amino acid Disruption of both copies of hemojuvelin has been shown to
63 of the protein (H63D). This polymorphism is found in cause profound hepcidin deficiency. It thus appears that the
about one-fifth of the world’s population. Although it may greater severity of the juvenile form of the disease is due to
occasionally be associated with iron overload, particularly the lower levels of hepcidin compared with the adult form of
when it is found in individuals heterozygous for the C282Y hemochromatosis.
mutation, its clinical significance is probably minor, though
clinical laboratories test for it. Most other HFE mutations are
TFR2-associated hemochromatosis
quite rare and are not identified by routine screening tests.
In the past, the proportion of at-risk C282Y homozygous A third form of autosomal recessive hemochromatosis
individuals who develop clinical hemochromatosis was esti- (type 3) has been reported to be indistinguishable from HFE-
mated to be about 20–40%. However, Beutler and colleagues associated hemochromatosis in its clinical manifestations,
published the results of a large questionnaire-based study but patients have no mutations in the HFE gene. Instead, their
of patients seen by a health maintenance organization, in disease is caused by mutations in the TFR2 gene on chro-
which they concluded that less than 1% of C282Y homozy- mosome 7q. TFR2 is a protein that is highly homologous to
gotes would have severe disease. There is no consensus yet the transferrin receptor (also known as TFR1), and highly
on laboratory parameters that identify C282Y homozygotes expressed by hepatocytes and hematopoietic cells. TFR2 is
at risk for clinically significant organ damage, but recent data capable of binding transferrin and transporting it into the
indicate that few if any patients with serum ferritin levels cell, but it does so at much lower efficiency than does trans-
below 1000 ng ml−1 develop cirrhosis, the main complication ferrin receptor. TFR2 is stabilized by interaction with iron–
of hemochromatosis. transferrin and is also able to bind HFE. Patients and mice
with homozygous TFR2 mutations have low hepcidin expres-
sion, implicating this protein in the regulation of hepcidin
Juvenile hemochromatosis
synthesis, possibly as a sensor of transferrin saturation. Some
Juvenile hemochromatosis is similar to HFE-associated patients have been shown to have nonsense mutations, indi-
hemochromatosis, but very rare, and characterized by earlier cating that TFR2 is not an essential protein for survival. Type
onset of iron loading and its complications. The target organs 3 hemochromatosis is effectively treated by phlebotomy.
are the same as those affected in HFE hemochromatosis, Two siblings with combined HFE and TFR2 mutations
but cardiac and endocrine dysfunction are more severe, and were reported to have the severe juvenile form of the disease,
untreated patients typically die from cardiomyopathy by age suggesting that HFE and TFR2 have additive effects on hep-
30 years. Liver cirrhosis and failure are uncommon. There are cidin expression.
several possible explanations for this pattern. First, experi-
ence with patients who develop siderosis from chronic trans-
Ferroportin-associated hemochromatosis
fusion therapy suggests that rapid iron loading is especially
toxic for the heart and endocrine tissues. Secondly, patho- Most reported ferroportin mutations cause the loss of ferro-
logical iron deposition in the adolescent years may be partic- portin function and the clinical picture of macrophage iron
ularly bad for young hearts which are growing to meet the overload (siderosis) with few if any clinical consequences
demands of a larger body mass; this is analogous to the prob- (see later). A rare form of hereditary hemochromatosis is
lems noted with doxorubicin cardiotoxicity in this age group. an autosomal dominant disorder (type 4 hemochromato-
Furthermore, endocrine problems are probably more appar- sis) due to ferroportin mutations that confer resistance to
ent in adolescents because they fail to go through normal hepcidin-induced internalization (gain of function). Such a
pubertal development. Juvenile hemochromatosis is a partic- lesion would be expected to act dominantly and mimic hep-
ularly lethal disorder, but it can be effectively treated by phle- cidin deficiency. Its most severe form involves serine or tyro-
botomy. sine substitutions in C326 in the hepcidin-binding site of fer-
Studies of families with juvenile hemochromatosis have roportin. Affected patients presented with early onset of liver
shown that there are at least two genetic loci responsible. disease and arthritis.
Some individuals are homozygous for ablative mutations in
the hepcidin gene on human chromosome 19q. However,
Other forms of hemochromatosis
most juvenile hemochromatosis families have homozygous
or compound heterozygous mutations in hemojuvelin (also Patients have been reported with relatively mild hemochro-
called HFE2, or repulsive guidance molecule RgmC) on matosis who have no apparent mutations in the HFE, TFR2,
170 Molecular Hematology

hemojuvelin, or hepcidin genes. Recent studies implicated common (2–13%) in southern Africa and may cause mild
dominantly acting mutations in BMP6 propeptide, which resistance to hepcidin and tendency to iron overload. It is
may interfere with BMP6 secretion. not yet known whether European and American individuals
of African descent are also more susceptible to iron overload
as a result of the same iron-loading gene.
Autosomal dominant siderosis

Autosomal dominant siderosis, formerly also called auto-


Abnormal iron distribution
somal dominant (or type 4) hemochromatosis, has a dis-
tinct clinical picture. Patients with this disorder may have There are two well-characterized (but very rare) disorders
iron-deficiency anemia early in life, but later present with due to mutations in plasma proteins important in iron
(sometimes massively) increased serum ferritin concentra- metabolism. These mutations do not directly affect intestinal
tion and macrophage iron accumulation. Some may even- iron absorption. Rather, they perturb tissue iron distribution.
tually develop parenchymal iron deposition in addition to
macrophage iron loading, probably because their specific
Atransferrinemia
mutations may also cause resistance to hepcidin.
This disorder has a very interesting pathogenesis. It is Atransferrinemia is a severe deficiency of the plasma iron-
due to missense mutations in ferroportin, the cellular iron binding protein transferrin, due to mutations that truncate
exporter. This seems paradoxical at first, because the muta- or alter the coding sequence of the transferrin gene. As a
tions alter ferroportin function, and ferroportin acts as the result, erythroid precursors are iron starved and severe ane-
basolateral enterocyte transporter involved in intestinal iron mia results. Paradoxically, all non-hematopoietic tissues are
absorption (see Figure 13.1). However, it is important to con- iron loaded, probably because intestinal iron absorption is
sider that ferroportin also plays a major role in macrophage enhanced to try to provide more iron to erythroid precursors,
iron release. The lack of nonsense mutations in this disor- and because NTBI is avidly taken up by many parenchymal
der, and evidence from cellular and animal models of this cell types. Mice and humans with this disorder have low hep-
disease, suggest that the mutated ferroportin exerts a dom- cidin, indicating that iron–transferrin is an important regu-
inant negative effect on the normal version of the molecule. lator of hepcidin synthesis. This disorder can be treated by
The resulting loss of functional ferroportin is severe enough transfusion of packed red blood cells or, more appropriately,
to impair macrophage iron release, resulting in accumula- by infusion of human transferrin.
tion of iron in macrophages and a decrease in the amount
of plasma iron available to developing erythroid precursors.
Aceruloplasminemia
Iron-restricted erythropoiesis probably signals for a compen-
satory increase in intestinal iron absorption, overcoming the Aceruloplasminemia is a deficiency or absence of plasma
genetic deficiency of ferroportin in the enterocytes. It appears ceruloplasmin. Ceruloplasmin was once thought to be a
that iron loading of macrophages has few if any clinical plasma copper carrier, but it is now clear that its primary
consequences. role is as a ferroxidase, aiding in the release of iron from
macrophages, hepatocytes, and cells of the central nervous
system. Patients with this disorder are generally well early in
African siderosis
life, but gradually develop tissue iron deposition in the liver,
The pathology of African siderosis is strikingly similar to pancreas, and brain. They typically present in middle age
that of autosomal dominant siderosis due to ferroportin gene with retinal degeneration, dementia, hepatic iron deposition,
mutations, although the genetic basis of African siderosis and diabetes. Treatment with deferoxamine is ineffectual;
has not been described. Once called Bantu siderosis because treatment with normal plasma may provide some benefit.
of the population affected, this disorder was originally
attributed to excessive dietary iron intake. It is common in
sub-Saharan Africans, many of whom drink a traditional Conclusions
alcoholic beverage brewed in non-galvanized steel drums.
The iron content of the brew is substantial, resulting in Iron disorders are among the most common of human
massive iron ingestion. However, the observations that not afflictions. They invariably result from abnormalities of iron
all drinkers develop iron overload and that some individuals balance, most often due to defects in the iron-regulatory
develop similar iron overload without drinking the beverage hormone hepcidin, its receptor/iron channel ferroportin, or
support the notion that there is a genetic component to their interaction. Iron overload is underdiagnosed because
this disorder. The ferroportin mutation Q248H is relatively it produces signs and symptoms that are common in adult
The molecular basis of iron metabolism 171

populations. However, iron overload disorders are usually Nicolas, G., Chauvet, C., Viatte, L. et al. (2002). The gene encoding the
easy to treat and clinicians should be vigilant in considering iron regulatory peptide hepcidin is regulated by anemia, hypoxia, and
them. inflammation. J. Clin. Invest. 100: 1037–1044.

Iron overload disorders


Further reading
Bardou-Jacquet, E. and Brissot, P. (2014). Diagnostic evaluation of
hereditary hemochromatosis (HFE and non-HFE). Hematol. Oncol.
General Clin. North Am. 28: 625–635.
Coffey, R. and Ganz, T. (2017). Iron homeostasis: an anthropocentric Beutler, E. (2006). Hemochromatosis: genetics and pathophysiology.
perspective. J. Biol. Chem. 292: 12727–12734. Annu. Rev. Med. 57: 331–347.
Hentze, M.W., Muckenthaler, M.U., Galy, B., and Camaschella, C. Kanwar, P. and Kowdley, K.V. (2013). Diagnosis and treatment of heredi-
(2010). Two to tango: regulation of mammalian iron metabolism. Cell tary hemochromatosis: an update. Expert Rev. Gastroenterol. Hepatol.
142: 24–38. 7: 517–530.
Pietrangelo, A. (2007). Hemochromatosis: an endocrine liver disease.
Hepatology 46: 1291–1301.
Regulation of iron homeostasis
Babitt, J.L., Huang, F.W., Wrighting, D.M. et al. (2006). Bone morpho- Abnormal iron distribution
genetic protein signaling by hemojuvelin regulates hepcidin expres-
sion. Nat. Genet. 38: 531–539. Bartnikas, T.B. (2012). Known and potential roles of transferrin in iron
Canali, S., Zumbrennen-Bullough, K.B., Core, A.B. et al. (2017). biology. Biometals 25: 677–686.
Endothelial cells produce bone morphogenetic protein 6 required for Xu, X., Pin, S., Gathinji, M. et al. (2004). Aceruloplasminemia: an inher-
iron homeostasis in mice. Blood 129: 405–414. ited neurodegenerative disease with impairment of iron homeostasis.
Nemeth, E., Tuttle, M.S., Powelson, J. et al. (2004). Hepcidin regulates Ann. N.Y. Acad. Sci. 1012: 299–305.
cellular iron efflux by binding to ferroportin and inducing its inter-
nalization. Science 306: 2090–2093.
Chapter 14 Hemoglobinopathies due
to structural mutations
D. Mark Layton & Steven Okoli
Center for Hematology, Imperial College London, London, UK

Introduction, 173 Homozygous HbE and Hb E/β thalassemia, 187


Normal hemoglobin structure and function, 173 Unstable hemoglobins, 188
Sickle cell anemia, 175 High oxygen affinity hemoglobins, 188
Sickle/β thalassemia, 184 Low oxygen affinity hemoglobins, 189
Hemoglobin C disease, 185 M hemoglobins, 189
Hemoglobin SC disease, 185 Conclusions, 190
Other sickle hemoglobinopathies, 186 Further reading, 191

The shape of the oxygen equilibrium curve of hemoglobin


Introduction is sigmoid. This shape is determined by the extent of
cooperativity, a phenomenon whereby the hemoglobin
This chapter discusses those hemoglobin disorders that are
molecule modulates its affinity for oxygen when partially
caused by mutations in the exon (i.e. coding) portion of the
saturated. The first portion of the curve has a gentle slope,
α or β globin genes. Alterations of globin gene expression
reflecting hemoglobin’s low affinity for oxygen at initiation
(thalassemias) are reviewed in Chapter 1. We confine our-
of the loading process. In other words, when hemoglobin is
selves mainly to the structural variants that are responsible
totally deoxygenated, it has a rather poor avidity for oxygen
for clinically significant hemoglobinopathies.
(Figure 14.2). As loading proceeds and hemoglobin binds
more oxygen molecules, the slope of the curve steepens,
Normal hemoglobin structure and indicating that the affinity for oxygen has increased. This
function occurs after two molecules of oxygen have bound to the
heme of deoxyhemoglobin tetramers, a property which
The adult major hemoglobin molecule, HbA, is a tetramer helps hemoglobin to rapidly become fully oxygenated. If red
formed by four polypeptide chains: two α chains of 141 cells are exposed to sufficient oxygen to saturate only half of
amino acids each and two β chains of 146 amino acids each. the hemes available, most molecules will either not be oxy-
Each globin chain harbors a prosthetic group (heme) formed genated at all or will be entirely oxygenated, with a very small
by protoporphyrin IX in a complex with a single iron atom component of partially oxygenated molecules. The sigmoid
(Figure 14.1). The heme, embedded in globin, is surrounded shape of the oxygen dissociation curve allows hemoglobin
by a hydrophobic niche which favors maintenance of iron in to release oxygen efficiently. Abnormal hemoglobins such as
the ferrous state. The heme pocket is large enough for oxygen Hb Bart’s (γ4 ) which lack cooperativity are unable to deliver
to penetrate, but larger ligands capable of binding to the iron, oxygen to the tissues efficiently.
such as carbon monoxide, have difficulty gaining access. At the molecular level, cooperativity is accounted for by
Oxygen transport to tissues, the principal function of the fact that hemoglobin exists stably in only two structural
hemoglobin, depends on blood flow, which in turn is forms, the R (relaxed) and T (tense) states, which facilitate
governed by cardiac output and microcirculatory resistance the switch between the oxy and deoxy states. Transition from
and distribution, and by hemoglobin concentration. Oxy- the deoxy (T) to the oxy (R) state occurs when a molecule
gen extraction by the tissues depends on the shape of the of hemoglobin has bound two or three molecules of oxygen.
oxygen dissociation curve of hemoglobin and on tissue Po2 . While this concept has been challenged, postulated interme-
diate stable states have not been generally recognized.
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. The heme-triggering mechanism for conformational
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. change has been resolved. The iron in deoxyhemoglobin is

173
174 Molecular Hematology

Hemoglobin tetramer

β2 β1
F'

A F A' F
E'
β2 β1
A
H'
H H'
B E'
E G G' H H' E
C
E'
GH C'
B
G
C' G' C
D' B D
F' E
H'
H E
H'
H'
E' F A
A' E' F'
A
A' α1 α2

α2 α1

(a) Front view (b) Side view


Fig. 14.1 Hemoglobin molecule. (a) The front view depicts the hemoglobin tetramer and the three axes of symmetry. The vertical line (marked
by a solid ellipse) tracks the true twofold axis of symmetry (if you look down this axis you will see first the central cavity constituted by the β
chains). The dashed ellipses and dashed lines mark the two pseudoaxes of symmetry, since the symmetry is only approximate. Only the 21 carbons
are shown, with none of the side chains. Numbers in bold type depict the residues in direct contact between the α1 and β2 chains. The α1 β2
dimer (green) never dissociates and interacts with the α2 β1 dimer (light shading) to change the conformation from T to R. Alterations in this area
can produce high- or low-affinity hemoglobins. (b) The side view of the tetramer depicts the stable dimer, the one that does not move or dissociate.

Horse methemoglobin Horse deoxyhemoglobin

α2 α2 β1
β1

β2 α1 α1
β2

Fig. 14.2 The allosteric transition of hemoglobin: R → T. Right: Deoxygenated (T, tense) conformation of hemoglobin. Notice that the center
cavity (space between the two β chains, indicated in green) in the hemoglobin molecule is larger than in the oxygenated tetramer (left). This space
is occupied by 2,3-diphosphoglycerate (2,3-DPG) when the tetramer is in the blue cell. The allosteric effector, 2,3-DPG, is in a little over equimolar
concentration with the tetramer. The T form has low affinity for oxygen. Left: Oxygenated (R, relaxed) conformational state of hemoglobin. The
central cavity has been reduced in size by the movement of the β chains toward each other. This tetramer cannot bind 2,3-DPG. The R form has
high affinity for oxygen. The change in conformation forms and breaks bonds between the β chains (green) and the α chains (light shading) on
each side of the central cavity. The title says “methemoglobin” but this hemoglobin form is identical to oxyhemoglobin.
Hemoglobinopathies due to structural mutations 175

slightly out of the plane of the heme (domed configuration)


because the pyrrole rings are also slightly pyramidal. When
the ligand binds the sixth coordinating position of the iron,
significant steric stresses are introduced, and to relieve this
strain the distal histidine moves 8° to become perpendicular
to the heme, significantly decreasing the doming of the
iron (the angle between iron and the heme decreases to 4°).
There is also the displacement of FG5 in the same direction
of the histidine F8 (a conserved residue to which heme iron
is covalently linked in all hemoglobins). The configuration
around the heme has now changed to the oxygenated
(R state), and a chain of events takes place involving the
critical interactions that change the conformation of the
hemoglobin tetramer.
Several heterotropic ligands within the red cell shift
the oxygen dissociation curve to the right, toward lower
oxygen affinity. These include CO2 , hydrogen ions, and
2,3-diphosphoglycerate (2,3-DPG). Hemoglobin binds CO2
while it is offloading oxygen and releases CO2 when it binds
oxygen, serving to dissipate the increase in concentration of
CO2 in the tissues and conveniently delivering this metabolic
Fig. 14.3 Sickled cell. This is an HbS homozygous (SS) red cell that
end product to the alveoli of the lungs. It accomplishes this
has been deoxygenated. Notice the digitations stemming in all
particular task with ease, because CO2 inhibits hemoglobin’s directions. These digitations are the product of the presence of
oxygen-carrying capacity by decreasing the oxygen affinity of fascicles of fibers, which are the polymerized form of deoxy HbS.
the molecule.
Hemoglobin binds hydrogen ions more efficiently in a low-
pH environment. The Bohr effect describes the changes in proposed that sickling reflected an abnormality of the
oxygen affinity secondary to pH changes within a certain hemoglobin molecule, based on the observation of Irving
range: the lower the pH, the lower the oxygen affinity or the Sherman, then a medical student, that under the polarizing
higher the P50 . This means that an increased concentration of microscope sickle cells, induced by deoxygenation, exhibited
protons favors a low-affinity state in hemoglobin. The phys- birefringence. Birefringence signified to Pauling that some
iological advantage this confers is twofold: in the tissues the form of molecular alignment or orientation existed in these
lower pH consequent on higher CO2 tension favors oxygen cells, and since the predominant molecule in the red cell
unloading, whereas in the lungs there is an increase in pH is hemoglobin, it had to be this that was involved in the
with CO2 release which enhances oxygen uptake. pathology. Electrophoretic studies, which demonstrated
the altered mobility of sickle hemoglobin (HbS), confirmed
this interpretation and the concept of molecular disease
Sickle cell anemia was born.
The chemical structure of HbS was defined by Vernon
Ingram, who had developed a technique for probing the pri-
Genetics
mary sequence of a protein based on its fingerprint after
The genetic basis of sickle cell anemia is central to the history cleavage by trypsin. This revealed that the structure of HbS
of medical genetics. Pre-dated for generations by vernacular differed from normal hemoglobin by only one tryptic pep-
descriptions among West African tribes, the first account tide, subsequently found to have a single amino acid change
of sickle cell disease (SCD) in Western medical literature is at position 6 of the β chain, where a polar glutamic acid
attributed to James Herrick, the Chicago physician, who in residue was replaced by a non-polar valine. At the DNA
1910 observed sickle-shaped red cells (Figure 14.3) in the level this corresponds to an A → T transversion at codon 6
blood of a dental student from Grenada who suffered from (GAG → GTG) of the β chain.
chronic hemolytic anemia. In 1947 James Neel concluded The globin genes were among the first in the human
that with respect to the mode of inheritance, sickle cell genome to be cloned. The β and β-like globin genes are
anemia was a homozygous state and sickle cell trait (the located on the short arm of chromosome 11 at 11p15.5 and
asymptomatic carrier state) the heterozygous state for a the α and α-like genes at chromosome 16p13.3. Utilizing
genetic character that had yet to be defined. Linus Pauling polymorphic restriction sites distributed across the β globin
176 Molecular Hematology

gene cluster, it has been established that in Africa the βS gene fitness in the homozygous state (balanced polymorphism).
is associated with four distinct chromosomal haplotypes, This selection pressure in favor of HbS heterozygotes is
Benin, Bantu or Central African Republic, Senegal, and attributable to partial protection against the life-threatening
Cameroon, named after the geographical areas in which the complications of Plasmodium falciparum malaria, with a
haplotypes are found (Figure 14.4). The geographical reach consequent reduction in mortality. The exact mechanism(s)
of the sickle mutation spread further with the discovery of of protection has yet to be fully elucidated.
a different haplotype linked to the βS gene in the eastern The sickle gene has also spread through gene flow; for
oasis of Saudi Arabia and among tribal population groups example, the βS allele linked to the Benin haplotype has
in central India. It was postulated this Indo-European sickle spread to North Africa, Sicily, Greece, Turkey, most of the
mutation originated in the Harappa people of the Indus Arab world, and the Americas, through the vicissitudes of
Valley, and then dispersed, probably during the Sassanian war (e.g. Sudanese troops in Sicily during the Arab conquest),
Empire, to regions in the Middle East (now eastern Saudi commerce, and the horrors of the Atlantic slave trade. It
Arabia, Bahrain, Kuwait, and Oman). Based on these haplo- is estimated that worldwide approximately 300 000 children
types a multicentric model has been proposed in which the with sickle cell anemia are born annually, and that the figure
sickle gene arose independently around the world on at least could rise to 400 000 by 2050.
five occasions. This has recently been challenged by analysis Despite its simple Mendelian inheritance, the clinical
of whole-genome sequencing data which suggest a single course of SCD is extremely heterogeneous. The basis for
origin for the sickle mutation approximately 7300 years the striking variability in the clinical course and severity
ago. The present-day gene frequency, which equates to of sickle cell anemia has been the subject of intense study.
carrier rates that reach 25% in sub-Saharan Africa, reflects Both genetic and non-genetic factors are important, though
a heterozygote advantage that outweighs the reduced their relative contribution to disease severity is still not

Senegal Arab-
India
2
Benin

3 Bantu

60 50 40 30 20 10 0 Kb

ε Gγ Aγ ψβ δ β
5ʹ 3ʹ
Fig. 14.4 The β-globin gene cluster
haplotypes linked to βS in Africa and the
Hinc Xmn Hind Pvu Hinc Hinf Ava Hpa Bam HI Middle East/Pakistan/India. At the top of the
II I III II II I II I figure are shown the geographical distributions
Hgi
AI Gγ corresponding to the haplotypes described
underneath. A haplotype is a particular array of
SS Benin – – – – + – + – ++ – + polymorphic sites (i.e. sites that vary among
SS Bantu – – + – + – – – ++ ++
individuals), defined here by the capacity of
SS Senegal – + + – + + + + ++ ++ endonuclease enzymes to recognize the short
SS Arab-India + + + – + + + + –
sequence and cut the DNA.
Hemoglobinopathies due to structural mutations 177

well defined. A pilot study of identical twin pairs revealed important implications for the development of strategies for
concordance in growth, hematological parameters, and HbF induction to treat β hemoglobinopathies.
certain complications, such as priapism and splenomegaly, Alpha thalassemia, an extremely common genetic
but variance with respect to other clinical manifestations. variant in populations where the βS allele is prevalent,
Socioeconomic status and access to medical care as well as exerts a beneficial effect through lowering intracellular
the prevalence of infections that carry increased morbidity HbS concentration, a key determinant of the rate of poly-
and mortality in SCD are important variables. Increased fetal merization. Individuals who co-inherit sickle cell anemia
hemoglobin (HbF) production is a major ameliorating factor and α-thalassemia have higher hemoglobin levels and a
in SCD. The switch from predominant expression of HbF to lower risk of complications linked to hemolysis, including
adult hemoglobin (HbA) after birth coincides with the stage stroke, priapism, leg ulcers, nephropathy, and cholelithiasis.
at which children with SCD first develop symptoms, as noted Unfortunately, the favorable impact of α-thalassemia is
by Janet Watson in 1948. Data from the large prospective offset by an increased risk of vaso-occlusive crises and
Cooperative Study of Sickle Cell Disease (CSSCD) showed avascular necrosis, possibly due to the higher hematocrit.
higher levels of fetal hemoglobin were independently associ- There are conflicting data on the effect of α-thalassemia
ated with improved survival and reduced complication rates. on mortality in SCD, though several studies describe an
This beneficial effect was also observed in the Jamaican increase in prevalence of α-thalassemia with age, implying
SCD cohort. Patients with a steady-state Hb F level of 8–9% an association with improved survival.
or more have a more favorable course. A further striking It is inevitable genetic factors beyond effects on
example is found in compound heterozygotes for the βS hemoglobin synthesis contribute to the clinical hetero-
allele and deletional forms of hereditary persistence of geneity of SCD given its complex pathophysiology. A
fetal hemoglobin, whose red cells contain 20–30% HbF. substantial number of genetic association studies have been
Despite most of their remaining hemoglobin being HbS, conducted, but with few exceptions the discovery of genetic
they are largely asymptomatic. The protective effect of HbF modifiers has proved challenging (reviewed in Lettre 2012).
is attributable to differences in the amino acid sequence of Convincing associations have been shown for polymor-
γ-globin and β-globin that affect the biophysical properties phism of the UGT1A1 gene and risk of cholelithiasis, as
of the corresponding hemoglobin tetramer. The γ-globin well as the increased risk and progression of nephropathy
chain has a glutamine rather than a threonine residue at among individuals with SCD who are homozygous or
position 87, which weakens hydrophobic interactions impor- compound heterozygous for the G1 or G2 variant alleles of
tant for stabilization of the HbS polymer. Consequently, HbF the APOL1 gene, found at high frequency in some African
as well as the hybrid tetramer HbFS (α2 γβS ) have a low populations. Causality is less clear in other cases. An SNP
probability of copolymerization with HbS. located upstream of COMMD7, a gene expressed in the lung
To date, three quantitative trait loci associated with implicated in NF-ΚB signaling, and HMOX1 gene promoter
variation in HbF level in patients with SCD have been polymorphisms that alter heme oxygenase activity have
identified. The first of these, a single-nucleotide polymor- been associated with acute chest syndrome. A genome-wide
phism (SNP) in the promoter of HBG2, the gene which association study in a large group of children with SCD
encodes G γ-globin, identified by the restriction enzyme enrolled in the CSSCD and Silent Infarct Transfusion
Xmn I, is linked to the Senegal and Saudi Arabian-Indian (SIT) trial revealed preliminary evidence of an association
βS haplotypes associated with higher HbF levels. Only part between an SNP at a locus on chromosome 4q27 previously
of the difference in HbF levels observed in SCD is accounted associated with autoinflammatory disorders and acute,
for by genetic variation at the β globin locus. Genome-wide severe vaso-occlusive pain. Recently, differences in blood
association studies have identified two further quantitative mononuclear transcriptomic profile have been found to
trait loci, the intergenic region between HBS1L and MYB correlate with clinical severity of SCD and, in conjunction
on chromosome 6q23 and BCL11A on chromosome 2p16, with key clinical markers, to be superior in prediction of
which account for variability in HbF in normal non-anemic mortality than clinical criteria alone. This represents a step
populations and up to 50% of that seen in patients with toward the discovery of much-needed biomarkers to aid
sickle cell anemia. The HBS1L-MYB intergenic region has disease and treatment stratification.
been shown to regulate erythropoiesis and HbF production,
though the functional variant at this locus has yet to be
Clinical features
defined. BCL11A encodes a zinc finger protein that represses
transcription of the HBG1 and HBG2 genes. SNPs associated Sickle cell anemia refers to the homozygous state for the βS
with HbF level co-locate with an erythroid-specific enhancer gene, in which the majority of the hemoglobin in the red cells
in intron 2 of BCL11A that is essential for gene expression. is HbS. Polymerization of HbS when ambient oxygen ten-
The mechanisms that underlie these quantitative traits have sion is reduced induces sickling (marked changes in red cell
178 Molecular Hematology

shape and deformability with the formation of irreversibly and ultimately vaso-occlusion. This may be an oversimpli-
sickled cells, ISCs), leading to decreased pliability, lowering fication and other mechanisms, for example reflex shunt-
of rheological competence, and hemolysis. In addition, there ing of blood away from the bone marrow and perturbation
are myriad pleiotropic effects, including increased adherence of vasoregulation, have been invoked. Precipitating factors
of sickle and other blood cells to endothelium, induction of include cold, infection, dehydration, altitude, excessive exer-
red cell dehydration, pro-coagulability, and perturbance of cise, emotional stress, sleep apnea, obstruction to the periph-
vasoregulation due to reduced nitric oxide (NO) bioavailabil- eral circulation (e.g. tourniquet use), and possibly cocaine
ity. As discussed, the primary pathophysiological events as abuse.
well as the pleiotropic effects are subject to modification by The clinical complications of sickle cell anemia are pro-
epistatic genes, which may be polymorphic. The combined tean (Figure 14.5) and involve almost every organ system.
impact of all these factors accounts for the great interindi- The lungs and kidneys are particularly affected. There is
vidual variability of the clinical phenotype: some patients are an increase in glomerular filtration rate as well as hypos-
severely affected, while others have only mild disease, and the thenuria from early childhood. Papillary necrosis with hema-
majority span these two extremes. turia, renal insufficiency, and nephrotic syndrome may occur
Anemia is present in all cases, with a mean hemoglobin (Figure 14.6). The lungs are the target of both acute and
concentration of around 8 g dl−1 (higher levels occur in chronic complications. Acute chest syndrome may be asso-
Indian and some Middle Eastern populations), reticu- ciated with infection (viral, bacterial, or atypical organisms)
locytosis (typically 5–20%), dense cell fraction – mean and fat embolism. The latter is associated with a more severe
corpuscular hemoglobin concentration (MCHC) >38 g dl−1 clinical course. Repeated episodes of acute chest syndrome
or density >1.1009 – of up to 40%, an elevated white cell predispose to the development of chronic sickle lung dis-
count (mean 18 × 109 l−1 ), and commonly thrombocytosis. ease. Cerebral vasculopathy may lead to stroke, silent cerebral
Anemia is tolerated relatively well due to the reduced oxygen infarction, and neurocognitive defects. From infancy there
affinity of HbS and increased 2,3-DPG concentration within is progressive splenic atrophy, against which co-inheritance
the red cell. Several hematological variables, including of α thalassemia affords protection. The resulting loss of
total hemoglobin, are modulated by the level of HbF and splenic reticuloendothelial function leads to susceptibility to
co-inheritance of α thalassemia (−α/−α genotype more so infection with capsulated bacteria. Before the advent of new-
than −α/αα), which occurs at a significant frequency among born screening and prophylaxis, overwhelming Streptococcus
sickle cell anemia patients (20–50% of cases). Higher levels pneumoniae infection was the leading cause of death in chil-
of HbF are associated with the Senegal and Arab-Indian dren with SCD.
β globin haplotypes. Gender also exerts an influence; for The prevalence of avascular necrosis of the femoral or
example, females with sickle cell anemia have a significantly humeral head reaches 20% in adulthood and is greater
higher HbF level. In males hemoglobin concentration among patients whose hematocrit is higher, including those
increases between 10 and 24 years, contributing to a higher with coexistent thalassemia. Peripheral retinopathy, charac-
frequency of vaso-occlusive crises. terized by microvascular occlusion and choroidal infarcts, is
The cardinal manifestation of sickle cell anemia is the common in SCD. Abnormal arteriovenous communications
painful crisis. This commonly involves multiple sites in the develop at the border of vascular and ischemic retina and are
extremities, joints, spine, abdomen, ribs, and sternum. The the precursor of proliferative sickle retinopathy. In patients
onset of pain can be insidious or rapid, and may affect sev- with sickle cell anemia, and even heterozygotes, glaucoma
eral sites simultaneously or extend over time from one loca- may occur after traumatic hyphemas, since the hypoxic
tion to others. The pathological basis of the painful crisis milieu of the anterior chamber favors sickling of red cells,
is infarction of the bone marrow. Systemic embolization of which are unable to exit through the canal of Schlemm. As in
bone marrow or fat to the lung, brain, or kidneys may occur, other chronic hemolytic anemias, cholelithiasis is common.
and is an ominous development that may herald acute chest The prevalence of gallstones increases with age and is also
syndrome or multiorgan failure. The site first affected in correlated directly with the number of TA repeats in the
infancy is often the hands or feet (dactylitis). Necrosis of promoter of the hepatic uridine diphosphate glucuronosyl-
the epiphysis may lead to premature fusion with shorten- tranferase (UGT1A1) gene, and inversely with the presence
ing of the digits. Paradoxically, painful crises are more fre- of α-thalassemia. Hepatopathy in sickle cell anemia encom-
quent in patients with greater red cell deformability. Serial passes several entities that include intrahepatic cholestasis
observations indicate that during painful crises a decrease and hepatic sequestration. In the latter, rapid enlargement of
in low-density cells of greater deformability precedes that of the liver is associated with a precipitous fall in hemoglobin
dense cells. This may be reconciled with a model whereby akin to that in acute splenic sequestration.
cells that are less dense but more adherent to the endothe- Congestive cardiac failure may develop in the absence of
lium bedeck the vessels, facilitating entrapment of dense cells other recognized causes in sickle cell anemia. This implies
Hemoglobinopathies due to structural mutations 179

Pain
Dactylitis
Long bones
Trunk

Sequestration
Splenic
Hepatic
Chest syndrome
Mesenteric syndrome

Infection
Pneumococcal
Parvovirus
Salmonella

Priapism

Upper airway obstruction

Stroke
Subarachnoid hemorrhage
Retinopathy

Gallstones
Avascular necrosis

Hyposthenuria

Delayed growth and development

Leg ulcers

Fig. 14.5 Age dependency of complications in Chronic renal failure


sickle cell anemia. Chronic sickle lung
Source: Davies S.C., Oni L. (1997). Management of
patients with sickle cell disease. BMJ, 315:
656–660. Reproduced with permission of British 0 5 10 15 20 25 30
Medical Journal. Age (years)

that the myocardium may be the target of vaso-occlusive


damage, although overt myocardial infarction is not com-
mon. The infrequency of vaso-occlusion in the heart may
be the consequence of the very high perfusion pressure of
coronary vessels, as well as the squeezing effect of ventricular
contraction. Pulmonary arterial hypertension has emerged as
a common and serious complication. Estimated pulmonary
artery pressure is increased in up to 30% of sickle cell anemia
patients based on echocardiographic findings. Multiple stud-
ies have shown this to be associated with an increased risk of
death, though the magnitude of risk varies between studies.
In a study conducted at the National Institutes of Health with
a median follow-up of four years, the mortality in patients
with a tricuspid regurgitant velocity ≥2.5 m s−1 was 36%.
In sickle cell anemia, pregnancy poses a significant risk to
Fig. 14.6 Renal medulla of transgenic mice expressing βS and the mother and fetus. The role of prophylactic transfusion
βS-Antilles . Note the extensive obstructive aggregation of sickle red during pregnancy remains controversial and many centers
cells in the renal medullary vasculature. adopt a selective approach, allied to close monitoring of
180 Molecular Hematology

mother and fetus in the context of a high-risk obstetric propagation of the polymer ensues at an exponential rate.
program, with generally favorable outcomes. The self-association of deoxy HbS molecules to form a
nucleus is a stochastic process, hence the reaction has a
delay time. This is shortened by an increase in intracellular
Pathophysiology
hemoglobin concentration, lowering the pH, and an increase
There are five major contributors to the pathophysiology of in temperature, which may be one reason dehydration and
sickle cell anemia. infection trigger vaso-occlusive events in sickle cell disor-
ders. The final product of polymerization is a rope-like fiber
composed of seven double strands twisted helically around a
Polymerization of HbS
vertical axis. Depending on their orientation within the cell,
The substitution of valine for glutamic acid at β6 sub- these polymers deform the shape of the red cell to produce
verts an essential property of hemoglobin, that it should one of several morphological forms (Figure 14.7).
maintain solubility at high concentration within the red The intermolecular contacts within and between the
cell. The initial step in the polymerization of HbS, known double strands have been extensively characterized. Lateral
as nucleation, involves the assembly of small aggregates contact between adjacent tetramers in the double strand
of deoxy HbS molecules. Once nucleation has occurred, involves the mutant β6 Val, which is buried in a hydrophobic

Strand 2

Strand 1

(a)

(c) 1β2 2β1


1α2
Axial
1β1
(b)

2β2
(d)
1β1

2α2
Lateral 2β1 Axial

Fig. 14.7 HbS polymer structure. This is


the assembly unit of the HbS polymer: the
Werner–Love double strand. This double strand is
part of the HbS crystal, but in the crystal it
propagates in all directions to form a lattice. The
sickle fiber wraps around to form a helical fiber. The
squares depict the four different areas of contact
that form the double strand.
Hemoglobinopathies due to structural mutations 181

acceptor pocket formed by β88 Leu and β85 Phe, allowing integrin, which mediates adhesion to the endothelium. The
ingress of water molecules which form bridging hydrogen expression of P-selectin by activated endothelium mediates
bonds that stabilize the contact. The failure of HbF and HbA2 abnormal rolling and adhesion of sickle red cells and initi-
(and the corresponding hybrid tetramers) to copolymerize ates neutrophil binding. Intravital microscopy in a murine
with HbS has been attributed to specific amino acid residues, sickle model has shown that following an inflammatory stim-
but how these disrupt axial and lateral contacts in the HbS ulus, neutrophils are recruited to the endothelium of post-
polymer is still not fully resolved. The reduced oxygen capillary venules, where they capture sickle red cells, possibly
affinity of HbS favors the formation of deoxy-HbS and this is through recognition of surface IgG, complement, or inter-
exacerbated by the high 2,3-DPG content of sickle red cells. cellular adhesion molecule (ICAM)-4 (Landsteiner–Weiner
Elevated sphingosine kinase activity may also contribute blood group antigen).
through the generation of sphingosine-1-phosphate, which Platelets also contribute to the vaso-occlusive process. The
also lowers hemoglobin oxygen affinity. platelets of patients with sickle cell anemia show increased
expression of activation and adhesion markers, includ-
ing activated αIIbβ3 integrin (glycoprotein IIb/IIIa) and
Adhesion of sickle red cells and other blood cells
P-selectin (CD62P), which mediate the formation of circu-
to the endothelium
lating heterocellular aggregates with red cells, monocytes,
Based purely on the kinetics of HbS polymerization, it is pre- and neutrophils. Thrombospondin released from activated
dicted that the majority of red cells will not undergo sickling platelets and shown to be elevated in plasma during painful
in vivo, because the delay time for polymerization exceeds the crisis may bridge CD36 or αVβ3 integrin expressed on
time taken to traverse the microvascular and venous circula- microvascular endothelial cells and CD36 or sulfated gly-
tion and reach the lungs, where reoxygenation occurs. From cans on sickle red cells. Increased plasma levels of platelet-
this has emerged the concept that sickle red cells are more derived soluble CD40 ligand, which potentially contribute to
adherent to the vascular endothelium, which has shaped our endothelial activation and pro-coagulant activity, are found
understanding of the pathophysiology of sickle cell anemia. in patients with sickle cell anemia and correlate with the fre-
Adhesion of sickle red cells to the vascular endothelium plays quency of acute pain episodes.
a key role in vaso-occlusion by retarding transit, thereby Adhesive interactions represent a promising target for
favoring HbS polymerization and contributing to mechani- therapeutic intervention in SCD. Transgenic sickle mice that
cal obstruction. lack E-selectin and P-selectin are protected against vaso-
In early studies Hebbel and colleagues, originally using occlusion. Several clinical trials of selectin blockade are now
human umbilical vein endothelial cell monolayers, estab- underway (see later). Von Willebrand factor and αVβ3 inte-
lished that sickle red cells were abnormally adherent to grin appear to be other good candidates, since antibodies
endothelial cells, and postulated that this was linked to dis- against both inhibit cell adhesion in living circulatory prepa-
ease severity. The enhanced adherence of sickle red cells has rations. Endothelial injury, which may be mediated by sickle
been reproduced in more physiological systems. The mech- cells or NO depletion, leads to activation of endothelial cells,
anisms proposed for these adhesive interactions are complex with upregulation of the transcription factor nuclear factor
and full discussion is beyond the scope of this chapter. Not (NF)-κB and enhanced expression of multiple cell adhesion
all have been validated in microcirculatory preparations or molecules, and establishes a vicious circle that potentiates the
proven to be relevant to the microvascular bed in which vaso- adherence of sickle red cells and vaso-occlusion.
occlusion occurs.
Sickle reticulocytes, which express the integrin α4β1 very
Red cell dehydration
late antigen 4 (VLA-4) that binds to the vascular cell adhe-
sion molecule (VCAM)-1 on the surface of endothelial cells Red cell dehydration represents an integral component of
and the subendothelial protein fibronectin, show increased the pathophysiology of sickle cell anemia, because poly-
adherence to the endothelium of post-capillary venules, the merization of HbS is critically dependent on intracellular
principal site of vaso-occlusion. Increased expression of other hemoglobin concentration. Although the MCHC is close to
adhesion molecules, including platelet glycoprotein (CD36) normal, sickle red cells have a wide density distribution.
and basal cell adhesion molecule (BCAM), by sickle red cells After they enter the circulation, a proportion of young sickle
promotes their adherence to the endothelium. Obstruction red cells undergo rapid dehydration, which accelerates HbS
of the microcirculation is preceded by the adhesion of young polymerization with conversion to dense cells and ultimately
sickle red cells, which trap dense cells and ISCs in those areas ISCs. Cell dehydration is due to loss of potassium accompa-
to which they are adherent. Leukocytes also participate in nied by water efflux.
vaso-occlusion. Circulating activated leukocytes are present Three membrane transport pathways have been implicated
in sickle cell anemia and show increased expression of αMβ2 in the dehydration of sickle red cells. The best characterized
182 Molecular Hematology

are the Ca2+ -dependent K+ (Gardos) channel and K+ /Cl− The hemolysis is also complex because of the heterocellular
cotransport. In addition, perturbance of cation homeostasis nature of red cells in sickle cell anemia. There are actually
in sickle cell anemia is due to a less well-defined conduc- three red cell populations: the most dense red cells, which
tance pathway, referred to as Psickle, that may reflect abnor- survive only 4–5 days; F cells (which contain predominantly
mal function of the mechanosensitive ion channel PIEZO1, HbF), which have close to a normal lifespan; and the remain-
defects of which cause hereditary xerocytosis (stomatocyto- ing majority of red cells, which have an intermediate survival.
sis). Activation of K+ /Cl− cotransport occurs in sickle cell Coexistent α thalassemia or high HbF levels reduce
anemia as well as other hemoglobinopathies due to posi- hemolysis and consequently anemia. Interestingly, the
tively charged hemoglobin variants such as HbC. It has been level of HbF mirrors the pattern of circulating progenitors
proposed that this is mediated by binding of the abnormal and hematopoietic cytokines seen in sickle cell anemia.
hemoglobin to the inner surface of the cell membrane. In Low-HbF sickle cell anemia patients have an increased
normal red cells K+ /Cl− cotransport is confined to reticu- number of peripheral blood burst-forming units-erythroid
locytes, but in sickle cell anemia activity persists in mature (BFU-E), which are in active cycle. In addition, these
red cells. The Ca2+ -dependent K+ (Gardos) channel is acti- patients have constitutively elevated levels of erythropoietin,
vated by intracellular accumulation of Ca2+ , which occurs on granulocyte/macrophage colony-stimulating factor, and
deoxygenation in sickle red cells. Cells containing HbF are stem cell factor, and lower levels of the inhibitory cytokine
less susceptible to this effect, suggesting it is due to polymer- transforming growth factor (TGF)-β, presumably reflecting
ization. When red cells undergo sickling, there is increased the need to maintain a more intense erythropoietic drive.
cation permeability due to distortion of the membrane suffi- Sickle cell anemia patients are susceptible to an acute anemia
cient to activate the Ca2+ -dependent K+ (Gardos) channel, superimposed on that resulting from chronic hemolysis,
leading to K+ efflux and cell dehydration. Dense cells and due to infection with parvovirus B19. This can produce
ISCs have markedly shortened survival and reduced oxygen a life-threatening fall in hemoglobin, which is generally
affinity. self-limiting (after 7–15 days). This results from the ability
of parvovirus B19 to infect BFU-E through the blood group
P system that serves as its receptor.
Anemia

Anemia is not simply a product of hemolysis in sickle cell


Nitric oxide effects
anemia. The extent of marrow expansion is less than in
thalassemia major (five as opposed to ten times normal). In recent years, a new paradigm linking vascular dysfunction
One explanation for this is the right shift in the oxygen equi- and hemolysis in the pathophysiology of sickle cell anemia
librium curve, to which high red cell 2,3-DPG contributes, has emerged. This is underpinned by evidence that in sickle
as described earlier. This results in increased oxygen delivery cell anemia, even in the steady state, the bioavailability of NO
to the tissues, and explains why erythropoietin levels do is decreased, leading to perturbance of vascular homeostasis.
not increase until the hemoglobin falls below 9 g dl−1 . This arises because free hemoglobin, released into the plasma
Even below this threshold, however, the erythropoietin as a consequence of intravascular hemolysis at a rate which
response is blunted (i.e. inappropriately low for the degree exceeds haptoglobin binding capacity, reacts rapidly with NO
of anemia). The reason for this is not fully understood, produced by the endothelium to form methemoglobin and
but the fact that this relative deficiency of erythropoietin nitrate. This scavenging mechanism does not occur under
increases with age suggests that occult renal damage might be physiological conditions because of the diffusion barriers
responsible. presented by the red cell membrane and the cell-free zone
Hemolysis results from damage to and dysfunction of adjacent to the endothelium.
the red cell membrane caused by polymerization of HbS. The release of arginase-1 contained in red cells compounds
Oxidative damage to membrane proteins and exposure of the effect on NO bioavailability through the conversion of l-
phosphatidylserine on the outer surface of the red cell pro- arginine, the substrate for NO synthase, to l-ornithine. Renal
mote phagocytosis by macrophages. Sickle red cells are under dysfunction in sickle cell anemia may further compromise
increased oxidative stress, the mechanism of which is com- the supply of arginine, as the kidneys are the primary site of
plex and includes the generation of superoxide and hydrogen de novo synthesis. Low plasma arginine levels are found in
peroxide from the autoxidation of intracellular and extracel- adults with SCD and predict early mortality. Hemolysis also
lular HbS (the latter being amplified in the absence of cellular leads to release of asymmetric dimethylarginine (ADMA)
antioxidant systems), generation of hydroxyl radicals via the from red cells. ADMA inhibits NO synthase, which may
Fenton reaction, activation of the xanthine–xanthine oxidase further limit NO production in SCD. The consequences of
pathway through recurrent ischemia–reperfusion injury, and reduced NO bioavailability include vasoconstriction, acti-
the depletion of reduced glutathione and enzymatic antiox- vation of endothelial cells with production of endothelin-1
idants, including superoxide dismutase and peroxiredoxins. (a potent vasoconstrictor and inflammatory mediator), and
Hemoglobinopathies due to structural mutations 183

upregulation of adhesion molecules, platelet activation, tis- a decrease in hospitalization and transfusion requirements.
sue factor expression, and thrombin generation, all of which Predictors of response to hydroxycarbamide include high
may contribute to the complex pathophysiology of sickle white cell count and absence of the Bantu haplotype. The
cell anemia. This model is supported by identification of a mechanism by which hydroxycarbamide, a ribonucleotide
clinical subphenotype of sickle cell anemia, associated with reductase inhibitor, acts involves the recruitment of erythroid
impaired NO bioavailability, characterized by more severe progenitors programmed to produce HbF. This appears to
hemolysis and increased risk of vascular complications, be mediated through NO-dependent activation of soluble
including ischemic stroke, pulmonary hypertension, leg guanylyl cyclase. Although predicated on its action as an
ulcers, priapism, and possibly nephropathy. inducer of HbF, it is apparent that hydroxycarbamide exerts
other effects of potential benefit in SCD. These include
reduced expression of adhesion molecules, enhanced NO
Treatment
production and red cell hydration, and reduction in the
Despite the in-depth understanding of the molecular basis leukocyte, platelet, and reticulocyte counts.
of sickle cell anemia, prefigured by Pauling’s landmark study Several targeted drug therapies for SCD have emerged
almost 70 years ago, improvement in clinical outcome has recently. Agents that block red cell and leukocyte adhe-
been slow and remains elusive in low-income settings, where sion are attractive candidates. Crizanlizumab, the first
childhood mortality is high. Progress in the development drug designed to target a specific cell adhesion molecule
of disease-modifying treatments has been disappointing and implicated in vaso-occlusion, is a humanized monoclonal
lagged behind advances in pathophysiology. The concept that antibody against P-selectin which blocks interaction with
adhesive interactions between circulating sickle red cells, leu- its cognate ligand, P-selectin glycoprotein ligand 1. The
cocytes, and endothelial cells play a central role in the patho- rationale for its use builds on extensive pre-clinical studies
physiology of SCD first evolved over 30 years ago. Despite (see earlier). The SUSTAIN study, a phase 2, multicenter,
this, to date no therapy that targets cell adhesion in SCD has placebo-controlled, double-blind trial, assessed the safety
been licensed. and efficacy of this novel therapy, with or without hydrox-
In high- and middle-income countries, the introduction ycarbamide, in patients with SCD. Over the course of the
of newborn screening combined with preventive measures 12-month trial, crizanlizumab significantly reduced the rate
that include penicillin prophylaxis, vaccination, and par- of pain crises compared to placebo. The results of a phase
ent/carer education have proved highly effective in reducing 3 clinical trial of rivipansel (GMI-1070), a pan-selectin
childhood mortality. The judicious use of red blood cell inhibitor that in earlier studies reduced the duration of pain
transfusion in devastating complications such as acute crises, length of hospital stay, and opioid requirement, are
chest syndrome, stroke, splenic sequestration, and aplastic awaited. In 2017 the FDA approved oral L-glutamine for use
crisis has also dramatically improved outcomes. With these in SCD based on the results of a phase 3 clinical trial which
measures, mortality rates have improved to 0.5 per 100 000 over 48 weeks showed a significant reduction in crisis and
affected children. The picture in adults is less favorable, with hospitalization rate in the intervention arm. The proposed
average life expectancy still well below that of the general mechanism of action of L-glutamine protects red cells against
population. Pivotal randomized controlled trials conducted oxidative damage through promoting NADH generation.
since the 1990s provide an evidence base for the use of Another promising avenue for drug development is inhibi-
transfusion for primary and secondary prevention of stroke tion of HbS polymerization, the initiating pathophysiological
and a rationale for the adoption of chronic transfusion pro- event. Previous efforts toward this goal have been thwarted
grams to reduce the frequency of vaso-occlusive crises and by unfavorable pharmacological properties and off-target
acute chest syndrome. In the acute setting, current therapies effects. Aromatic aldehydes such as 5-hydroxymethylfurfural
are supportive, with emphasis on rapid assessment of the (5-HMF) and its derivative GBT440 are allosteric effectors of
intensity, quality, and distribution of pain and the presence HbS which increase its oxygen affinity, maintain the R state,
of comorbidities, coupled with prompt administration of and consequently inhibit polymerization. GBT440, which is
opioid analgesia, antibiotic therapy where indicated, and active orally, binds to the N-terminal valine of the α-chain
hydration. Individually tailored opioid analgesia regimens of hemoglobin, forming a reversible Schiff base. Based on
with the use of adjunctive non-steroidal anti-inflammatory pre-clinical and early-phase clinical trial data, GBT440 has
drugs are favored by most centers. received Breakthrough Therapy and Priority Medicines des-
Hydroxycarbamide (hydroxyurea) was approved by the ignation from the FDA and European Medicines Agency,
Food and Drug Administration (FDA) in 1998, and is the respectively. A phase 3, double-blind, randomized, placebo-
first licensed drug that has been shown to modify the natu- controlled, multicenter study of GBT440, the HOPE study, is
ral history of SCD. Hydroxycarbamide therapy reduces the in progress.
incidence of painful crises and acute chest syndrome in The options for curative therapy in SCD are expanding.
adult and pediatric sickle cell anemia patients, and leads to Allogeneic bone marrow transplantation (BMT) for SCD
184 Molecular Hematology

has been performed since the 1980s, though access is insertions/deletions seems a more realistic prospect for the
limited, even in developed countries. Current consensus treatment of SCD and other β hemoglobinopathies.
guidelines recommend this option for young patients, The manipulation by gene editing of factors which silence
ideally pre-school age, with symptomatically severe SCD γ-globin expression offers an attractive approach. BCL11A
and a human leukocyte antigen (HLA)-matched sibling is a potent repressor of fetal hemoglobin, but it also has key
donor. A recent international survey of the results of 1000 non-erythroid functions in hematopoietic stem cells, neu-
HLA-identical sibling hematopoietic stem cell transplants ron development, B-cell lymphopoiesis, and dendritic cells.
showed the five-year event-free and overall survival, using A solution to the problem of selectivity might be to tar-
predominantly conventional myeloablative conditioning, get only one of the two BCL11A alleles, as in the mouse
were 91.4% and 92.9%, respectively. Transplant-related model BCL11A repression of γ-globin is exquisitely sensi-
mortality was 7% and the risk of graft rejection 2.3%. Both tive to gene dosage. SNPs affecting BCL11A which result
increase with age. Overall survival was higher in patients in a modest decrease in gene expression, preserving around
younger than 16 years (95%) than those aged 16 years or 65% of normal activity, are associated with an approxi-
older (81%). Reduced intensity conditioning has produced mately threefold increase in fetal hemoglobin in patients
encouraging results and holds promise as a safer option with sickle cell disease. Individuals who are haploinsuffi-
in older patients. Only a minority of eligible patients have cient for BCL11A have an elevated fetal hemoglobin level.
an HLA-matched related donor. This has prompted con- An elegant alternative is disruption by genome editing of an
sideration of alternative stem cell sources in the form of erythroid-specific enhancer for BCL11A to achieve selective
unrelated umbilical cord blood or marrow and haploiden- reduction of expression in erythroid precursors. Strategies
tical donors, which opens the possibility of cure to a wider which take advantage of the competitive expression of globin
group, though at the cost of increased graft rejection and genes at the β locus to replace HbA with a non-sickling
mortality. form such as HbF hold particular promise in the treatment
The long road to gene therapy for SCD reached a turning of SCD.
point in 2017 with the report by Ribeil et al. of the success-
ful treatment of a 13-year-old boy with a severe sickle cell
phenotype, on a prophylactic red cell transfusion program, Sickle/β thalassemia
using a lentiglobin vector carrying a β-globin gene modified
to enhance the anti-sickling effect (LentiGlobin BB305 vec- This syndrome is observed in geographical regions in which
tor βA-T87Q ). At 15 months, the patient was asymptomatic both HbS and β thalassemia are frequent, such as Africa,
and transfusion independent, with sustained production of Sicily, Greece, Turkey, the Arab countries, and regions of
the anti-sickling β-globin and correction of disease mark- the Americas with African and southern Mediterranean
ers. No safety concerns were reported. Additional results admixture.
will follow from the HGB-205 and HGB-206 clinical trials There are two phenotypic forms: S/β+ thalassemia, in
of LentiGlobin BB305 in SCD. Several additional gene ther- which the red cells contain between 20 and 40% HbA, the
apy trials using alternative gene cassettes are ongoing. In the remainder comprising HbS, HbF, and HbA2 ; and S/β0 tha-
long term gene therapy holds considerable promise for the lassemia, in which the red cells contain only HbS, HbF,
treatment of SCD, though to confirm the durability of clini- and HbA2 . The latter may sometimes be difficult to dis-
cal benefit and safety, greater patient numbers and extended tinguish from the homozygous state for the βS gene and
follow-up are needed. this may require pedigree or genotypic analysis. The rel-
Since the discovery of the clustered regularly interspersed ative frequency of S/β0 and S/β+ thalassemia reflects the
short palindromic repeats (CRISPR)/Cas9 complex, gene prevalence of individual β thalassemia mutations, which
editing has emerged as a potential tool for the treatment varies from one geographical region to another. In peo-
of SCD. The complex consists of a Cas9 endonuclease ple of African descent S/β+ thalassemia predominates,
and a sequence-specific single-guide RNA, which directs whereas in the Mediterranean S/β0 thalassemia is more
the endonuclease to the target DNA sequence. Following common.
cleavage by the Cas9 endonuclease, one of two mechanisms Though similar to that of sickle cell anemia, the clin-
may be employed to repair the double-strand break: non- ical spectrum of S/β thalassemia is broader and in S/β+
homologous end joining (NHEJ) or homology-directed thalassemia significantly milder. However, certain complica-
recombination (HDR). HDR enables genetic correction, tions, including proliferative retinopathy, osteonecrosis, and
but currently this is difficult to achieve at therapeutic hypersplenism, are more common.
efficiency in quiescent hematopoietic stem cells in which The hemoglobin level is higher and mean cell volume
NHEJ is the dominant repair pathway. At present, genetic (MCV) as well as mean corpuscular hemoglobin (MCH) typ-
disruption which relies on NHEJ to produce targeted ically lower than in sickle cell anemia.
Hemoglobinopathies due to structural mutations 185

dehydration. The kinetics of K+ /Cl− cotransport are altered


Hemoglobin C disease in HbC disease red cells, with delayed switch-off when the
stimulus for activity (cell volume increase or low pH) is
Worldwide, hemoglobin C is the third most common struc-
removed. Whether this alteration is sufficient to explain
tural hemoglobin variant (after HbS and HbE). Individuals
the pathophysiology of HbC disease and its relation to
who are homozygous for HbC display a mild hemolytic ane-
interaction of HbC with the red cell membrane remains to
mia that is generally asymptomatic. Splenomegaly is com-
be determined.
mon, but usually clinically benign. The anemia is mild to
Epidemiological data from Burkina Faso reveals that indi-
moderate in degree and accompanied by a reduced MCV. The
viduals homozygous for HbC are particularly resistant to
peripheral blood film is fairly characteristic and, with some
death from malaria. This is congruent with previous obser-
experience, diagnostic. The red cells appear hypochromic
vations that the red cells of HbC homozygotes are less able
due to their flatness (rather than low MCH or MCHC; in fact,
to release merozoites than normal or heterozygote (HbAC)
the red cells have a normal MCH and an increased MCHC).
red cells. By virtue of this greater protective effect, HbC may
Folded and target cells are prominent. Red cells may, though
eventually supersede the HbS gene with respect to preva-
infrequently in the absence of splenectomy, show intracellu-
lence in those populations where the two mutants coexist.
lar tetragonal crystals of HbC, best detected in reticulocyte
The multiplication rate of P. falciparum is lower in the red
preparations, where they retain their red color.
cells of HbC homozygotes than in normal red cells, proba-
The βC gene (β6 Glu → Lys) has a frequency one-quarter
bly due to disintegration of ring forms and trophozoites. It
that of the βS gene among African Americans. The βC
appears that only a subset of red cells of HbC homozygotes
mutation most likely originated in Burkina Faso, where
supports normal parasite replication.
the highest gene frequency (up to 0.5) is found, decreasing
The structural basis for HbC crystallization has been elu-
concentrically, and encompassing Mali, Ivory Coast, and
cidated. HbC crystals form by nucleation and grow by the
Ghana, all of which lie east of the Niger river.
attachment of individual HbC molecules from the solution.
The pathophysiology of HbC disease is dominated by
HbC crystallization is possible because of the huge entropy
the reduced solubility of hemoglobin, which results from
gain, likely stemming from the release of up to 10 water
replacement of β6 glutamic acid by lysine and its effect on
molecules per protein intermolecular contact–hydrophobic
membrane transport. The former is manifest as a tendency
interaction. The propensity of oxy HbC to form crystals is
for oxy HbC to form tetragonal crystals (Figure 14.8).
attributable to increased hydrophobicity resulting from the
Although less deformable, the red cells in HbC disease do
conformational changes that accompany the β6 Lys muta-
not produce vaso-occlusion, even when present in signifi-
tion. The oxy ligand state is thermodynamically driven to a
cant numbers after splenectomy. This may be explained by
limited number of aggregation pathways, with a high propen-
dissolution of oxy HbC crystals under conditions of low Po2
sity to form the tetragonal crystal structure. This contrasts
in the microvascular circulation. The red cells are uniformly
with deoxy HbC, which energetically favors equally multiple
denser. This characteristic is associated with stimulation
pathways of aggregation, not all of which will culminate in
of K+ /Cl− cotransport (higher than in SS cells, despite a
crystal formation, and which has a different crystal form.
lower reticulocyte count), a transport system which, through
efflux of K+ and accompanying water loss, leads to red cell
Hemoglobin SC disease

The compound heterozygous disorder hemoglobin SC dis-


ease combines two structural variants, HbS and HbC, neither
of which is associated with a clinical phenotype in the simple
carrier state. The clinical basis of Hb SC disease, a significant
sickling disorder, therefore demands an explanation. The
pathophysiology of Hb SC disease derives from two mech-
anisms. The relative proportion of HbS to HbC is 50 : 50, in
contrast to sickle cell trait in which the ratio of HbS to HbA
is 40 : 60. This reflects the variable electrostatic affinity of
structurally distinct β chains for α globin, which governs the
proportion of different αβ subunits and hemoglobin assem-
bly within the red cell. The red cells of Hb SC individuals
have a higher MCHC, reflecting dehydration due to increased
Fig. 14.8 Hb C tetragonal crystals. K+ /Cl− cotransport. Since polymerization is dependent on
186 Molecular Hematology

Density MCHC
g/ml g/dl
AA 1.065 26.0
Reticulocytes
Fig. 14.9 Percoll density gradients of AA, CC,
1.085 34.0 and SC red cells. This shows the separation of red
cells by density. Light-density cells are on top,
high-density cells on the bottom. Note that SS
1.095 38.0
blood has normal-density red cells as the largest
1.107 40.5 compartment, then some very dense cells, and
Dense SC cells 1.125 49.0 some intermediate-density cells between the two.
ISC 1.143 55.0
In contrast, all the cells of the CC patients are
denser than normal red cells. The SC red cells are
AA AC SC SC CC SS Beads intermediate.

intracellular HbS concentration, both these factors favor Glu → Gln) interact with HbS to produce a severe clinical
polymer formation. In keeping with this, in vitro studies have phenotype akin to sickle cell anemia. This is explained by the
shown that increasing the proportion of HbS from 40% to fact that these substitutions at the β121 residue, a contact
50% increased the rate of polymerization almost 15-fold. Hb point in the HbS polymer, increase the nucleation rate for
SC red cells are denser than normal or the majority of sickle polymer formation. Only a small number of cases of Hb SE
cell anemia red cells (Figure 14.9). Interestingly, HbS favors have been described, all of which displayed a mild pheno-
HbC crystallization; this explains, in addition to the loss of type. Other compound heterozygous sickling disorders due
splenic function, why crystals are generally more prominent to a second structural variant are rare. Co-inheritance of Hb
in Hb SC than HbC disease. However, for the reasons already Quebec-Chori (β87 Thr → Ile) and HbS is an interesting
given, this does not contribute to vaso-occlusion. example. Since Hb Quebec-Chori is electrophoretically
Hb SC disease is milder than sickle cell anemia. Virtually silent, the compound heterozygous state with HbS mimics
all patients survive to adulthood and average life expectancy sickle cell trait; however, its presence can be distinguished
is close to that of matched controls. The clinical spectrum by mass spectrometry. The proposed mechanism for the
resembles that of sickle cell anemia, but the frequency and interaction between the two variant globin chains is that
severity of most specific complications are generally less. substitution of a bulky hydrophobic isoleucine residue at β87
Two exceptions show a disproportionate prevalence or sever- creates a more favorable environment in the acceptor pocket
ity in Hb SC disease – osteonecrosis and proliferative sickle for β6 Val, which stabilizes intermolecular contacts in the
retinopathy the frequency of which is similar and higher polymer.
respectively to that in sickle cell anemia. Splenomegaly is Several dominant sickle mutations in which there is a sec-
more frequent and functional hyposplenism delayed. Mean ond substitution in addition to β6 Val in the same β chain
red cell survival is around 27 days compared with 17 days in have been described. This results in assembly of a “super-
sickle cell anemia. Hematologically, Hb SC disease is char- Hb S,” which manifests with symptoms in the heterozygote.
acterized by a higher hemoglobin, lower MCV, lower retic- Several examples have been well characterized. Hb S-Antilles
ulocyte count, fewer dense red cells, and lower HbF level (β6 Glu → Val; 23 Val → Ile) is expressed in the heterozygote
compared with sickle cell anemia. The blood film in Hb at a level of around 40% and produces a syndrome resem-
SC disease may be diagnostic. In addition to the red cell bling a mild sickle cell anemia phenotype. The mechanism
forms seen in HbC disease, there are frequent dense cells, is complex: the second mutation reduces the solubility from
some of which resemble a broad-beamed canoe and occa- a Csat of 18 g dl−1 for Hb S to 12 g dl−1 , with the result that
sionally ISCs. Intraerythrocytic crystals are best visualized polymerization is promoted. Hb S-Antilles also has reduced
with supravital stains (Figure 14.10) or after incubation of oxygen affinity. This oxygen equilibrium shift favors sickling.
blood with 3% sodium chloride (NaCl). A similar effect is seen with Hb Jamaica Plain (β6 Glu → Val;
68 Leu → Phe), in which the amino acid substitution at β68
reduces oxygen affinity, producing symptoms of sickling in
Other sickle hemoglobinopathies the heterozygous state. The other well-documented dom-
inant sickle mutation is Hb S-Oman (β6 Glu → Val; 121
Many compound heterozygous states have been described in Glu → Lys). This generates an even more powerful “super-
which HbS is co-inherited with a second β globin variant, Hb S.” Even at expression levels as low as 20%, resulting
most of which are of no clinical significance. Exceptions from concomitant inheritance of the α thalassemia geno-
are Hb SO-Arab, Hb SD-Punjab (Los Angeles), and Hb type −α/−α, this is associated with a phenotype similar to
SE. Hb O-Arab (β121 Glu → Lys) and Hb D-Punjab (β121 Hb S-Antilles. Since the two dominant forms of Hb S have
Hemoglobinopathies due to structural mutations 187

Fig. 14.10 SC red cells from a double-stained


smear. This blood smear is from a patient with Hb
SC and is stained with Wright’s and reticulocyte
stain. Note the fl at thin cells (which generate
target cells), folded cells, and those with
intracellular tetragonal crystals (arrows).

similar solubility in the deoxy state, the more severe pheno- chain variants, HbS and HbC, this proportion is reduced in
type of Hb S-Oman implies that the β121 mutation (identical the presence of α thalassemia. Homozygotes exhibit a benign
to that in Hb O-Arab) possesses intrinsic pathophysiologi- thalassemic phenotype because the mutation responsible
cal properties. This hypothesis is supported by the hemol- produces both a structurally altered β chain and reduced
ysis and perturbance of red cell cation transport evident in expression due to the generation of an alternative splice site.
individuals homozygous for Hb O-Arab. Rare symptomatic The clinical syndrome of homozygous HbE disease is very
HbS heterozygotes in whom deficiency of pyruvate kinase mild, with no or minimal anemia (once nutritional causes
has been identified have been described. The mechanism in of anemia are treated) and low MCV, with target cells in the
these cases is attributed to the increased 2,3-DPG content of smear and hypochromia. Density gradient separation reveals
red cells, with a block in glycolysis distal to the Rapoport– that although the red cells are small they have a normal
Luebering shuttle that shifts the hemoglobin oxygen equilib- MCHC, as a consequence of a diminished MCH, due to the
rium in favor of deoxy Hb S. thalassemic component of this disease. People who inherit
HbE and β thalassemia trait present with the features of
thalassemia major or intermedia (see Chapter 1).
Homozygous HbE and Hb E/β The protective effect of HbE against P. falciparum malaria
thalassemia has been assessed in a mixed erythrocyte invasion assay. The
parasite preferentially invaded normal (AA) red cells com-
Hemoglobin E (β26 Glu → Lys), the most common struc- pared with AE, EE, or E/β thalassemia red cells. Heterozy-
tural hemoglobin variant worldwide, is prevalent across a gote (AE) red cells were the poorest target, showing inva-
wide geographical area, from the eastern provinces of India to sion restricted to approximately 25% of the red cells. It has
the Philippines. Populations living near the common border been postulated that AE red cells might have an uniden-
of Cambodia, Laos, and Thailand (the Khmer people) have tified membrane abnormality that renders the majority of
the highest incidence (gene frequency reaching 0.5) of HbE, the red cell population relatively resistant to invasion by P.
the selection pressure for which is resistance to infection by falciparum, consequently reducing parasitemia and hence
P. falciparum malaria. the lethality of infection. This is consistent with the Hal-
In heterozygotes HbE constitutes 25–30% of the dane hypothesis of heterozygote protection against severe
hemoglobin. In common with the other widespread β malaria.
188 Molecular Hematology

(e.g. Hb Hammersmith). Hb Zurich (β63 His → Arg) is


Unstable hemoglobins another interesting variant that affects multiple hemoglobin
functions. The structural change in Hb Zurich alters the
Cases of non-spherocytic hemolytic anemia due to a struc-
heme pocket, resulting in reduced oxygen affinity but little
turally unstable hemoglobin were first recognized because
effect on carbon monoxide (CO) binding. The higher levels
precipitation occurred on thermal incubation of hemolysate,
of carboxyhemoglobin (CO-Hb) found in patients with Hb
a phenomenon which forms the basis of the heat stability test.
Zurich stabilize hemoglobin, with the irony that smoking
In many patients Heinz bodies were present. There are over
affords protection against hemolysis.
100 mutations that render the hemoglobin molecule unsta-
In most cases treatment revolves around supportive mea-
ble, of which only a minority are associated with significant
sures that include folic acid supplementation, prompt treat-
clinical symptoms. About 75% are β chain variants. Unstable
ment of infection and fever, and avoidance of oxidant drugs.
hemoglobins are generally inherited in an autosomal dom-
In severe cases splenectomy has been performed and is gen-
inant manner, although rare homozygous cases, for exam-
erally, though not universally, beneficial. Hydroxycarbamide
ple Hb Bushwick (β74 Gly → Val) and Hb Taybe (α1 38/39
has been used to stimulate Hb F in unstable β chain variants.
Thr deletion), have been described. In a significant number
Some unstable hemoglobin variants result in low hemoglobin
of cases there is no antecedent family history, indicating that
oxygen saturation measurements by pulse oximetry due to
the mutation has arisen spontaneously. The unstable variant
their altered absorbance spectra.
usually accounts for 10–30% of the total hemoglobin.
Structural alterations may render hemoglobin unstable by
five mechanisms:
High oxygen affinity hemoglobins
r destabilization of the attachment of heme to globin;
r interference with stability of the α β contact area, which
1 1 The sigmoid curve of the oxygen binding of hemoglobin
does not normally dissociate; reflects the transition from the low-affinity deoxy (T) to
r introduction of either a bulky or charged side chain to the
the oxy (R) state. With this molecular switch, hemoglobin
hydrophobic interior of the hemoglobin molecule; acquires a high affinity for oxygen. Mutations that stabilize
r disruption of the secondary structure of the molecule by
the R state or destabilize the T state produce hemoglobins
a side chain that is not α-helix-friendly (commonly as a with increased oxygen affinity. In response, the oxygen-
result of proline substitution); sensing mechanism in the kidney triggers an increased
r elongation of a globin subunit.
output of erythropoietin, which in turn stimulates red cell
Hb Köln (β98 Val → Met) is the most common of the production. This results in erythrocytosis and an elevated
unstable hemoglobins and shows no ethnic predilection. hematocrit.
Many unstable hemoglobin variants do not alter the sur- There are approximately 100 high-affinity hemoglobin
face charge of the hemoglobin molecule and therefore have variants, most of which are rare. Almost all show a domi-
an electrophoretic mobility identical to that of HbA. Light nant mode of inheritance. Variants associated with a marked
bands representing dissociated globin subunits, in some cases increase in hemoglobin oxygen affinity are likely to be lethal
devoid of heme, may be observed. in homozygotes, and the few examples of homozygous high-
Clinically, unstable hemoglobins are characterized by affinity hemoglobins reported are confined to less severe phe-
hemolytic anemia, which ranges from mild to severe depend- notypes. The first high-affinity variant to be described was
ing on the causative mutation, and the presence of pigmen- Hb Chesapeake (α2 98 Arg → Leu), which has a P50 on whole
turia (due mostly to dipyrroles). Unstable hemoglobins due blood of 19 mmHg (normal 26 mmHg) and exhibits reduced
to γ chain variants, such as Hb Poole (G γ130 Trp → Gly), are cooperativity. This mutation results in substitution at a con-
a rare cause of transient neonatal hemolysis. The mechanism served residue of the α1 β2 interface between dimer subunits
of red cell destruction, much of which occurs in the bone that is important for stability of the T conformer. The substi-
marrow, involves displacement of heme, which destabilizes tution of leucine stabilizes the R state.
the hemoglobin tetramer and its composite globin subunits The molecular defects responsible for high-affinity
and leads to the formation of hemichrome. Globin subunits hemoglobins may be divided into those that occur at sites
aggregate and precipitate, leading to reduced deformability directly involved in the R → T transition (the α1 β2 contact
and removal or entrapment of red cells, which contain Heinz area or the switch region of the molecule, particularly the
bodies. Febrile episodes and ingestion of oxidant drugs may C and N terminals), such as Hb Chesapeake; those which
accelerate hemolysis. Hemoglobin level may be modulated disrupt the 2,3-DPG binding site in the central cavity, such
by altered oxygen affinity, being near normal in those as Hb Old Dominion (β143 Val → Met); mutations at the
unstable variants with increased oxygen affinity (e.g. Hb α1 β1 interface that produce a structural change favoring the
Köln) and much lower in those with reduced oxygen affinity R state, such as Hb San Diego (β109 His → Tyr); and those
Hemoglobinopathies due to structural mutations 189

which constrain quaternary conformational change through of overt cardiopulmonary causes and anemia are evident, the
polymerization, such as Hb Porto Alegro (β9 Ser → Cys). diagnosis of a low-affinity hemoglobin variant should be con-
The clinical course of high-affinity hemoglobin variants is sidered, partly to avoid unnecessary investigation. However,
benign. The reduction in tissue oxygen delivery is compen- neither cyanosis nor anemia is universal. In patients in whom
sated for by an increased red cell mass and treatment is not the P50 is markedly increased, anemia is not usually present.
generally required. Maternal high-affinity hemoglobin vari- Hemoglobin separation techniques are helpful only if the
ants do not compromise fetal well-being, arguing against a amino acid substitution alters the charge of the molecule and
physiologically important role for the greater affinity for oxy- measurement of the oxygen dissociation curve (P50 ) is the
gen of Hb F than Hb A in normal pregnancy. Curiously, the gold standard. The oxygen delivery properties of high- and
degree of erythrocytosis may differ among individuals who low-affinity hemoglobins are illustrated in Figure 14.11.
inherit high-affinity variants with similar P50 values, suggest-
ing other factors influence the adaptation to hypoxia.
M hemoglobins

Low oxygen affinity hemoglobins M hemoglobins result from mutations which stabilize heme
iron in the oxidized ferric (Fe3+ ) state. Inheritance is auto-
Hemoglobins with low oxygen affinity deliver oxygen to somal dominant. Nine M hemoglobin variants have been
the tissues more efficiently and are associated with ane- described, most of which are the product of substitution, in
mia due to reduced erythropoietic drive. Relatively few all but one case by tyrosine, of the proximal or distal histidine
hemoglobin variants with low oxygen affinity have been interacting with the heme group. All are rare, with the excep-
described. Cyanosis will be evident if the deoxyhemoglobin tion of Hb Iwate (α2 87 His → Tyr), originally described in
level exceeds 5 g dl−1 . This may be present at birth in low- the Iwate prefecture of Japan. The interactions between the
affinity α chain variants, but its onset is delayed in low- tyrosine-substituted histidine residues and heme iron have
affinity β chain variants because of the latency in β globin been resolved for some M hemoglobins by X-ray crystallog-
expression. raphy and nuclear magnetic resonance (NMR) spectroscopy,
The first low-affinity variant to be described was Hb and suggest a model whereby the side group of tyrosine
Kansas (β102 Asn → Thr). Two other substitutions at β102 acts as an internal ligand preventing interaction with the
are associated with low affinity, Hb Beth Israel (β102 residual histidine residue. One other molecular defect, Hb
Asn → Ser) and Hb St Mande (β102 Asn → Tyr). This is M-Milwaukee 1 (β67 Val → Glu), has been described in
explained by the fact that β102 Asn, a highly conserved which the carboxyl group of the substituted glutamic acid
residue, is required to stabilize the hemoglobin molecule at residue binds and stabilizes oxidized heme iron. Predictably,
the α1 β2 interface in the R state. If cyanosis in the absence some M hemoglobins have reduced oxygen affinity.

100 20
High
in
ob Normal
gl
yo

80
M

y
nit

15
ffi
Oxygen saturation (%)

ha

O2 content (vol %)
Hig

al
rm

60
No

10
nity Low
Fig. 14.11 Oxygen-binding curves of 40 affi
Low
hemoglobins with high and low oxygen
affinity. Notice that the extraction of oxygen by
the tissues, which is the difference between 5
20
pulmonary oxygen pressure (100 mmHg) and
capillary oxygen pressure (40 mmHg), is lower than
normal in high-affinity hemoglobins (low P50 ) and
higher than normal in low-affinity hemoglobins.
0 20 40 60 80 100
This is why the former have erythrocytosis and the
latter (most of the time) anemia. Oxygen tension (mmHg)
190 Molecular Hematology

Hemoglobin A Hemoglobin M Boston

G He G He
lix lix

E Helix 87 Tyr
87 His E Helix

58 His 58 Try
F Helix F Helix

Hemoglobin A Hemoglobin M Milwaukee

lix lix
G He G He
Fig. 14.12 Structure of M hemoglobins. Heme
environment of two M hemoglobins compared
67 Val with HbA. Note the diverse changes in the distal
67 Glu
92 His 92 His and proximal tyrosines (which have replaced the
normal histidines) in each of the M hemoglobins.
E Helix E Helix These hemoglobins have a characteristic visible
F Helix F Helix
spectrum around 610 nm.

M hemoglobins resulting from mutation of the α, β, r HbC in the homozygous state is associated with mild
or G γ genes have been described. The latter were iden- chronic hemolysis, but when co-inherited with HbS pro-
tified in newborns with “cyanosis” that resolved sponta- duces a syndrome similar to but generally milder than
neously in the first few months of life. Affected individuals sickle cell anemia.
appear slate gray in color due to pseudocyanosis (there may r homozygous HbE results in hemolysis and a mild micro-
in addition be an element of true cyanosis in low-affinity cytic anemia, but in combination with β thalassemia pro-
M hemoglobin variants), but are otherwise asymptomatic duces a phenotype of remarkable variability, ranging from
and have a normal life expectancy. Pulse oximetry mea- mild thalassemia intermedia to transfusion-dependent
surements are unreliable. Although standard hemoglobin thalassemia major.
separation techniques may be helpful, definitive diagno- r unstable hemoglobins lead to a hemolytic anemia of vari-
sis requires examination of the visible absorbance spec- able severity.
trum between 450 and 650 nm of a hemolysate, which r low oxygen affinity variants and M hemoglobins are char-
reveals individual spectral patterns that differ from that acteristically associated with cyanosis or pseudocyanosis,
of methemoglobin (Figure 14.12). Differential diagnosis though if unstable may also be associated with hemolysis
includes enzymopathic or acquired methemoglobinemia, of mild degree.
sulfhemoglobinemia, and unstable hemoglobin variants such r high oxygen affinity mutant hemoglobins present with ery-
as Hb Chile (β28 Leu → Met) associated with accelerated throcytosis.
methemoglobin formation. The main value in identification This panoply of clinical syndromes can be attributed to
of M hemoglobins lies in the avoidance of unnecessary med- the following alterations of the hemoglobin molecule: (i)
ical intervention. modification of the biophysical properties of the hemoglobin
molecule (e.g. polymerization of deoxy HbS); (ii) altered
ligand binding affinity; (iii) changes in the heme environ-
Conclusions ment (e.g. M hemoglobins); (iv) instability of the functional
tetramer; and (v) reduced globin synthesis (e.g. HbE). Over
Structurally abnormal hemoglobins may present as one of 1000 genetic variants of hemoglobin have been described.
several well-defined clinical syndromes: These are predominantly due to missense mutation and most
are rare and clinically silent. Only HbS, HbC, and HbE have
r homozygous or compound heterozygous inheritance of reached a high frequency in human populations as a result
HbS produces a picture of chronic anemia and cumulative of the selective pressure of malaria. These together with less
organ damage, punctuated by intermittent painful crises common variants have provided a wealth of insight into the
and other acute manifestations. structure and function of human hemoglobin.
Hemoglobinopathies due to structural mutations 191

Nagel, R.L., Johnson, J., Bookchin, R.M. et al. (1980). Beta-chain contact
Further reading sites in the haemoglobin S polymer. Nature 283: 832–834.
Porteus, M. (2016). Genome editing: a new approach to human thera-
General peutics. Annu. Rev. Pharmacol. Toxicol. 56: 163–190.
Prengler, M., Pavlakis, S.G., Prohovnik, I. et al. (2002). Sickle cell
Eaton, W.A. and Hofrichter, J. (1990). Sickle cell hemoglobin polymer-
disease: the neurological complications. Ann. Neurol. 51: 543–
ization. Adv. Protein Chem. 40: 63–279.
552.
Henry, E.R., Bettati, S., Hofrichter, J. et al. (2002). A tertiary two-state
Ribeil, J.A., Hacein-Bey-Abina, S., Payen, E. et al. (2017). Gene ther-
allosteric model of hemoglobin. Biophys. Chem. 98: 198–164.
apy in a patient with sickle cell disease. N. Engl. J. Med. 376: 848–
Nagel, R.L. (2002). Disorders of hemoglobin function and stability. In:
855.
Blood: Principles and Practices of Hematology (eds. R.I. Handon, S.E.
Serjeant, G.R. and Serjeant, B.E. (2001). Sickle Cell Disease. Oxford:
Lux and T.P. Stossel). Philadelphia, PA: J.B. Lippincott.
Oxford University Press.
Steinberg, M., Forget, B., Higgs, D. et al. (eds.) (2009). Disorders of
Ware, R.E., de Montalambert, M., Tshilolo, L. et al. (2017). Sickle cell
Haemoglobin: Genetics, Pathophysiology and Clinical Management.
disease. Lancet 390: 311–323.
Cambridge: Cambridge University Press.
Thom, C.S., Dickson, C.F., Gell, D.A. et al. (2013). Hemoglobin variants:
biochemical properties and clinical correlates. Cold Spring Harb. Per- Hemoglobin C
spect. Med. 3: a011858.
Dewan, J.C., Feeling-Taylor, A., Puius, Y.A. et al. (2002). Structure of
mutant human carbonmonoxyhemoglobin C (betaE6K) at 2.0 Å res-
Sickle cell disease olution. Acta Crystallogr. D Biol. Crystallogr. 58: 2038–2042.
Fabry, M.E., Romero, J.R., Suzuka, S.M. et al. (2000). Hemoglobin C
Chaturvedi, S., Bhatnagar, P., Bean, C.J. et al. (2017). Genome-wide asso-
in transgenic mice: effect of HbC expression from founders to full
ciation study to identify variants associated with acute severe vaso-
mouse globin knockouts. Blood Cell Mol. Dis. 26: 331–347.
occlusive pain in sickle cell anemia. Blood 130: 686–688.
Fairhurst, R.M., Fujioka, H., Hayton, K. et al. (2003). Aberrant develop-
Desai, A.A., Lei, Z., Bahroos, N. et al. (2017). Association of circulating
ment of Plasmodium falciparum in hemoglobin CC red cells: implica-
transcriptomic profiles with mortality in sickle cell disease. Blood 129:
tions for the malaria protective effect of the homozygous state. Blood
3009–3016.
101: 3309–3315.
Dever, D.P., Bak, R.O., Reinisch, A. et al. (2016). CRISPR/Cas9 beta-
Modiano, D., Luoni, G., Sirima, B.S. et al. (2001). Haemoglobin C pro-
globin gene targeting in human haematopoietic stem cells. Nature
tects against clinical Plasmodium falciparum malaria. Nature 414:
539: 384–389.
305–308.
Gluckman, E., Cappelli, B., Bernaudin, F. et al. (2017). Sickle cell dis-
Nagel, R.L. and Lawrence, C. (1991). The distinct pathobiology of SC
ease: an international survey of results of HLA-identical sibling
disease: therapeutic implications. Hematol. Oncol. Clin. North Am. 5:
hematopoietic stem cell transplantation. Blood 129: 1548–1556.
433–451.
Halsey, C. and Roberts, I.A. (2003). The role of hydroxyurea in sickle
Vekilov, P.G., Feeling-Taylor, A.R., Petsev, D.N. et al. (2002). Inter-
cell disease. Br. J. Haematol. 120: 177–186.
molecular interactions, nucleation, and thermodynamics of crystal-
Herrick, J.B. (1910). Peculiar elongated and sickle-shaped red blood cor-
lization of hemoglobin C. Biophys. J. 83: 1147–1156.
puscles in a case of severe anemia. Arch. Intern. Med. 6: 517.
Kato, G.J., Steinberg, M.H., and Gladwin, M.T. (2017). Intravascular
hemolysis and the pathophysiology of sickle cell disease. J. Clin. Hemoglobin E
Invest. 127: 750–760.
Chotivanich, K., Udomsangpetch, R., Pattanapanyasat, K. et al. (2002).
Kaul, D.K., Fabry, M.E., and Nagel, R.L. (1996). The pathophysiology of
Hemoglobin E: a balanced polymorphism protective against high
vascular obstruction in the sickle syndromes. Blood Rev. 10: 29–44.
parasitemias and thus severe P falciparum malaria. Blood 100: 1172–
Lettre, G. (2012). The search for genetic modifiers of diseases sever-
1176.
ity in the β-hemoglobinopathies. Cold Spring Harb. Perspect. Med.
Fucharoen, S. and Weatherall, D.J. (2012). The hemoglobin E tha-
2: a015032.
lassemias. Cold Spring Harb. Perspect. Med. 2: a011734.
Lettre, G. and Bauer, D.E. (2016). Fetal haemoglobin in sickle-cell dis-
Schrier, S.L. (2002). Pathophysiology of thalassemia. Curr. Opin. Hema-
ease: from genetic epidemiology to new therapeutic strategies. Lancet
tol. 9: 1231–1226.
387: 2554–2564.
Chapter 15 Molecular pathogenesis of malaria
David J. Roberts1 , Arnab Pain2,3 & Chetan E. Chitnis4
1 National Health Service Blood and Transplant (Oxford), John Radcliffe Hospital, Oxford, UK
2
Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology, Jeddah, Saudi Arabia
3 Nuffield Division of Clinical Laboratory Sciences (NDCLS), The John Radcliffe Hospital, University of Oxford Headington, Oxford, UK
4
Malaria Group, Pasteur Institute, Paris, France

Introduction, 193 Merozoite surface protein 1 and invasion, 199


Epidemiology of malaria, 193 Adhesion of infected red blood cells to host cells, 200
Life cycle of malaria, 195 Malarial anemia, 202
Invasion of red blood cells, 196 Ineffective erythropoiesis in malaria, 204
Structural basis for the interaction of EBPs with host receptors, 198 Future prospects, 205
Reticulocyte-binding proteins, 199 Further reading, 206
Apical membrane antigen 1 and rhoptry proteins, 199

malaria. In this chapter we examine the basic features of


Introduction malaria the parasite and malaria the disease, the molecular
aspects of the invasion of red blood cells, and the adhesion of
Malaria is a major global public health problem and it
infected red blood cells to host cells and receptors and their
is estimated that it is responsible for over 400 000 deaths
role in pathogenesis. We also consider the pathology of ane-
annually and 160 million infections. The vast majority of
mia, which is a prominent feature of the disease, and outline
morbidity and mortality from malaria is caused by infection
drug treatment and vaccine development for malaria, as well
with Plasmodium falciparum, although Plasmodium vivax,
as genomic approaches to the study of the parasite, pathology,
Plasmodium ovale curtisi, Plasmodium ovale wallikeri and
and prevention of the disease.
Plasmodium malariae also cause infection and illness. In a
few areas, Plasmodium knowlesi, Plasmodium cynomolgi and
Plasmodium simium can cause zoonotic infection and illness. Epidemiology of malaria
The pathology of malaria is entirely related to the growth and
development of the parasite in red blood cells, and indeed a Approximately 1000 million people live in areas where
significant proportion of the mortality and morbidity is due malaria parasites are endemic. According to the World
to anemia. Malaria could therefore quite reasonably, if some- Health Organization (WHO), 161 million clinical cases of
what unconventionally, be considered the greatest single malaria were reported worldwide in 2016, giving rise to
cause of years of lost life due to a hematological disease. A 430 000 deaths per year; this represents a 45% decrease in
combination of insecticide-treated bed nets, highly effective mortality since 2000. These spectacular statistics are partly
artemisinin-based drug regimens, and better medical care due to the massive use of artemisinin-based combination
has greatly reduced deaths from malaria in the last decade. therapies (ACTs) as a first-line treatment (Figure 15.1).
The malarial parasite has a complex life cycle, alternating There is substantial evidence that the incidence of severe
between humans and the female Anopheles mosquito, a liver disease, particularly in East and West Africa, is falling, due
and blood stage of growth, complex eukaryotic metabolic to the use of artemisinin combination treatment, leading to
systems, and a panoply of mechanisms to evade protective the recent award of the Nobel Prize to Youyou Tu for the
host responses. The vast landscape of interactions between discovery of artemisinin, and impregnated bed nets as part
humans, parasites, and mosquitoes has been illuminated by of the Gates Foundation and WHO initiatives (available
genomics and genetic epidemiology. A short review can only at https://www.gatesfoundation.org/What-We-Do/Global-
highlight some of the many key molecular processes involved Health/Malaria and https://rollbackmalaria.com). Because
in the growth, pathology, and prevention or treatment of of these recent advances, the WHO announced support
of a more ambitious goal: progressive malaria elimination
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. in endemic countries, leading to its complete eradication.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. Although there is understandable and appropriate interest

193
194 Molecular Hematology

Fig. 15.1 Global malaria endemicity. Areas are colored according to malaria endemicity (prevalence): light blue, hypoendemic (areas in which
childhood infection prevalence is less than 10%); medium blue, mesoendemic (areas with infection prevalence between 11% and 50%); dark
blue, hyperendemic and holoendemic (areas with an infection prevalence of 50% or more). Gray areas are a combined mask of areas outside of
the transmission limits and areas of population density less than 1 person per km2 .
Source: Snow R.W., Guerra C.A., Noor A.M., Myint H.Y., Hay S.I. (2005). The global distribution of clinical episodes of Plasmodium falciparum
malaria. Nature 434: 214–217, with permission.

in the molecular aspects of the disease and new therapies, it The clinical presentation of malaria infection is also wide
remains true that in endemic areas the disease is associated and influenced by age, immune status, and pregnancy and
with poverty and poor access to established treatments or by the species, genotype, and perhaps the geographical ori-
simple preventive measures. gin of the parasite. In endemic areas, many infections present
Malaria occurs when the mean temperature is above 20 °C as an uncomplicated febrile illness. In more severe forms of
for P. falciparum and 15 °C for other human malarias, per- the disease, children may present with prostration or inabil-
mitting development of the mosquito stages of development ity to take oral fluids, or in younger children an inability to
(sporogony). Transmission rarely occurs above 1500 m, in suckle. Alternatively, they may exhibit several syndromes of
arid regions, or in the Central and South Pacific, where suit- severe disease, including anemia, coma, respiratory distress,
able vectors are absent. Moreover, P. vivax malaria is rare in and hypoglycemia, and may also have a high rate of bac-
Africa, where the population frequency of the blood group teremia, due to susceptibility to coincident bacterial infec-
Duffy negative (Fy(a)- Fy(b)-) is high. In malaria-endemic tion. In most age groups, anemia is frequently accompanied
areas, the pattern of clinical malaria is determined by the by more than one syndrome of severe disease, and the already
number and timing of infective mosquito bites. Mosquito substantial case fatality rate of 15–20% for severe malaria in
numbers increase after rainfall, leading to an increase in new African children rises significantly when multiple syndromes
infections; in naı̈ve individuals, parasites can cause chronic of severe disease are present.
infection lasting many months, either as a result of long blood The age distribution of anemia and other syndromes of
stage infection or repeated waves of blood stage parasitemia severe disease is a consistent, but puzzling feature, of the
from activation of dormant liver-stage hypnozoites in P. vivax epidemiology of clinical malaria. Children born in endemic
and P. ovale. areas are protected from severe malaria in the first six months
The intensity of transmission determines the distribution of life by the passive transfer of maternal immunoglobulins
of clinical symptoms in different age groups. In general, and by fetal hemoglobin. Beyond infancy, the most com-
in areas of high transmission younger children suffer from mon presentation of severe disease changes from anemia
severe disease; as transmission decreases, older children suf- in children aged one to three years, in areas of high trans-
fer from severe disease. When transmission is sporadic, the mission, to cerebral malaria in older children, in areas of
population can lose much of its natural immunity, and a sud- lower transmission. As transmission intensity declines fur-
den increase in mosquitoes can result in epidemics where ther, severe malaria is most frequently found in older age
large numbers fall ill in all age groups. groups.
Molecular pathogenesis of malaria 195

There is scant evidence to explain the relationships A E


between age, transmission, and predominant clinical syn-
drome of severe malaria. One possible explanation is the age-
dependent increase in expression of complement receptor 1
(CR1) on red blood cells, and so a parallel age-dependent
increase in the capacity to inactivate complement compo- Liver
nents absorbed or deposited directly onto the surface of the
red blood cell. Another explanation, by no means mutually
B D
exclusive, is that erythropoietin (EPO) has a powerful cyto-
protective effect on neural, cardiac, and other tissues and,
indeed, increased levels of EPO are found in younger chil-
dren with anemia and with malaria. A clinical study showed
that there was a significant protective effect of high levels of Asexual
Growth
endogenous EPO on survival after malaria-induced coma,
particularly in children under two years old.
In endemic areas, even today, it is believed that selection C
for malaria-protective alleles has taken place over the last Adhesion to endothelial
5000 years, after the widespread introduction of agriculture and other host cells
allowed significant increases in population density, transmis-
sion, and parasite virulence. Sickle cell trait and thalassemia
traits and allele for glucose-6-phosphate dehydrogenase
(G6PD) protect from infection and are truly polymorphic Key:
characteristics in many parts of the world. Understanding RBC Merozoites Ring Trophozoites/
the genetic epidemiology has provided classic examples of Schizonts
the principle of genetic selection in vivo, for example bal- Fig. 15.2 Life cycle of Plasmodium. (a) The asexual life cycle begins
ancing selection for sickle cell trait and negative epistasis for when sporozoites from a female mosquito taking a blood meal enter
sickle cell trait and α thalassemia. The homozygous forms the circulation and invade hepatocytes. (b) Up to 30 000 merozoites
of these disorders cause significant clinical disease, such as are formed. The hepatocyte becomes detached and migrates to the
sickle cell disease, β thalassemia, and G6PD deficiency. In liver sinusoid, where budding of parasite-filled vesicles or
“merosomes” occurs. Following rupture of the merosome, infective
endemic areas these genetic diseases represent major public
merozoites are released and invade erythrocytes (RBC). (c) Within
health problems.
RBCs, the parasite develops through the stages of rings, trophozoites,
and schizonts. Mature schizonts burst to release erythrocytic
merozoites, which invade new RBCs. (d) A small proportion of
merozoites in RBCs transform into male and female gametocytes,
Life cycle of malaria which are ingested by the mosquito. (e) The male and female gametes
fuse and transform into an oocyst, which divides asexually into many
In P. falciparum malaria, the infective sporozoite forms are sporozoites that migrate to the salivary gland, from where they are
inoculated from the salivary glands of a female Anophe- released during the next blood meal.
les mosquito into the subcutaneous tissues and sometimes
the bloodstream (Figure 15.2). These thin, needle-shaped
cells, 10–12 μm in length, circulate briefly with a half-life induces hepatocyte cell death, causing the release of mero-
of approximately 30 min, traversing sinusoidal Kupffer cells zoites in membrane-enclosed structures or “merosomes” that
and several hepatocytes before residing in a single hepato- are extruded from the infected cell, so avoiding host-cell
cyte, requiring sporozoite microneme protein essential for defense mechanisms.
cell traversal (PfSPECT) or perforin-like protein 1 (PfPLP1). The merozoites bind and then invade red blood cells
Invasion depends, at least partly, on the thrombospondin (see later). The host plasma membrane is invaginated to
domains on the circumsporozoite protein (CSP) and the form the parasitophorous vacuole. Immediately after inva-
thrombospondin-related adhesive protein (TRAP). CSP and sion, the developing parasites appear as fine “ring forms.”
TRAP bind to heparan sulfate proteoglycans on the surface Between 10 and 15 hours, the cytoplasm appears to thicken,
of hepatocytes. Within these cells rapid multiplication takes and 16 hours after invasion granules of the black pigment
place over 4–10 days to produce a liver schizont containing hemozoin, the product of hemoglobin digestion, begin to
about 30 000 merozoites. When ready to leave, the parasite appear. At this time receptors are expressed at the surface
196 Molecular Hematology

of the infected red blood cell that mediate adhesion to host malaria infection from non-human primates may follow.
ligands not only on venular endothelium, but also on pla- Indeed, a zoonotic outbreak of Plasmodium simium parasites
centa, uninfected red blood cells, platelets, and dendritic in humans in Brazil and a first case of naturally infected Plas-
cells, and many of these interactions have been associated modium cynomolgi in human have been described recently.
with pathology. These mature infected red blood cells or
trophozoites are sequestered in tissue beds as they adhere to
post-capillary venular endothelium. Nuclear division begins, Invasion of red blood cells
at about 30 hours, to form schizonts containing up to 32
merozoites. At 48 hours the red blood cell is ruptured to The invasion of erythrocytes by malaria parasites is a com-
release the merozoites into the circulation to continue fur- plex process and requires many specific molecular interac-
ther cycles of asexual multiplication. tions. These interactions are self-evidently required for the
The erythrocytic cycle of growth may yield a 10-fold survival of the parasite, and it would seem likely that the par-
increase in parasitemia in vivo and a patent or microscop- asite proteins involved in these interactions would be promis-
ically detectable infection six days after the liver stage is ing candidates for malaria vaccines.
completed. After two or more cycles, the infection is often Erythrocyte invasion by malaria parasites is a multistep
manifested by the paroxysms of fever that accompany the process. After initial binding with an erythrocyte, the mero-
release of merozoites. Growth continues until parasite multi- zoite reorients itself so that its apical end faces the erythro-
plication is reduced by chemotherapy, specific or non-specific cyte. A junction forms between the apical surface of the
defense mechanisms, or occasionally the demise of the host. merozoite and the erythrocyte, and several apical organelles
When fully grown, some merozoites become committed such as the rhoptries, micronemes, and dense granules dis-
during the previous erythrocytic cycle to form male or female charge their contents to create an invagination in the erythro-
gametocytes, which are distinguished by dispersed pigment cyte surface. As the merozoite moves into this invagination,
in a single nucleus. They are sequestered for the first five the junction at the apical end transforms into a circumferen-
days of their development in the peripheral circulation, but tial ring that moves around the merozoite and pinches closed
later appear as circulating, mature, crescent-shaped gameto- behind the merozoite when invasion is complete.
cytes. The sexual phase of the parasite life cycle begins after The receptors that mediate the invasion of red blood cells
male and female gametocytes are ingested by a feeding female are found in the rhoptries, micronemes, and on the cell sur-
Anopheles mosquito. In the midgut of the mosquito the game- face of merozoites. The intracellular location of many of
tocytes shed the red blood cell membrane, which appears to these parasite receptors and a rapid invasion process may, at
be precipitated by a drop in ambient temperature. A female least in part, allow the parasite to evade protective immune
gametocyte forms a single macrogamete, but male gameto- responses.
cytes undergo several rounds of nuclear division to produce
flagellated motile microgametes, which migrate to fertilize a
Use of host ligands during red blood
macrogamete. The resulting zygote forms a mobile ookinete
cell invasion
and migrates through the epithelial wall of the midgut to
rest on the external surface. The oocyst divides repeatedly to Plasmodium vivax and knowlesi are completely dependent
form about 10 000 sporozoites, which enter the acinar cells on interaction with the Duffy blood group antigen for the
of the salivary glands and are infective when injected into the invasion of human red blood cells. Electron microscopy
human host. studies have shown that the interaction of P. knowlesi, and
In P. vivax and P. ovale infections, some sporozoites enter- by analogy P. vivax, merozoites with the Duffy antigen as
ing the liver form dormant forms or hypnozoites, which only ligand is necessary for junction formation during invasion.
begin to divide some months later to cause further relapses. This blood group antigen is a 38-kDa glycoprotein with
P. ovale is now officially two parasites – P. ovale wallikeri and 7 putative transmembrane segments and 66 extracellular
P. ovale curtisi. In P. malariae, erythrocytic development takes amino acids at the N-terminus (Figure 15.3). The Duffy
72 hours and thus causes quartan fever (i.e. on days 1 and 4). antigen serves as a receptor for a family of proinflammatory
A fifth human malaria parasite has been identified in cytokines that includes the chemokines interleukin (IL)-8,
Southeast Asia. P. knowlesi, whose normal hosts are long- melanoma growth-stimulating activity, and monocyte
tailed and pig-tailed macaques, has been shown to be a major chemotactic peptide 1. Indeed, IL-8 and melanoma growth-
cause of malaria in Malaysia – particularly on the island of stimulating activity inhibit the invasion of Duffy-positive
Borneo. This example of a zoonotic malaria should not be human erythrocytes by P. knowlesi, with 50% inhibition
surprising. Malaria is a historic zoonosis: both P. falciparum achieved at nanomolar concentrations in vitro, suggesting
and P. vivax became human parasites after independent that it may be possible to block the critical step of junction
host switches from African primates. Further examples of formation during invasion. The binding site for P. vivax and
Molecular pathogenesis of malaria 197

Parasite (protein) Host receptors


P. vivax DBL Duffy blood group
(PvDBP)
P. falciparum DBL DBL Glycophorin A
(EBA-175)

P. falciparum
M1/2 Unknown
(AMA-1)
P. vivax
Reticulocytes
(RBP-1)
P. falciparum Trypsin-resistant and neuraminidase-
(Pf-NBP1) sensitive receptor on red cells
P. vivax
Transferrin receptor
(Pv-RBP2b)
P. falciparum
CR1
(Pf-RH4)
P. falciparum
Basigin
(Pf-RH5)
Fig. 15.3 Apical protein families involved in erythrocyte invasion. Bar diagrams represent the various protein families found in apical
organelles of Plasmodium species merozoites that mediate erythrocyte invasion. Multiple P. falciparum PvRBP-2 orthologs, referred to as PfRH1,
PfRH2a, PfRH3, PfRH4, and PfHR5, have also been identified.

P. knowlesi has been mapped to a 35-amino-acid segment and severe malarial anemia. The allele encodes a protein
of the extracellular region at the N-terminus of the Duffy where the extracellular domain of GYPB joins the transmem-
antigen. Sulfation of tyrosine 41 within this region of the brane and intracellular domains of GYPA, creating a peptide
Duffy antigen is critical for recognition by P. vivax and sequence at their junction that defines the Dantu antigen in
P. knowlesi. the MNS blood group system. Parasite growth has previously
Unlike P. vivax and P. knowlesi, P. falciparum does not use been shown to be impaired in Dantu+ cells, consistent with
the Duffy antigen as a ligand during invasion. Instead, stud- the new epidemiological findings that the Dantu blood group
ies have shown that sialic acid residues of glycophorin A are antigen is associated with protection from malaria.
ligands for P. falciparum during invasion. A 175-kDa P. fal- While P. vivax is completely dependent on a single path-
ciparum protein, also known as erythrocyte binding antigen way for invasion, P. falciparum uses multiple invasion path-
(EBA)-175, binds sialic acid residues on glycophorin A dur- ways and is not confined to using sialic acid residues of
ing invasion. EBA-175 does not bind En(a)- erythrocytes, glycophorin A. Some P. falciparum laboratory strains use
which lack glycophorin A, but retain glycophorin B and other sialic acid residues on glycophorin B as ligands. Other strains
sialoglycoproteins, suggesting that sialic acid residues and are capable of invading neuraminidase-treated erythrocytes
the peptides from glycophorin A are required for binding. using sialic acid–independent ligands.
A 63-amino-acid tryptic glycopeptide from the extracellular Field isolates collected from patients with malaria com-
region of glycophorin A, which blocks binding of EBA-175 to monly use alternative invasion pathways independent of
human erythrocytes, serves as the binding site. It is possible sialic acid residues of glycophorin A, and recently the Ok
that EBA-175 makes contact directly with the peptide back- blood group antigen, basigin, has been identified as a recep-
bone of glycophorin A, or that the peptide backbone presents tor for PfRh5, a parasite ligand that is essential for blood
sialic acid residues in the correct conformation for binding. stage growth. These multiple invasion pathways may provide
A novel malaria resistance locus was identified close to the P. falciparum with a survival advantage when faced with host
cluster of genes encoding glycophorins, and a haplotype at immune responses or receptor heterogeneity in natural infec-
this locus provides 33% protection against severe malaria. tions, and pose a daunting challenge for vaccine design.
The protective allele was not immediately obvious, and a
series of careful genetic analyses demonstrated complex copy
Parasite receptors used during invasion
number variants at this locus. Genome sequencing defined 8
deletions and 8 duplications, as well as 11 singleton variants. Parasite proteins that bind erythrocyte receptors during
The combined allele frequency of glycophorin copy number invasion belong to the erythrocyte-binding protein (EBP)
variants in African populations was 11% compared to 1.1% family (Figure 15.3). The EBP family includes multiple
in non-African populations. One of the variants, DUP4, was P. vivax and P. knowlesi Duffy-binding proteins (PvDBP
associated with a 40% decreased risk of both cerebral malaria and PkDBP), P. knowlesi β and γ proteins, which bind
198 Molecular Hematology

Duffy-independent receptors on rhesus erythrocytes, and The ability to transfect malaria parasites has allowed func-
P. falciparum EBA-175 and EBA-140. tional analysis of parasite proteins. Replacement of the EBA-
The extracellular region of each EBP contains two con- 175 gene with a mutant copy encoding a truncated EBA-175
served cysteine-rich domains, regions II and VI, which protein without region VI and the transmembrane and cyto-
contain conserved cysteines and hydrophobic amino acid plasmic segments has no effect on localization of EBA-175 in
residues. P. falciparum EBA-175 contains a tandem dupli- micronemes. Disruption of the EBA-175 gene in the P. falci-
cation in region II of the N-terminal cysteine-rich region. parum strain Dd2/NM, which uses sialic acid–independent
The functional binding domain of each EBP maps to region invasion pathways, has no effect on invasion or growth rates.
II. Region II of PvDBP binds the human Duffy antigen, However, when the EBA-175 gene is disrupted in the P. fal-
region II of PkDBP binds both human and rhesus Duffy ciparum strain W2Mef, which is dependent on sialic acid
antigens, P. knowlesi β region II binds sialic acid residues residues on glycophorin A, invasion is reduced. P. falci-
on rhesus erythrocytes, and P. knowlesi γ region II binds parum W2Mef transfectants expressing truncated EBA-175
as yet unidentified Duffy-independent receptors on rhesus use a sialic acid–independent invasion pathway, demonstrat-
erythrocytes. A member of the reticulocyte binding protein ing that P. falciparum can switch to alternative invasion path-
(RBP) family, normocyte binding protein Xa (NBPXa), is ways. The ability to switch to alternative invasion pathways
responsible for human red cell infectivity in P. knowlesi. In may allow P. falciparum to escape intervention strategies that
the case of P. falciparum EBA-175, region II binds sialic acid target EBA-175.
residues on human glycophorin A. Region II of EBA-175 and PvDBP have been expressed as
The EBA-140 ligand is a paralogue of P. falciparum secreted functional proteins that bind erythrocytes with the
EBA-175 protein. They share homology of domain struc- correct specificity. Antibodies directed against region II of
ture, including region II, with two homologous F1 and F2 PvDBP block the binding of PvDBP to erythrocytes, and anti-
domains, and are responsible for ligand–erythrocyte receptor bodies to region II of EBA-175 inhibit the invasion of ery-
interaction during invasion. Recombinant EBA-140 region throcytes by P. falciparum in vitro, providing support for their
II binds to the endogenous and recombinant glycophorin C, development as malaria vaccines. Somewhat surprisingly,
but does not bind to Gerbich-type glycophorin C nor to gly- high-affinity antibodies to EBA-175 inhibit invasion by P. fal-
cophorin D, the truncated form of glycophorin C which lacks ciparum strains that use alternate sialic acid– glycophorin A–
the N-glycan, or to desialylated GPC. independent invasion pathways. Antibodies directed against
EBA-175 region II may inhibit invasion via such other path-
ways by steric hindrance after binding to EBA-175 at the api-
cal surface. Given the limited homology between region II
Structural basis for the interaction of of EBP homologs, it is unlikely that antibodies raised against
EBPs with host receptors EBA-175 will cross-react with its homologs to block receptor
binding.
Regions containing binding residues have been mapped to The crystal structures of PvDBP region II (PvRII) and
domains of PvDBP and P. knowlesi β protein, which contain region II (F1–F2) of EBA-175 (EBA175-RII), together with
approximately 350 amino acids with 12 conserved cysteines. the identification of binding residues, reveal interesting fea-
Chimeric binding domains containing stretches of P. vivax tures of receptor recognition by these diverse DBL domains.
region II fused to stretches of P. knowelsi β region II have While the three-dimensional structures of these domains
been expressed and used to define binding residues for are similar, EBA175-RII forms dimers, whereas PvRII is
the Duffy antigen; these residues map to a 170-amino-acid monomeric. The receptor recognition site on PvRII is fully
stretch between the fourth and seventh cysteines of P. vivax exposed, whereas four sialic acid–binding sites lie in chan-
region II, and binding residues for sialic acid map to a nels at the dimer interface in the case of EBA175-RII. Signif-
53-amino-acid stretch between the fourth and fifth cysteines icant conformational changes are predicted to be necessary
of P. knowlesi β region II. EBPs are localized in micronemes, for receptor engagement by EBA175-RII. Regions of poly-
not on the surface of merozoites. Invasion studies using morphic clusters map to the face opposite the binding site,
another Apicomplexan parasite, Toxoplasma gondii, have suggesting that the receptor recognition site is not under
shown that discharge of microneme proteins such as MIC2 selective immune pressure upon natural infection. Indeed,
is regulated by cytoplasmic-free calcium. It is possible that in a field study conducted in children residing in a malaria
cytoplasmic calcium levels may serve as a second messenger endemic region of Papua New Guinea, high-titer binding
to trigger release of EBPs to the merozoite surface from inhibitory antibodies against PvRII were rare. However, once
micronemes during invasion. The external signals that children developed anti-PvRII binding inhibitory antibodies,
trigger microneme release in Apicomplexan parasites during they were strain transcending and protected against P. vivax
host cell invasion are not yet known. infection. These observations support the development of a
Molecular pathogenesis of malaria 199

vaccine for P. vivax malaria based on region II of P. vivax binding, and provide a molecular basis for the restriction of
Duffy-binding protein. P. falciparum to its human host among other primates.

Reticulocyte-binding proteins
Apical membrane antigen 1 and
rhoptry proteins
Plasmodium vivax preferentially invades reticulocytes, which
account for only about 1–4% of circulating red blood cells.
Apical membrane antigen 1 (AMA-1) is a well-characterized
Two P. vivax reticulocyte-binding proteins (PvRBP-1 and
rhoptry protein and a leading malaria vaccine candidate.
PvRBP-2) have been identified and are localized to the apical
Sequence analysis of AMA-1 from different primate and
surface of P. vivax merozoites (Figure 15.3). They may there-
rodent malaria parasites reveals the presence of 16 conserved
fore be responsible for the recognition of reticulocytes by P.
cysteine residues. The pattern of disulfide linkages formed
vivax during invasion. The Duffy antigen is expressed on the
by these cysteine residues in P. falciparum AMA-1 has
surface of mature erythrocytes as well as reticulocytes, but an
been experimentally determined. Based on the pattern of
irreversible junction forms only when the P. vivax merozoite
disulfide linkages, AMA-1 can be divided into three domains
encounters a reticulocyte.
(Figure 15.3). Regions M1 and M2 from the N-terminal
Now, transferrin receptor 1 (TfR1) has been identified
cysteine-rich regions of MAEBL share homology with the
as the receptor for P. vivax reticulocyte-binding protein 2b
first two domains of AMA-1. AMA-1 is detected in the
(PvRBP2b). TfR1 mutant cells deficient in PvRBP2b bind-
rhoptries of mature merozoites in late-stage schizonts, is
ing were refractory to the invasion of P. vivax, but not to
proteolytically processed at the time of schizont rupture,
the invasion of P. falciparum. PvRBP2b monoclonal antibod-
and is subsequently evenly distributed over the merozoite
ies that can inhibit reticulocyte binding also block P. vivax
surface. Anti-AMA-1 antibodies inhibit erythrocyte inva-
entry into reticulocytes, providing concrete evidence that the
sion, and immunization with purified AMA-1 provides
TfR1–PvRBP2b invasion pathway is critical for the recogni-
protection against parasite challenge. Furthermore, the
tion of reticulocytes during P. vivax invasion.
gene for P. falciparum AMA-1 cannot be knocked out using
Multiple P. falciparum PvRBP-2 orthologs, referred to as
gene-replacement methods. This evidence suggested that
PfRH1, PfRH2a, PfRH3, PfRH4, and PfHR5, have also been
AMA-1 plays a critical, non-redundant role in invasion.
identified (Figure 15.3). Like PvRBP-2, the P. falciparum
Invasion of merozoites into a host red cell requires
orthologs are localized at the apical surface of merozoites and
formation of an electron dense ring at the parasite–host cell
bind erythrocyte receptors to mediate invasion. Erythrocyte
interface called the moving junction (MJ), through which
binding has been established for PfRH1, PfRH2, and also for
the parasite inserts itself into the cell in an active motile
PfRH4, which binds to CR1.
process, akin to the migration of cells over a surface. In
However, the most exciting development has been the
the MJ, AMA1 binds to its receptor, the parasite-encoded
identification of the host receptor for the P. falciparum
rhoptry neck protein (RON) complex, which is targeted to
ligand PfHR5. A series of elegant experiments revealed
the host cell membrane from merozoite organelles early in
an unexpected pathway for red cell invasion. Erythrocyte
invasion. Crystal structures of AMA1 have shown that a par-
invasion was inhibited by soluble basigin or by knocking
tially mobile loop, termed the DII loop, forms part of a deep
down basigin, and invasion could be completely blocked
groove in domain I and overlaps with the RON2 binding site.
using low concentrations of anti-basigin antibodies across all
Rhoptry proteins also play an important role in the forma-
laboratory strains. Furthermore, Ok(a-) erythrocytes, which
tion of new permeability pathways (NPPs) in Plasmodium-
express a basigin variant that has a weaker binding affinity
infected erythrocytes, which allow nutrient uptake. RhopH2
for PfRh5, had reduced invasion efficiencies. However, it is
interacts with RhopH1, RhopH3, the erythrocyte cytoskele-
puzzling that Ok blood group antigens are not obviously
ton, and exported proteins involved in host cell remodeling. If
polymorphic in malaria endemic regions. Nevertheless,
RhopH2 expression is reduced, uptake of nutrients and vita-
these findings have provoked renewed interest in further
mins and parasite growth are reduced.
explorations of PfRh5 as a vaccine candidate antigen. A
series of papers has demonstrated that recombinant PfRh5
can induce antibodies in humans that block the invasion of
red blood cells by merozoites, and a recombinant chimeric Merozoite surface protein 1 and
antibody (Ab-1) against basigin inhibited the PfRH5–basigin invasion
interaction and blocked erythrocyte invasion across multiple
parasite strains. Intriguingly, binding of PFRh5 to basigin Merozoite surface protein (MSP)-1 was the first merozoite
may be responsible for the species specificity of PfRH5 surface protein to be identified and has been extensively
200 Molecular Hematology

investigated. Homologs of MSP-1 have been identified from


a number of primate malaria parasites, as well as several
Adhesion of infected red blood cells to
rodent malaria parasites. Sequence analysis shows that,
host cells
within a species, MSP-1 contains conserved and more poly-
Plasmodium falciparum is distinguished from the other
morphic regions. Interestingly, MSP-1 undergoes extensive
human malarias by the adherence of a very high propor-
proteolytic processing, either during schizogeny or soon after
tion of the more mature infected red blood cells, containing
merozoite release. Proteolysis results in four proteolytic frag-
asexual parasites, and the first stages of gametocytes to post-
ments of 83 kDa (N-terminus), 30 kDa, 38 kDa, and 42 kDa
capillary venular endothelium. The host ligands and parasite
(C-terminus). These polypeptides remain non-covalently
receptors that mediate the sequestration of ring-stage para-
associated at the merozoite surface. The C-terminal
sites have been widely studied, as there is considerable evi-
42-kDa fragment is attached to the plasma membrane of the
dence that the adhesion of infected red blood cells to some
merozoite by a glycosylphosphatidylinositol (GPI) anchor.
host ligands is associated with the development of severe
Final processing steps cleave the 42-kDa fragment into a
clinical disease and evasion and indeed modulation of the
33-kDa fragment and then into a 19-kDa fragment. Because
immune system.
MSP-1 is evenly distributed over the merozoite surface, it
Sequestration of infected red blood cells begins with a
is suggested that MSP-1 may mediate the initial interaction
similar sequence of events to that seen during the adhesion
with the erythrocyte. Full-length P. falciparum MSP-1 binds
of leukocytes to endothelium. Using a flow cell chamber to
erythrocytes in a sialic acid–dependent manner.
mimic the shear forces believed to be present in vivo, infected
Antibodies directed against MSP-119 block erythrocyte
red blood cells can be seen to roll across the substrate (indi-
invasion, and immunization with recombinant MSP-119 pro-
cating receptor–ligand interactions with very rapid on and off
tects mice as well as monkeys against parasite challenge, pro-
rates). Most host receptors are involved with tethering and
viding support for a blood-stage vaccine based on MSP-1.
rolling, but only two receptors, CD36 and chondroitin sul-
Invasion-inhibitory antibodies directed against P. falciparum
fate A (CSA), support the adhesion of infected red blood cells
MSP-119 block proteolytic processing of MSP-1. Following
under flow.
natural exposure to P. falciparum infections, individuals in
Electron microscopy of post-mortem tissues has shown
endemic areas acquire antibodies that can block the bind-
that parasites are sequestered in various organs, including
ing of processing-inhibitory antibodies to MSP-1. It may be
the heart, lung, brain, liver, kidney, subcutaneous tissues, and
important to design a subunit vaccine based on MSP-119
placenta. Post-capillary venular endothelium and placental
that elicits processing-inhibitory antibodies without eliciting
syncytiotrophoblasts in placenta express a variety of indu-
antibodies that block their binding to MSP-1.
cible and non-inducible receptors. Parasites isolated from
It has not been possible to knock out the P. falciparum
natural infections or laboratory strains can bind in variable
MSP-1 gene using gene-targeting methods, indicating that
numbers to many different receptors. The total number of
MSP-1 is essential for invasion and survival. Moreover, P.
parasites, the specificity and degree of binding to host ligands,
falciparum MSP-119 has limited sequence diversity, possibly
and the expression of these receptors are thought to deter-
because of functional constraints, and this provides a fur-
mine the tissue and organ distribution of parasites, and quite
ther advantage for vaccine development. However, it has been
possibly the clinical syndromes observed in severe malaria.
shown that Plasmodium chabaudi MSP-119 can function-
Adhesion of P. falciparum-infected red blood cells to
ally replace the P. falciparum domain, even though the two
endothelium and other host receptors is mediated by a par-
sequences are highly divergent, suggesting that the sequence
asite protein expressed at the cell surface, the aptly named
of MSP-119 may not be tightly constrained by function.
P. falciparum erythrocyte membrane protein 1 (PfEMP1).
This observation raises the possibility that introduction of
PfEMP1 is encoded by a large var gene family. As the name
a vaccine based on MSP-119 may result in the selection of
suggests, these proteins are highly variable, not only antigeni-
P. falciparum parasites with mutant MSP-119 domains that
cally but also in their ability to bind host ligands. Each par-
are functional, but antigenically divergent, allowing parasites
asite cell contains 60 or so different var genes, but only one
to escape host immune responses. Several trials have used
is expressed at any one time and the cell can rapidly switch
MSP-1 as a candidate vaccine antigen, but immunogenicity
expression from one gene to another at a rate of 2–4% per
has been poor, highlighting some of the problems faced in
cycle, a phenomenon known as clonal antigenic variation.
designed a complex subunit vaccine for a protozoal pathogen.
Isolates of parasites from patients or laboratory lines there-
Erythrocyte invasion by malaria parasites is a multistep
fore include infected red blood cells that contain heteroge-
process that is mediated by highly specific molecular interac-
neous antigenic and adhesive phenotypes.
tions that, once understood, may help in the rational design
PfEMP1 has a complex domain structure (Figure 15.4).
of vaccines that attempt to block erythrocyte invasion and
The extracellular region of PfEMP1 is composed of variable
prevent malaria.
Molecular pathogenesis of malaria 201

Parasite (protein) Host receptors

P. falciparum CD36, ICAM-1,


DBL1α CIDR1α DBL2β C2 DBL3γ DBL4ε DBL5δ CIDR2β ATS
(PfEMP1) CSA, etc.
Fig. 15.4 Plasmodium falciparum erythrocyte membrane protein (PfEMP)-1. The extracellular region of PfEMP-1, which is expressed on the
infected red blood cell surface, is composed of variable numbers of five possible DBL domains, named after their homology to the DBL domains
involved in red blood cell invasion, and one or two cysteine-rich interdomain regions (CIDRs).

numbers of different DBL domains, named after their cytoprotective pathways, and may have a significant role on
homology to the DBL domains involved in red blood cell the pathology of cerebral malaria and aid the development
invasion, and one or two cysteine-rich interdomain regions of new malaria interventions.
(CIDRs). DBL domains have been classified into five different However, the closest linkage between tissue-specific adhe-
classes, and the binding domains for several host receptors sion and pathology is for malaria during pregnancy (Fig-
have been mapped to various DBL and CIDR domains. ure 15.5). Pregnant women are more susceptible to malaria
PfEMP1 is involved in many pathogenic processes (Fig- and not only suffer severe acute malaria and chronic anemia,
ure 15.5). Almost all infected red blood cells can bind but also experience premature delivery, low birthweights, and
to thrombospondin and the platelet glycoprotein CD36, increased neonatal mortality. Parasitized red blood cells are
while a subset of infected cells expressing different mem- sequestered in the placenta in pregnant women and, unlike
bers of the PfEMP1 family can bind to intercellular adhe- isolates from other groups of patients, can bind to CSA but
sion molecule (ICAM)-1, platelet endothelial cell adhesion not to CD36. This adhesive phenotype is linked to expression
molecule (PECAM)-1, P-selectin, αvβ3 integrin, vascular of a single PfEMP1 with a DBL domain that binds CSA and a
cell adhesion molecule (VCAM)-1, and, perhaps of relevance non-CD36-binding CIDR1, while CD36-adherent parasites
for cerebral malaria, endothelial protein C receptor (EPCR). express a PfEMP1 with a CD36-binding CIDR1. The appar-
Adhesion of infected red blood cells or “rosetting” has been ently restricted molecular structures associated with placen-
associated with severe disease, and in different strains may be tal malaria suggest that the CSA-binding PfEMP1 may be a
mediated by CR1, ABO blood group antigens, glycosamino- good candidate antigen to prevent malaria in pregnancy.
glycans, and/or IgM. Similarly, the ability of infected red Immunity to malaria has a major role in controlling
blood cells to adhere to platelets and form large clumps disease. Intriguingly, epidemiological evidence suggests that
has also been associated with severe disease and is medi- immunity to severe forms of malaria may be gained after
ated by the adhesion of infected red blood cells to CD36 or one or two episodes of infection, and experimentally this has
to the complement receptor gC1qR. The exact relationship been achieved in the absence of an antibody response. Even
between these molecular adhesive phenotypes and disease is after many tens of exposures, people may develop immunity
not entirely clear, but it seems possible that the formation to disease, but are not refractory to asymptomatic infection.
of multicellular aggregates and/or binding of infected cells Again, the immune responses that underlie this form of
to a ligand expressed on a particular endothelial bed could immunity are not clear. However, anti-PfEMP1 antibodies
contribute to microvascular obstruction and tissue pathology do play some role in protecting against pathogenic infec-
and organ dysfunction (Figure 15.5). tions. Exposure to parasites that sequester in the placenta
For example, sequestration of parasites in the brain may during pregnancy induces strain-transcending immunity
be related to cerebral malaria or coma and may involve that stops infected red blood cells adhering to CSA and
adhesion of infected red blood cells to the ICAM-1 receptor may protect a multigravid mother and fetus from placental
and/or EPCR (Figure 15.5). Severe childhood malaria is malaria. During the development of clinical immunity,
associated with expression of specific PfEMP1 subtypes particularly during early childhood, strain-specific anti-
containing domain cassettes (DCs) 8 and 13. EPCR, which bodies to PfEMP1 are important in preventing infection
mediates the cytoprotective effects of activated protein C, with previously encountered isolates. It also appears that
has been identified as the endothelial receptor for DC8 virulent isolates from children with severe malaria are not
and DC13 PfEMP1. EPCR binding is mediated through the rare, but, on the contrary, are more commonly recognized
amino-terminal cysteine-rich interdomain region (CIDR1α) by children’s sera than isolates from cases with mild malaria.
of DC8 and group A PfEMP1 subfamilies, and this CIDRα1 It remains possible that the functionally significant regions
interferes with protein C binding to EPCR. This PfEMP1 of PfEMP1 that mediate binding to CD36, CSA, or EPCR
adhesive property links P. falciparum cytoadhesion to a host have conserved epitopes that can be targeted for vaccine
receptor, EPCR, involved in anticoagulation and endothelial development.
202 Molecular Hematology

HA CSA TSP ICAM-1 ELAM-1 VCAM-1 CD36 PECAM


P-Sel (CD31)

Placenta Brain Microvasculature


CR1
HS-like GAGs, IgM,
blood group A

Rosetting

Placental malaria Cerebral malaria Microvascular adhesion


CD36
Sequestration

PfEMP1 variant antigens Clumping

CD36

A B C D E F

Dendritic cell
Antigenic distinct waves of parasitaemia
Fig. 15.5 Plasmodium falciparum erythrocyte membrane protein (PfEMP)1 and pathology. PfEMP1 expressed on the surface of mature
red blood cells infected with P. falciparum undergoes clonal antigenic variation and can bind to many host receptors through its multiple adhesion
domains. The different properties of PfEMP1 – sequestration for evading clearance in the spleen and antigenic variation for evading
antibody-dependent killing – contribute to the virulence and pathogenesis of P. falciparum and are essential for survival of the parasite. Parasite
sequestration in the brain and placenta contribute to the complications of cerebral malaria and placental malaria, respectively. Simultaneous
binding to several receptors, binding of uninfected erythrocytes (rosetting), and clumping of infected erythrocytes through platelets are associated
with the pathogenesis of malaria. Parasite-infected red blood cells binding to dendritic cells downregulate the host immune response. CR1,
complement receptor 1; ELAM-1, endothelial/leukocyte adhesion molecule 1; HA, hyaluronic acid; HS-like GAGs, heparin sulfate-like
glycosaminoglycans; P-Sel, P-selectin; PECAM (CD31), platelet endothelial cell adhesion molecule 1; TSP, thrombospondin; VCAM-1, vascular cell
adhesion molecule 1.
Source: Miller L.H., Baruch D.I., Marsh K., Doumbo O.K. (2002). The pathogenic basis of malaria. Nature 415: 673–679, with permission.

The exact relationship between the adhesion of infected


red blood cells and pathology may involve obstruction
Malarial anemia
of the microcirculation, initiation of a local inflammatory
In endemic areas, the etiology of anemia is complex. In
response, and/or endothelial activation and immune mod-
children, acute or chronic malaria infection is a major pre-
ulation. Understanding such processes may define possible
cipitating factor of severe anemia causing admission to hos-
routes for therapy of severe disease.
pital. Not infrequently, patients present with a hemoglobin
Molecular pathogenesis of malaria 203

level of less than 5 g dl−1 with or without respiratory distress The anemia of P. falciparum malaria is typically nor-
secondary to metabolic acidosis. The sudden appearance mocytic and normochromic, with a notable absence of
of hemoglobin in the urine, indicating severe intravascular reticulocytes. The anemia of malaria may be accompanied
hemolysis leading to hemoglobinemia and hemoglobinuria by modest leukocytosis, leukopenia, leukemoid reaction,
(so-called blackwater fever), was described in early studies of monocytosis, lymphocytosis, and thrombocytopenia, but
anemia in expatriates living in endemic areas, but has been these changes are neither in themselves diagnostic, nor do
more common in Southeast Asia and Papua New Guinea, they help guide management. However, malarial pigment
where some cases are associated with G6PD deficiency is often seen in neutrophils and monocytes and has been
and treatment with a variety of drugs including quinine, associated with severe disease and an unfavorable outcome.
mefloquine, and artesunate. However, some recent cases Malaria provides ample reasons for both increased red
of severe intravascular hemolysis have been identified in cell destruction and reduced red cell production (see Fig-
Africa, prompting speculation that hemolysis may be linked ure 15.6 for an overview). Destruction of red blood cells
to the use of artemisinin derivatives. is inevitable as parasites complete their 48-hour growth

Clearance of non-iRBC
RSP-2 and specific antibody
RSP-2 Immune complexes Spleen
Loss of deformability


Clearance of iRBC


Clearance of debris
iRBC and debris
Activation of MØ
Pigment and GPI-
containing monocyte
Direct effect of
parasite
Indirect effect by
cytokines and other
– mediators e.g. TNF-αα,
+ MIF, IL-12, and IL-10

+
Anemia +
EPO
Malaria infection
Erythropoiesis

KEY: Reticulocyte RBC iRBC iRBC


Ring stage Schizont stage

Fig. 15.6 Pathogenesis of malarial anemia. Severe malarial anemia is characterized by destruction of infected red blood cells (iRBC) following
schizogony and clearance of both iRBCs and uninfected RBCs. During malarial infection, ring surface protein (RSP)-2 may be deposited on the
membrane, immune complexes may be absorbed, and senescent-type changes may occur. The resultant immune complexes of RBCs, antigen, and
immunoglobulin, e.g. RBC : RSP2 : Ig, are cleared by splenic macrophages (MØ). Pigment-containing macrophages may release inflammatory
cytokines and other biologically active mediators such as 4-hydroxynonenal. Macrophage inhibitory factor (MIF) may be released by macrophages
or a plasmodial homolog may suppress erythropoiesis. Malarial pigment or other parasite products may have a direct inhibitory effect on
erythropoiesis. Inhibition of erythropoiesis may be at one or more sites in the growth and differentiation of hematopoietic progenitors. Both
indirect and direct effects may cause suppression of the bone marrow and spleen, resulting in inadequate reticulocyte counts for the degree of
anemia. EPO, erythropoietin; GPI, glycosylphosphatidylinositol anchors of merozoite proteins; Hz, hemozoin.
204 Molecular Hematology

cycle and lyse their temporary host cell. Some parasites may defense mechanism, maximizing the clearance of parasitized
be removed from erythrocytes as immature ring forms by erythrocytes.
phagocytic cells, leaving the red blood cells with residual par-
asite antigens to continue to circulate, albeit with reduced
survival. Infected erythrocytes may also be phagocytosed by Ineffective erythropoiesis in malaria
macrophages following opsonization by immunoglobulins
and/or complement components. Other signals for recogni- Reticulocytopenia has been confirmed in numerous clinical
tion of infected erythrocytes by macrophages include abnor- studies of malarial anemia. The histopathological study of
mally rigid membranes, exposure of phosphatidylserine, and the bone marrow of children with malarial anemia shows
other altered host antigens. erythroid hyperplasia with increased numbers of erythroid
Changes to uninfected red blood cells also contribute to precursors. However, maturation is abnormal as observed
their own enhanced clearance by phagocytes. The activ- by light and electron microscopy. Abdalla and Weatherall
ity and number of macrophages are increased in malarial described the hallmark characteristics of such abnormal
infection, and increased removal of uninfected cells may maturation, namely cytoplasmic and nuclear bridging and
occur. Moreover, the molecular signals for recognition of irregular nuclear outline. They later confirmed that the
uninfected erythrocytes for removal by macrophages are distribution of the erythroid progenitors through the cell
enhanced. Uninfected erythrocytes bind increased amounts cycle is abnormal in malarial anemia, with an increased
of immunoglobulin and/or complement, as detected by the proportion of cells in G2 phase compared with normal con-
direct antiglobulin test (Coombs test). These antibodies do trols. Ineffective erythropoiesis also contributes to anemia
not have a specificity, but may instead represent immune in animal models of malaria. A recent study has shown that
complexes adsorbed onto the surface of red blood cells. vaccinated Aotus monkeys, after a challenge infection, may
Moreover, red cells from malaria patients are not only more develop moderate to severe anemia following rapid clearance
susceptible to phagocytosis, but also show increased surface of uninfected erythrocytes but with low reticulocyte counts,
IgG and deficiencies in CR1 and CD55 compared with con- indicating bone marrow dysfunction.
trols. Uninfected red cells in children and adults with severe Given the importance of EPO to erythropoiesis, attention
disease are less deformable and this is a significant predictor has focused on the levels of this crucial cytokine in malarial
of the severity of anemia and indeed outcome, consistent with infection. Serum EPO was appropriately raised in a single
the notion that these cells are being removed by the spleen. study of African children suffering from malarial anemia.
Active erythrophagocytosis is a conspicuous feature within However, other studies in adults from Thailand and Sudan
the bone marrow during P. vivax and P. falciparum malaria, have suggested that EPO concentration, although raised,
and it is highly probable that this also occurs within the was inappropriate for the degree of anemia. The most recent
spleen. Children with acute P. falciparum malaria have high studies of EPO in African children have shown a supra-
circulating levels of interferon (IFN)-γ and tumor necrosis physiological rise in EPO levels compared with age-matched
factor (TNF)-α, a synergistic combination of cytokines that community controls with non-malarial anemia, and there
activates macrophages. appears to be a defective response to EPO in malaria, at least
Parasite proteins may also contribute to the clearance of in these children.
uninfected red blood cells. The ring surface protein (RSP)-2 The prime candidates for the host factors mediating
is expressed shortly after merozoite invasion of red blood dyserythropoiesis are imbalances in TNF-α, IFN-γ, and
cells and is widely deposited on uninfected red blood cells IL-10. The concentrations of TNF-α and IFN-γ have been
by contact with merozoites in the circulation. Opsonization correlated with the severity of the disease. While low con-
of these RSP-2-bearing uninfected red blood cells could centrations of TNF-α (<1 ng ml−1 ) stimulate erythropoiesis,
contribute to erythrophagocytosis. Indeed, high levels higher levels of TNF-α have been shown to suppress ery-
of antibodies that could facilitate complement-mediated thropoiesis. Furthermore, it is possible that high levels of
phagocytosis of cells expressing RSP-2 are found in sera from these inflammatory cytokines may contribute to reduced and
immune adults and children with severe anemia. The RSP-2 abnormal production of erythrocytes, and also to increased
antigen is also present on the surface of erythroblasts in the erythrophagocytosis.
bone marrow of P. falciparum-infected patients and it has High levels of the Th2 cytokine IL-10 might prevent the
been suggested that damage to developing erythroid cells by development of severe malarial anemia. Low levels of IL-10
RSP-2 and anti-RSP-2 antibody could also contribute to the have been described in African children with severe malar-
development of anemia. ial anemia. It has been suggested that IL-10 may induce
Thus, all the available evidence points to increased retic- heme oxygenase and so reduce oxidative damage to red blood
uloendothelial clearance in P. falciparum malaria, persisting cells and/or developing erythroid cells. Similarly, IL-12 may
long after recovery. These changes are presumably a host be associated with a protective innate immune response to
Molecular pathogenesis of malaria 205

malaria, and low IL-12 levels have been associated with severe Many possible molecular mechanisms of red blood cell
malaria in African children. clearance have been described in a variety of experimental
There is also substantial evidence that the lysate of infected and clinical systems. The challenges now are to determine
erythrocytes may directly modulate the function of host the relative contribution of these mechanisms to anemia in
cells. During its blood stage, the malaria parasite proteolyzes children with malaria, and to seek ways to prevent anemia
host hemoglobin in an acidic vacuole to obtain amino acids, without increasing susceptibility to disease.
releasing heme as a byproduct, which is autoxidized to
potentially toxic hematin: aquaferriprotoporphyrin IX or
H2 O-Fe(III)PPIX. β-hematin forms as a crystalline cyclic Future prospects
dimer of Fe(III)PPIX and is complexed with protein and
lipid products as malarial pigment or hemozoin. Arese and Molecular methods and the application of modern pro-
colleagues showed that the function of monocytes and of teomic and genomic technologies are likely to transform
monocyte-derived macrophages is severely inhibited after our approach to the diagnosis, treatment, and prevention of
ingestion of malaria pigment or hemozoin. These cells are malaria. The diagnosis of malaria is based on the identifi-
unable to repeat phagocytosis and to generate oxidative cation of circulating blood-stage parasites in thick and thin
burst when appropriately stimulated. Furthermore, after films. In endemic areas, laboratory staff are skilled at the
phagocytosis of hemozoin, myeloid cells are unable to kill examination of thick films and are routinely able to detect 1
ingested fungi, bacteria, and tumor cells and to respond to parasite in 100 high-power fields of a thick film, which corre-
IFN-γ stimulation, but instead respond by increased release sponds to a sensitivity of approximately 5–50 parasites μl−1 .
of IL-1β, TNF-α, and macrophage inflammatory protein Nevertheless, the diagnosis of malaria by microscopy in
(MIP)-1α and MIP-1β. non-endemic countries has proven problematic. Routine
The hemozoin polymer of heme moieties may be com- laboratories may only achieve sensitivities of the order of
plexed with biologically active compounds. The oxidation 500 parasites μl−1 using thick films. Detection of circulating
of membrane lipids catalyzed by ferric heme produces malarial antigens is another potentially attractive, but
lipoperoxides. There is accumulating evidence that 4- ultimately limited, alternative to the laborious method of
hydroxynonenal and other lipoperoxides, including 15- screening blood films. The widely available tests detect Plas-
hydroxy-arachidonic acid (15-HETE), may play a role in modium histidine-rich protein 2 and Plasmodium-specific
the pathophysiology of malaria. Endoperoxides produced lactate dehydrogenase by immunochromatography. The
in pigment-containing monocytes or macrophages may formulation of the tests using dipstick antigens allows rapid
impair erythroid growth. Hemozoin may also directly testing to be performed by laboratory staff. However, the
inhibit erythroid development in vitro and increased levels sensitivity is 100–1000 parasites μl−1 , which is comparable
of plasma hemozoin and pigment in monocytes have been to the sensitivity achieved by inexperienced microscopists.
associated with anemia. The current recommendations for malaria diagnosis in the
Hemozoin may have a direct effect as a proinflammatory UK emphasize clearly that these tests only have a place
danger signal that activates the NALP3 inflammasome, caus- when experienced staff are not available. Amplification of
ing the release of IL-1β. Similar to other NALP3-activating circulating parasite DNA using the repeated rRNA genes
particles, hemozoin activity is blocked by inhibiting phago- is an extremely sensitive method of malaria diagnosis.
cytosis, K(+) efflux, and NADPH oxidase. The sensitivity may be as low as 0.005 parasites μl−1 or
A further class of parasite products that may contribute 5 parasites ml−1 . This approach may be developed as a future
to malarial anemia are the GPI anchors of merozoite method for routine diagnostic use.
proteins, such as MSP-1, MSP-2, and MSP-4, which are Malaria requires urgent effective chemotherapy to prevent
found in plasma during infections. GPIs are likely to con- the progression of disease and this may be the most crucial
tribute to anemia, since their injection into mice results public health intervention to reduce the global mortality
in a transient decrease in the number of circulating red from malaria. Resistance to the first-line treatment of chloro-
blood cells, probably through induction of TNF-α from quine and to dihydrofolate reductase inhibitors has led to
macrophages. More recently, it has been demonstrated that artemisinin-based combination treatments. This group of
the proinflammatory response from human monocytes is drugs has a remarkable history and represents a novel class
through interaction of GPIs with TLR2 and, to a lesser of chemical compounds known as the artemisinins. Chinese
extent, TLR4. Antibodies specific to GPIs were present in scientists rediscovered the activity of extracts of the plant
the sera of adults from endemic regions in Kenya, but Artemisia annua from careful rereading of medieval pharma-
the level of these antibodies was less in younger children copeias. The active substance was crystallized and identified
who, in general, have more severe disease and malarial as artemisinin in 1973, and derivatives dihydroartemisinin
anemia. (DHA), artemether, artesunate, and arteether were first
206 Molecular Hematology

prepared in China in the 1970s. The mechanism of action of the parasite, human, and mosquito genomes are still being
these drugs was proposed to be within the “food vacuole,” applied, increasing our understanding not only of the patho-
where the drug could inhibit digestion of hemoglobin, but genesis of disease but of the biology of the parasite.
no single mechanism of action has been defined. In any These new approaches to advance the biology of malaria
event, artemisinin acts very early in the parasite life cycle, have been accompanied by a large international effort to
killing ring-stage parasites and so preventing the maturation develop new drugs for the disease, which are likely to be
and sequestration of malarial-infected erythrocytes. There needed, not only if artemisinin drug resistance emerges on
has been increased drug resistance, mainly in Southeast Asia, a wide scale, but also to treat the dormant liver stages (or
where drug resistance has been strongly associated with hypnozoites) of P. ovale and P. vivax. Novel high-throughput
mutation in the falciparum gene encoding a K13 propeller screening platforms, large chemical libraries, and coordi-
mutation, but this mutation has arisen against a background nated international effort has allowed the Medicines for
of a complex genetic phenotype that may have allowed the Malaria Venture to assemble 400 unique compounds with
parasite to adapt to many different drug regimens and/or blood-stage antimalarial activity for continued future devel-
grow in erythrocytes with hemoglobin variants. There are opment (see https://www.mmv.org/research-development).
now isolated case reports of emerging resistance or molecu- There are considerable scientific, technical, organizational,
lar markers of resistance to artemisinin-based combination and political problems faced in the development and imple-
therapy in Africa, where widespread resistance could reverse mentation of new methods and programs for diagnosing,
the recent advances in the burden of malaria. treating, and preventing malaria. However, the huge pro-
The search for a malaria vaccine could almost be a case grams now underway would be expected to continue to roll
study in the triumph of hope over experience. However, back the disease. In a remarkable public health achievement,
there have been some recent trials showing significant effi- Sri Lanka was certified as malaria free by the WHO in 2016,
cacy. The RTS,S/AS01 vaccine was developed by Joe Cohen which gives some hope that the eradication of malaria may be
at GlaxoSmithKline and uses a recombinant P. falciparum achieved one day, at least in areas where transmission is low
circumsprozoite protein, which is naturally expressed in and there is the necessary infrastructure to sustain complex
high levels on the surface of the infective forms injected public health programs.
by mosquitoes, fused to the hepatitis B surface protein. A
series of trials have shown a reduction in episodes of severe
malaria by 50% in phase 3 studies and Ghana, Kenya, and Further reading
Malawi partnered with the WHO in the Malaria Vaccine
Implementation Program (MVIP) to make the RTS,S/ASO1 Cowman, A.F., Healer, J., Marapana, D., and Marsh, K. (2006). Malaria:
vaccine available in selected areas of the three countries, biology and disease. Cell 167: 610–624.
beginning in 2018. This program will assess the feasibility Howes, R.E., Battle, K.E., Mendis, K.N. et al. (2016). Global epidemiol-
and safety of the vaccine in real-life settings. There are also ogy of Plasmodium vivax. Am. J. Trop. Med. Hyg. 95 (6): 15–34.
Kirchner, S., Power, B.J., and Waters, A.P. (2016). Recent advances in
over 40 subunit vaccines in different stages of development,
malaria genomics and epigenomics. Genome Med. 8: 92.
many of which are based on the antigens described in this
Lelliott, P.M., McMorran, B.J., Foote, S.J., and Burgio, G. (2015). The
chapter (see www.malariavaccine.org). influence of host genetics on erythrocytes and malaria infection: is
This chapter has described the essential features of the epi- there therapeutic potential? Malar. J. 14: 289.
demiology, clinical presentations, and life cycle of the parasite Rabinovich, R.N., Drakeley, C., Djimde, A.A. et al. (2017). malERA:
and the main features of our present molecular understand- an updated research agenda for malaria elimination and eradication.
ing of key features of the invasion of red blood cells, adhe- PLoS Med. 14 (11): e1002456.
sion of infected erythrocytes to host ligands, and how these Woodrow, C.J. and White, N.J. (2017). The clinical impact of
and other host–parasite interactions lead to cerebral and pla- artemisinin resistance in Southeast Asia and the potential for future
cental malaria and anemia. The data revealed by sequencing spread. FEMS Microbiol. Rev. 41 (1): 34–48.
Chapter 16 Molecular coagulation
and thrombophilia
Björn Dahlbäck1 & Andreas Hillarp2
1
Department of Translational Medicine, Section of Clinical Chemistry, Lund University, University Hospital, Malmö, Sweden
2
Department of Clinical Chemistry and Transfusion Medicine, Halland County Hospital, Halmstad, Sweden

Introduction, 207 Management of thrombophilia, 217


Blood coagulation, 207 Conclusions, 218
Molecular genetics of venous thromboembolism, 211 Further reading, 218

FVIIa bound to TF specifically cleaves and activates the


Introduction two vitamin K-dependent plasma proteins, factor IX (FIX)
and factor X (FX) (Figure 16.1). Activated FX (FXa) activates
Venous thrombosis is a major medical problem affecting
prothrombin to thrombin, whereas activated FIX (FIXa) acti-
millions of individuals worldwide each year. It is a typical
vates FX. Both FIXa and FXa are poor enzymes that require
multifactorial disease, the pathogenesis involving both
protein cofactors, calcium ions, and negatively charged phos-
environmental and genetic risk factors. A mutation in the
pholipid surfaces for expression of full biological activity.
factor V (FV) gene (Arg506Gln or FV Leiden) is the most
The protein cofactors for FIXa and FXa are the activated
common genetic risk factor known to date. Activated protein
forms of factor VIII (FVIIIa) and factor V (FVa), respectively
C (APC) regulates the activity of FVa by cleaving several sites
(Figure 16.2). As a result of multiple protein–protein and
in FVa, and Arg506 is one of them. APC resistance, which
protein–phospholipid interactions, enzymatically highly effi-
is the consequence of the FV mutation, results in a lifelong
cient complexes are assembled on the phospholipid surface.
hypercoagulable state. A point mutation in the prothrombin
The initiation of blood coagulation by TF is usually
gene is another relatively common risk factor, whereas
referred to as the extrinsic pathway or the TF pathway.
deficiencies of the anticoagulant proteins antithrombin
In association with injury, this is the physiologically most
(AT), protein C, or protein S are less common. Owing to
important mechanism of blood coagulation. However, coag-
the high prevalence of the FV and prothrombin mutations,
ulation can also be activated through the intrinsic pathway,
combinations of genetic defects are relatively common in the
triggered by activation of the contact phase proteins (FXII,
general population. Such individuals have a highly increased
FXI, prekallikrein, and high-molecular-weight kininogen)
risk of thrombosis.
that follows exposure of blood to certain negatively charged
surfaces. In recent years, it has been discovered that platelets
contain long-chain negatively charged polyphosphates that
Blood coagulation may serve as a natural activator for the intrinsic system.
The intrinsic pathway does not appear to be physiologically
At sites of vascular damage, circulating platelets adhere to important for injury-related coagulation in vivo, illustrated
subendothelial structures and undergo a series of reactions by the lack of bleeding problems in individuals with a defi-
that lead to primary hemostasis due to the formation of a ciency of FXII. However, the interest in the intrinsic pathway
platelet plug. Concomitant to these events, the subendothe- has increased dramatically in the last decade after the dis-
lial membrane protein tissue factor (TF) is exposed to blood. covery that it may be important for the formation of arterial
A small amount of activated factor VII (FVIIa) present in cir- thrombosis, and efforts are being made to develop inhibitors
culating blood binds to TF and triggers a series of proteolytic to the system for safe anticoagulation.
reactions that culminate in the formation of thrombin and Thrombin generated at sites of vascular injury expresses a
the conversion of fibrinogen to insoluble fibrin. number of procoagulant properties. It amplifies the coagu-
lation process by activating FXI, and in addition it activates
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. platelets and converts fibrinogen to fibrin. Moreover, in a pos-
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. itive feedback reaction, thrombin converts the procofactors

207
208 Molecular Hematology

VWF

IXa VIII
VIIa IX/X
T
IXa T
IXa T Xa
Xa
VIIIa Va
PT
Xa X Xa
IXa
TF

Extravascular cell Activated platelet


Fig. 16.1 Initiation and propagation of blood coagulation. Blood coagulation takes place on the surface of cell membranes where enzymes
and cofactors form complexes that efficiently convert their respective proenzyme substrates to active enzymes. The exposure of tissue factor (TF) to
blood, with subsequent binding of FVII/FVIIa and activation of FIX and FX, initiates the reaction sequence. The subsequent assembly of tenase
(FIXa/FVIIIa) and prothrombinase (FXa/FVa) complexes on the surface of negatively charged phospholipid membranes (mainly provided by platelets)
results in amplification, propagation of the process, and the generation of high concentrations of thrombin (T). Thrombin feedback activates FVIII
(circulates in complex with von Willebrand factor, VWF) and FV.

(a) (b)
A2 A2

R306 R506 R306

A1 A1
R506

C2 FXa ~70 A C2
~85 A
A3 A3
APC
(SP domain)
C1 C1

Membrane Membrane

Fig. 16.2 Molecular model of FVa, highlighting its interactions with FXa and APC. FVa is shown as a solid surface (domains A1, green; A2,
cyan; A3, brown; C1, pink; C2, purple) and FXa as an orange ribbon. The virtual membrane is represented as a gray box. (a) FVa/FXa complex, with
FVa residues probed experimentally to be important for FXa binding shown in white. (b) APC approaching the cleavage site at Arg506. The two
main cleavage sites of APC (Arg306 and Arg506) are indicated in red. The serine protease domain of APC is shown as a blue ribbon.
Source: Segers K., Dahlbäck B., Nicolaes G.A. (2007). Coagulation factor V and thrombophilia: background and mechanisms. Thromb Haemost
98: 530–542, with permission.
Molecular coagulation and thrombophilia 209

FV and FVIII into their biologically active counterparts (FVa to be determined. The endothelial surface also contains
and FVIIIa). shorter TFPI isoforms that are linked with glycosylphos-
phatidylinositol (GPI) anchors. They presumably serve to
inhibit premature TF-dependent activation of coagulation
Regulation of blood coagulation
on the vascular wall. Shorter isoforms are also present in
The efficient reactions of the coagulation system have plasma circulating bound to lipoproteins. The function of
considerable biological potential and strict regulation the lipoprotein-associated isoforms is not understood.
is required. For this purpose, several plasma proteins and The highly efficient procoagulant reactions of thrombin
protein–cell interactions are involved in constant monitoring are physiologically adequate at sites of vascular injury and
of the circulation. At each level of the coagulation pathway, are instrumental in efficient hemostasis. However, the same
membrane-bound molecules expressed on the surface of reactions pose a threat to the organism, as uncontrolled
intact endothelial cells, circulating inhibitors, and negative coagulation leads to thrombus formation. Nature has solved
feedback mechanisms provide efficient control. this dilemma in intricate and fascinating ways, one of which
Antithrombin (AT) is the most important serine pro- is the transformation of thrombin into an efficient initiator
tease inhibitor (serpin) involved in the regulation of blood of a natural anticoagulant pathway, the protein C system.
coagulation. AT inhibits thrombin as well as FXIa, FIXa, The conversion of thrombin from a procoagulant into an
and FXa and, under certain conditions, also FVIIa. AT anticoagulant enzyme depends on the presence of intact
forms a highly stable complex with the protease and, as a endothelium. Thus, thrombin generated at sites of intact
consequence, the protease is trapped and eliminated from vasculature binds to the endothelial membrane protein
the circulation. The activity of AT is stimulated by heparin, thrombomodulin, which is a potent modulator of thrombin
which accelerates the rate of formation of the AT–protease activity and a cofactor to thrombin in the activation of pro-
complexes. Under normal physiological conditions, heparan tein C (Figure 16.3). A receptor for protein C, the endothelial
sulfate proteoglycans present on the endothelial cell surface protein C receptor (EPCR), has been shown to stimulate the
stimulate the activity of AT, whereas heparin injections activation of protein C by the thrombin–thrombomodulin
are used in clinical situations. During the inhibition of complex. APC inactivates membrane-bound FVa and FVIIIa
thrombin, an important role of heparin is to function as by limited proteolysis in reactions that are potentiated by
a bridge between thrombin and AT. In addition, heparin a cofactor protein designated protein S and, in the case of
induces conformational changes in AT that are associated FVIIIa degradation, also by the non-activated form of FV
with the generation of a more efficient inhibitor. In the (Figure 16.4).
inhibition of FXa, the conformational change appears to be Under physiological conditions, procoagulant and antico-
more important than the bridging mechanism. agulant mechanisms are balanced in favor of anticoagulation,
The tissue factor pathway inhibitor (TFPI) regulates the whereas the anticoagulant system is downregulated and pro-
TF pathway. The full-length TFPI (TFPIα) is composed of coagulant forces prevail at sites of vascular damage. Defects
three protease inhibitory domains belonging to the Kunitz in this ingenious system are associated with increased throm-
type of inhibitors and a highly positively charged C-terminal bin generation and a hypercoagulable state, leading to an
tail. There are also shorter isoforms containing two or three increased risk of thrombosis.
Kunitz domains in circulation. TFPI has the unique capacity
to inhibit the FVIIa–TF–FXa complex and is therefore
Protein C anticoagulant system
highly efficient in turning off the TF pathway. The inhibition
mediated by TFPI occurs in two steps: the first involves Protein C is a vitamin K–dependent plasma protein that is
inhibition of FXa by the middle Kunitz domain; the first synthesized mainly in the liver. It is homologous to FVII, FIX,
Kunitz domain then binds and inhibits FVIIa. TFPI can also and FX and shares with them a common modular organi-
directly inhibit FXa, in a phospholipid-bound reaction that zation. From the N-terminus, these proteins contain a vita-
is stimulated by the vitamin K–dependent protein S. Protein min K–dependent γ-carboxyglutamic acid (Gla)-rich mod-
S interacts with the third Kunitz domain. The majority of ule, two epidermal growth factor (EGF)-like modules, and
TFPIα is bound to glycosaminoglycans on endothelial cells a serine protease module. The Gla domains bind calcium
(approximately 80%) and only a minor fraction of TFPIα is ions and provide the vitamin K–dependent clotting proteins
present in plasma, where it is mainly associated with coag- with phospholipid-binding properties. Upon activation by
ulation FV, which interacts with the C-terminus of TFPI. the thrombin–thrombomodulin complex, the serine protease
Recently, a minor splice variant of FV having a truncated module is converted to an active enzyme. APC is highly spe-
B-domain (702 amino acid residues deleted), which exposes cific in its proteolytic activity, cleaving a limited number of
a high-affinity binding site for TFPIα, is shown to be a carrier peptide bonds in FVa and FVIIIa.
for TFPIα in circulation. The relative distribution of TFPIα Intact FV is a high-molecular-weight protein and
between full-length FV and the FV splice variant remains shares with the homologous FVIII molecule the modular
210 Molecular Hematology

APC
PC APC
PC
EPCR TM EPCR
TM
T TM
PC TM PC
T T
T PC T

T
T

Fig. 16.3 Activation of protein C by thrombin–thrombomodulin. Thrombomodulin (TM) serves as a cofactor to thrombin (T) in the activation
of protein C. It is present on all endothelial cells. Endothelium also contains the endothelial protein C receptor (EPCR) that binds the Gla domain of
protein C and helps present protein C to the T/TM complex. Activated protein C (APC) remains in the bloodstream to control coagulation reactions.

arrangement A1, A2, B, A3, C1, and C2 (Figure 16.5). APC alone has poor anticoagulant activity and it is only
On activation of FV by thrombin or FXa, peptide bonds in the presence of its cofactors protein S and FV that efficient
surrounding the B-module (positions 709 and 1545) are anticoagulant function is expressed. This was demonstrated
cleaved, releasing the B-module, which is thus not part of in an experimental system based on the degradation of FVI-
FVa. FVIII is activated by thrombin in a similar fashion, IIa. In this system, it was found that full anticoagulant activity
leading to release of the B-module. In circulation, FVIII is of APC was obtained in the presence of the combination of
bound to von Willebrand factor (VWF) and the thrombin- FV and protein S. The synergistic APC cofactor activity of
mediated activation of FVIII results in the release of FVIIIa FV requires APC-mediated proteolysis of at least the Arg506
from VWF. APC cleaves three peptide bonds in FVa at cleavage site (Figure 16.5). Thus, the Arg506 → Gln muta-
Arg306, Arg506, and Arg679, whereas FVIIIa is cleaved tion is important for understanding the mechanism of APC
at Arg336 and Arg526. As a consequence of the APC- resistance (FV Leiden; see later). The APC cofactor activity
mediated cleavages, FVa and FVIIIa lose their procoagulant of FV appears specific for the degradation of FVIIIa, whereas
properties. the FVa degradation is unaffected by this FV activity. FV

S
V

APC
V S

APC VIIIa APC


T
APC
VIIIi
S
V
Va
APC Vi
APC

Fig. 16.4 Degradation of FVa and FVIIIa by APC. Both FVa and FVIIIa are cleaved and inhibited by APC in phospholipid membrane-bound
reactions that also involve cofactors to APC. Protein S and APC are sufficient to inhibit FVa, whereas the regulation of FVIIIa additionally includes
FV, which in this situation serves as cofactor to APC.
Molecular coagulation and thrombophilia 211

FV A2
bound to C4b-binding protein (C4BP), a regulator of the
A1 B
A1 A2 B A3 C1C2 A3 complement system. The Gla module of protein S provides
C2 C1
both free protein S and the protein S–C4BP complex with
phospholipid-binding ability. This is important for the
Thrombin APC localization of coagulation and complement regulatory
FXa
activities to certain cell membranes, for example to the phos-
phatidylserine that is exposed on apoptotic cells. Protein
S binding to such cells has been shown to be involved in
Procoagulant FVa Anticoagulant FVac stimulation of phagocytosis of these cells.
A1 A2 During the degradation of free FVa (i.e. not bound to
Ca2+ A1 A2 B A3 C1C2 FXa) by APC, the cleavage at Arg506 is faster than that at
A3 C1C2
Arg306. The cleavage at Arg506 leads only to partial loss of
FVa activity, whereas the cleavage at Arg306 leads to efficient
APC
Thrombin inactivation of FVa. Protein S serves as a cofactor for the
FXa cleavage at Arg306, but has minor effects on the Arg506
cleavage. This, together with a specific protection of the
Arg506 site exerted by FXa, indicates that the Arg306 site
Inactive FVi is the most important site for regulation of FVa activity in
the prothrombinase complex. On the other hand, FVa that
A1 A2
Ca2+ is not part of a prothrombinase complex is first cleaved at
A3 C1C2 the Arg506 site, because the kinetics of this cleavage are
Fig. 16.5 Procoagulant and anticoagulant properties of factor more favorable than those for the cleavage site at Arg306. In
V. Proteolytic modification of single-chain FV results in the expression experiments in vitro, protein S has been shown to express an
of either procoagulant or anticoagulant properties. Thrombin and FXa anticoagulant activity that is independent of the presence of
cleave peptide bonds in and surrounding the B domain (positions 709, APC. The exact mechanism is still unclear, despite consid-
1018, 1545) and thus activate FV to procoagulant FVa, which erable research. An anticoagulant activity of protein S that is
functions as a cofactor to FXa in the activation of prothrombin. Intact
potentially physiologically important is due to its interaction
FV is sensitive to cleavage by APC (at positions 306, 506, and 679),
with the third Kunitz domain of TFPIα and the direct
which increases the anticoagulant activity of FV. FV modified by APC
(FVac) functions as a synergistic APC cofactor with protein S in the
stimulation of the TFPIα-mediated inhibition of FXa. Other
degradation of FVIIIa. The anticoagulant properties of FV are lost on proposals suggest that protein S inhibits prothrombin activa-
further proteolysis by thrombin or FXa. Likewise, the procoagulant tion through direct interactions of protein S with FVa, FXa,
effects of FVa are lost as a result of cleavage by APC. Thus, FV plays a and the phospholipid membrane. The in vivo physiological
crucial and central role, balancing procoagulant and anticoagulant significance of this APC-independent anticoagulant activity
forces. Arrowheads denote the three APC cleavage sites at positions is unclear. Regardless of its mode of action, protein S is an
306, 506, and 679. important anticoagulant protein in vivo, as demonstrated by
protein S knockout studies and by the association between
protein S deficiency and venous thrombosis.
loses its APC cofactor activity upon proteolysis by thrombin,
but it gains procoagulant properties as a cofactor to FXa.
Thus, FV is similar to thrombin in being able to express Molecular genetics of venous
both procoagulant and anticoagulant effects. However, thromboembolism
whereas the anticoagulant effects of thrombin depend on its
binding to thrombomodulin, the anticoagulant properties The annual incidence of venous thrombosis in Western soci-
of FV are potentiated by APC-mediated proteolysis of the eties is approximately 1–2 per 1000. Thrombotic episodes
non-activated form of FV. tend to occur in conjunction with surgery, fractures, preg-
Protein S is also a vitamin K–dependent plasma protein, nancy, the use of oral contraceptives, and immobilization. In
but, unlike the other vitamin K–dependent coagulation addition, genetic defects are frequently involved and many
proteins, it is not a serine protease. It is a multimodular patients report positive family histories. Genetic defects
protein containing a Gla module, a thrombin-sensitive known to predispose to thrombosis include inherited APC
module, four EGF-like modules, and a large module homol- resistance due to FV mutations (mainly FV Leiden), a
ogous to sex hormone-binding globulin (SHBG; see later point mutation in the prothrombin gene (G20210A), and
Figure 16.8). Protein S also has functions outside the protein deficiencies of anticoagulant protein C, protein S, or AT.
C system and 60–70% of protein S in plasma circulates These inherited causes of thrombophilia are autosomal
212 Molecular Hematology

dominant disorders and the prothrombotic mutations result as a sedentary lifestyle, surgery, and the use of oral contra-
in a lifelong increased risk of thrombosis. However, the ceptives, did not affect our ancestors.
penetrance of the disease may be variable and the individual The high prevalence of the FV Leiden allele in Western
risk profile is still difficult to predict. societies is the result of a founder effect. It has been estimated
that the mutation event occurred around 21 000 years ago,
after the “Out of Africa Exodus” that took place 100 000 years
Factor V gene mutations causing APC
ago and also after the separation of Asians from Europeans.
resistance
This explains why the mutant FV allele is common among
In 1993, APC resistance was described as a cause of inher- European populations, but is not present among Japanese,
ited thrombophilia and it was soon demonstrated to be Chinese, or in the original populations of Africa, Australia,
highly prevalent (20–60%) among thrombosis patients. In or America.
APC resistance, APC does not give a normal prolongation of A large number of studies have demonstrated a relation-
the clotting time. In more than 95% of cases, the molecular ship between the presence of APC resistance (FV Leiden)
defect associated with APC resistance is a single point muta- and an increased risk of venous thrombosis. Differences in
tion in the FV gene, F5 (GeneID 2153, OMIM ID 227400, selection criteria of patients and in the prevalence of the
188055, 612309). The mutation is a G → A substitution at mutant allele in the general population explain the wide vari-
nucleotide position 1691 in the FV gene, which predicts the ation in results obtained from different studies. However, the
replacement of Arg506 with a Gln (Figure 16.5). The mutant general consensus is that the FV Leiden allele is the most
FV is known as FVR506Q, FV Leiden, or FV:Q506 (R and Q common genetic risk factor for venous thrombosis in West-
are one-letter codes for Arg and Gln, respectively). Recently, ern societies. The odds ratio, describing the increased risk
additional rare FV mutations have been described in indi- of thrombosis in affected individuals, has been calculated to
viduals with venous thrombosis and APC resistance. In a be two- to threefold for those carrying the defect in a het-
Japanese family, FVNara (W1920R) is demonstrated to be erozygous form, whereas homozygous individuals are at an
the cause of the APC resistance phenotype. In six European even higher risk of thrombosis. The FV Leiden allele does
patients, APC resistance is caused by FVBonn (A512V). In not appear to be a strong risk factor for arterial thrombo-
both FVNara and FVBonn, the FV mutations were proven sis, such as myocardial infarction. The FV Leiden allele is
to be the cause of the APC resistance phenotype. In several associated with a hypercoagulable state, which is reflected
other venous thrombosis patients with APC resistance, FV by increased levels of prothrombin activation fragments in
mutations have been found, but not experimentally proven to plasma of individuals with inherited APC resistance. Two
be the cause of the APC resistance phenotype. These muta- molecular mechanisms are involved (Figures 16.5 and 16.4).
tions are FV E666D in a Chinese family, and three mutations One is that an APC cleavage site in FVa is lost, which impairs
in venous thrombosis patients with Chilean Amerindian eth- the normal degradation of FVa by APC. The other surpris-
nic background – FV M443L, FV E461Q, and FV G493E. Two ing observation is that FV Leiden is a poor APC cofactor in
different mutations affecting the Arg306 site have recently the degradation of FVIIIa, because the cleavage at Arg506
been found in thrombosis cases, FV Cambridge and FV Hong is required for the expression of the APC cofactor activity
Kong, but such mutations appear to be rare. They do not of FV.
result in APC resistance and are not major risk factors for In the degradation of normal FVa, the APC cleavage at
thrombosis. Arg506 has favorable kinetics compared with cleavages at
The FV Leiden allele is found only in Caucasians, and other sites. The Arg506 cleavage is approximately 10-fold
its prevalence varies considerably in the general population faster than the cleavage at Arg306, and the activity of
of Western societies. High prevalence (up to 15%) is found FVa:Q506 (FVa Leiden) is therefore inhibited at an approx-
in southern Sweden, Germany, Greece, Arab countries, and imately 10-fold lower rate than FVa:R506. Generated FVa
Israel. In the Netherlands, the UK, and the USA, around 3– Leiden persists longer than normal FVa and can form active
5% of the population carry the mutant allele. A lower preva- prothrombinase complexes with FXa. However, degradation
lence (around 2%) is found in Hispanics. The high prevalence of free FVa (i.e. FVa not bound to FXa) is different from
of the FV Leiden allele in certain populations suggests a pos- that of FVa, which is part of the prothrombinase complex.
sible survival advantage, and there is a reduced risk of bleed- In the prothrombinase complex, the Arg506 site is protected
ing after delivery in women carrying the mutation. In the his- from degradation by APC by both FXa and prothrombin. In
tory of humankind, the slightly increased risk of thrombosis addition, protein S functions as an APC cofactor primarily
associated with the FV Leiden allele has presumably not been for the Arg306 cleavage. As a consequence, APC-mediated
a negative survival factor, because thrombosis develops rela- degradation of FVa, which is part of the prothrombinase
tively late in life and does not influence fertility. In addition, complex, follows a different pathway compared with that of
many of the circumstantial risk factors for thrombosis, such free FVa. Therefore, when FVa:R506 and FVa:Q506 are part
Molecular coagulation and thrombophilia 213

0 2 4 6 8 10 12 14 kb

Exon 1 2 3A 3B 4 5 6

mRNA

200 bp

Type II
Mature polypeptide chain

Type I

100 aa
Fig. 16.6 Structure of the human antithrombin gene and location of detrimental missense mutations in the antithrombin molecule.
The gene for antithrombin (SERPINC1) is localized to chromosome 1q23–q25 and spans 13.4 kb of DNA (upper part). It comprises seven exons and
results in an mRNA of 1.7 kb (middle part). The antithrombin molecule (lower part) is synthesized as a single polypeptide chain comprising a
mature protein of 432 amino acid residues and a signal peptide (shaded) of 32 amino acid residues. Many mutations of different types causing
antithrombin deficiency have been described. Shown here are examples of missense mutations leading to amino acid substitutions associated with
type I deficiency (indicated by open circles below the polypeptide chain) or type II deficiency (open, shaded, and filled circles denote type II HBS,
type II RS, and type II PE variants, respectively).

of assembled prothrombinase complexes, the rates of their According to the Human Gene Mutation Database
degradation by APC plus protein S are similar. (HGMD; www.hgmd.cf.ac.uk), there are 353 distinct muta-
Laboratory investigation of inherited APC resistance due tions reported in the SERPINC1 gene (Table 16.1). In a major-
to the FV Leiden allele can be done using both a functional ity of cases, the genetic defect is a point mutation leading
APC-resistance test and molecular biology assays. A mod- to missense, nonsense, or splice-site mutations. Other muta-
ified APC-resistance test involving dilution of the patient’s tions involve small deletions or insertions, which result in
plasma in FV-deficient plasma is highly sensitive and specific frameshift alterations that are often deleterious. Partial or
for the presence of the FV Leiden allele. whole gene deletions are relatively uncommon causes of AT
deficiency (<10% of the reported mutations). Type II RS vari-
ants are defective in protease inactivation and mutations in
Deficiency of antithrombin
Subjects with inherited AT deficiency are, with few excep-
tions, heterozygotes. It is caused by mutations in the AT gene, Table 16.1 Mutations in genes of anticoagulant proteins retrieved
SERPINC1 (GeneID 462, OMIM ID 107300, 613118), which from the HGMD database (www.hgmd.cf.ac.uk), November 2016
is located at chromosome 1q23–q25. Heterozygous AT defi-
ciency is found in 0.02–0.05% of the general population and Mutation type SERPINC1 PROC PROS1
in 1–2% of thrombosis patients, suggesting that the genetic
defect is associated with at least a 10-fold increased risk of Missense/nonsense 196 (56%) 276 (74%) 249 (61%)
thrombosis – somewhat higher than estimated for APC resis- Splicing 25 (7%) 31 (8%) 44 (11%)
tance. AT deficiency may be of either type I or type II. Type Regulatory 1 (0.3%) 14 (4%) 4 (1%)
I deficiency is characterized by low levels of both immuno- Small deletions 65 (18%) 29 (8%) 49 (12%)
Small insertions 29 (8%) 16 (4%) 24 (6%)
logical and functional AT, whereas type II denotes functional
Small indels 3 (0.8%) 4 (1%) 5 (1%)
defects. Type II cases can be further divided into three sub-
Gross deletions 28 (8%) 3 (0.8%) 25 (6%)
types: RS (reactive site mutants), HBS (heparin binding site Gross insertions/duplications 2 (0.6%) 0 (0%) 7 (2%)
mutants), or PE (mutants giving pleiotropic effects). A large Complex rearrangements 4 (1%) 0 (0%) 2 (0.5%)
number of AT deficiencies are caused by missense mutations, Repeat variations 0 (0%) 0 (0%) 0 (0%)
which alter key amino acid residues in the primary structure Total no. of mutations 353 373 409
of the protein (Figure 16.6).
214 Molecular Hematology

the vicinity of the reactive site have been found. The type II deficiency in the population is estimated to be approximately
HBS deficiency carries mutations in the heparin-binding site, 0.3%. The relative risk of venous thrombosis associated with
and type II PE variants are caused by a limited number of protein C deficiency is similar to or slightly higher than that
mutations between amino acids 402 and 429. of FV Leiden. Protein C deficiency is not a risk factor for
arterial thrombosis, although there are studies claiming that
protein C deficiency may have an impact on the onset of arte-
Protein C deficiency rial occlusive diseases. Homozygous or compound heterozy-
Protein C deficiency is caused by mutations in the protein gous protein C deficiency is a rare condition (1 in 200 000 to
C gene, PROC (GeneID 5624, OMIM ID 176860, 612304), 1 in 400 000) that often leads to severe and fatal thrombosis
which is located on chromosome 2q13–q21. Clinically, there in the neonatal period. The clinical picture is that of purpura
are two types of protein C deficiency. Type I is characterized fulminans and the symptoms include necrotic skin lesions
by a parallel reduction in protein C antigen and functional due to microvascular thrombosis. Other major symptoms
activity. Type II is characterized by a functional defect in the are thrombosis in the brain and disseminated intravascular
protein, and its plasma concentration may be normal. The coagulation. Several cases have been successfully treated
majority of reported cases of protein C deficiency are of type with fresh frozen plasma or with protein C concentrates.
I. However, plasma assays can sometimes have limited dis- Genetic analysis has been performed in a large number
criminative power between the different types. There is also of cases with protein C deficiency and there are 373 muta-
an overlap between plasma concentrations at the low end of tions in the HGMD associated with protein C deficiency
the normal distribution and at the upper end of protein C (Table 16.1). Most genetic defects are missense mutations,
deficiency, which can complicate correct phenotyping. which lead to single amino acid substitutions, or nonsense
Heterozygous deficiency of protein C is identified in mutations located within the coding region of the gene (Fig-
2–5% of thrombosis patients. The prevalence of protein C ure 16.7). Missense mutations causing type I deficiency are

0 2 4 6 8 10 kb

Exon 1 2 3 45 6 7 8 9

mRNA

200 bp

Type II
GLA EGF1 EGF2 AP Serine protease domain

Type I

100 aa
Fig. 16.7 Structure of the human protein C gene and location of detrimental missense mutations in the protein C molecule. Human
protein C is encoded by the PROC gene, localized to chromosome 2q13–q14, which spans approximately 11 kb of DNA (upper part). The gene
comprises nine exons, which yield about a 1.8-kb mRNA transcript (middle part). The protein C mRNA encodes a pre-pro-protein C sequence of
461 amino acid residues (lower part). The pre-sequence (dark shading) serves as a signal peptide and the pro-sequence (light shading) functions as
a signal for proper γ-carboxylation of the protein. The mature protein consists of 419 residues and can be divided into a γ-carboxyglutamic acid
(GLA) domain, two epidermal growth factor (EGF) domains, and a serine protease domain. During processing of the protein, an internal dipeptide
is removed from the protein and the mature protein circulates as a covalently linked two-chain molecule. Between the second EGF domain and the
protease part of the molecule is an activation peptide (AP) region, which is released on protein C activation. Shown here are examples of missense
mutations resulting in amino acid substitutions associated with type I deficiency (indicated below the polypeptide chain) or type II deficiency (above
the polypeptide chain).
Molecular coagulation and thrombophilia 215

scattered over the entire polypeptide chain. Mutations lead- a similar picture to homozygous protein C deficiency, with
ing to type II deficiency are less common, but have also been purpura fulminans in the neonatal period.
found in almost all the modules of protein C, including the The PROS1 database, published by the International
propeptide, the Gla module, EGF1, the activation peptide Society on Thrombosis and Haemostasis in 2000, contained
(AP), and the serine protease domain. Mutations in the pro- more than 200 entries, in which 131 different mutations were
moter region of the gene, which affect the plasma protein considered to be detrimental. In the HGMD there are 409 dif-
concentration, and mutations affecting RNA splicing have ferent PROS1 mutations that may be associated with a protein
also been found. S deficiency phenotype. Most of the gene defects are missense
or nonsense mutations, and mutations affecting splicing
or small insertion/deletion defects are less common (Fig-
Protein S deficiency
ure 16.8). Due to the large size of the protein S gene and the
Protein S deficiency is linked to the protein S gene, PROS1 presence of a closely linked and highly similar pseudogene,
(GeneID 5627, OMIM ID 176880, 612336, 614514). The identification of mutations is not easy. Furthermore, in many
PROS1 gene is located close to the centromere at chro- families with phenotypically established protein S deficiency,
mosome 3p11.1–q11.2. A homologous pseudogene (PROSP, PROS1 gene mutations are not found, although linkage
GeneID 5628) is located in the near vicinity and shares more between the PROS1 locus and protein S deficiency has been
than 95% similarity with PROS1 exon and intron sequences, established. The reason for the difficulties in identifying pro-
which must be taken into consideration when designing tein S gene mutations in some families may be explained by
genetic investigations of PROS1. the fact that common genetic screening techniques may miss
Phenotyping for protein S deficiency is dependent on the large deletions as a cause for quantitative protein S deficiency.
correct measurement of protein S plasma concentration in This was also shown to be the case by extensive segregation
the laboratory. This is not a simple task to perform and will analysis using a dense set of genetic single-nucleotide
be further complicated by the presence of pools of free and polymorphism (SNP) and microsatellite markers in families
C4BP-bound protein S. However, the level of free protein S where mutations in the PROS1 gene have not been found. By
discriminates better between those with and without protein this approach, three of eight investigated families could be
S deficiency than the level of total protein S. This is because explained by large and unique deletions. If this finding, of an
the concentrations of protein S and C4BPβ+ , which is the unusually high frequency of large deletions, is reproduced
protein S-binding isoform of C4BP, are equimolar in protein in other family materials, then such mutations must be con-
S–deficient individuals and most of the protein S is bound sidered a major explanatory factor for protein S deficiency.
to C4BPβ+ . Protein S deficiency with low levels of both free
and total protein S is called type I, whereas protein S defi-
Prothrombin gain-of-function mutations
ciency with low free protein S and normal total protein S has
been believed to constitute a separate genetic type (type III). A point mutation in the prothrombin gene, F2 (GeneID
However, coexistence of the two types in many protein S– 2147, OMIM ID 176930.0009), has been identified as the
deficient families demonstrates that they represent different second most common independent risk factor for venous
phenotypic variants of the same genetic disease. Mutations thrombosis. The mutation involves a G → A base substitu-
that cause functional defective protein S are referred to as tion at nucleotide position 20210, which is located in the 3′
type II deficiency. To date, very few type II deficiencies have untranslated regions of the F2 gene (Figure 16.9). Thus, the
been found, presumably related to the poor diagnostic per- point mutation does not lead to protein structure alterations.
formance of available functional protein S assays. Instead, the mutation is associated with increased plasma
Heterozygous protein S deficiency is present in 1–10% levels of prothrombin through a gain-of-function phenotype
of thrombosis patients; the wide variety may be explained that has been explained by increased efficiency of the F2 gene
by several factors such as different inclusion and exclusion mRNA. The prevalence of the mutation in the general pop-
criteria and geographical/ethnic differences between studies. ulation is 0–5%, depending on the ethnicity of the studied
The prevalence of protein S deficiency in the general pop- population, and the mutation is associated with an approx-
ulation has been estimated to be 0.03–0.13% in European imately twofold increased risk of thrombosis. Similar to the
populations, but higher figures have been reported from FV Leiden mutation, the F2 20210A variant originated from
Japanese and Thai populations. Family studies suggest that a single mutational event and the allele is only found in Cau-
heterozygous carriers have at least a fivefold increased risk casian populations.
of thrombosis compared with their healthy relatives, which Another gain-of-function mutation was originally found
is similar to the rates in thrombophilic families with protein in a Japanese family with hereditary thrombosis. The muta-
C deficiency and APC resistance (FV Leiden). Homozygous tion is located in the last exon of the F2 gene and leads to a
protein S deficiency is extremely rare, but appears to give substitution of arginine for leucine at position 596 (OMIM
216 Molecular Hematology

0 50 60 70 80 90 100 kb

Exon 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15

mRNA

200 bp

Type II

GLA TSR EGF1EGF2 EGF3 EGF4 SHBG

Type I

100 aa
Fig. 16.8 Structure of the human protein S gene and location of detrimental missense mutations in the protein S molecule. The gene
for human protein S (PROS1) comprises 15 exons (upper part) and spans 100 kb of DNA and is localized to chromosome 3p11.1–q11.2. Exons are
denoted by open bars and introns by lines. Introns denoted by dashed lines between exons indicate gaps and are not drawn to scale. The PROS1
mRNA is approximately 3.5 kb in size (middle part). The mRNA is translated into a pre-pro-protein S of 676 amino acid residues (lower part). The
polypeptide chain can be divided into a signal peptide (dark shading), a pro-peptide (light shading), a thrombin sensitive region (TSR), a
γ-carboxyglutamic acid (GLA) domain, four epidermal growth factor (EGF)-like domains, and a large carboxy-terminal domain homologous to sex
hormone–binding globulin (SHBG). Shown here are examples of missense mutations resulting in amino acid substitutions associated with type I
deficiency (indicated below the polypeptide chain) or type II deficiency (above the polypeptide chain).

1 kb

2 4 6 8 10 12 14
Exon 1 3 5 7 9 11 13

13 14

3’ UT

A
ATGGTTCCCAATAAAAGTGACTCTCAGC AGCCTC
G
Fig. 16.9 Structure of the human prothrombin gene and location of the prothrombin 20210 G → A mutation. The human gene for
prothrombin (F2) comprises 14 exons and spans approximately 20 kb of DNA on chromosome 11p11–q12. The nucleotide sequence flanking the
G → A transition at nucleotide 20210 (indicated in bold) in the 3′ untranslated region of the F2 gene is shown at the bottom. The putative
polyadenylation signal is boxed. The 20210A allele represents a gain-of-function mutation associated with elevated levels of plasma prothrombin
and an increased risk of venous thrombosis.
Molecular coagulation and thrombophilia 217

100

80 FV Leiden
60

Free of thrombosis (%)


Fig. 16.10 Thrombosis-free survival curves for individuals 40 Protein S Protein S
with different FV genotypes and coinherited protein S deficiency deficiency
20
deficiency. (a) The increased risk of thrombosis with combined +FV Leiden
0 (a)
defects is illustrated with thrombosis-free survival curves for 21
individuals with single defects, FV Leiden or protein S deficiency, 100
FV: normal
and 18 individuals with both defects. There was no significant 80
difference between those with single defects, whereas differences
60 FV Leiden:
between individuals with the FV Leiden allele or protein S heterozygous
deficiency and those with combined defects were significant. (b) 40
Probability of being free of thrombotic events at a certain age for FV Leiden: homozygous
146 normal individuals, 144 heterozygotes and 18 homozygotes 20
for the FV Leiden allele (Kaplan–Meier analysis). Highly significant 0
differences were observed between normals and heterozygotes 0 20 40 60 80 100
and between heterozygotes and homozygotes. Age (years) (b)

ID 176930.0015). This arginine residue 596 is located in Leiden are expected to be present in between 1 in 3000 and 1
a highly conserved region important for the coordination in 10 000 individuals. A similar calculation for the combina-
of sodium and contains one of the antithrombin-binding tion of the prothrombin mutation and FV Leiden allele sug-
sites in the prothrombin molecule. It is anticipated that the gests the prevalence of combined defects to be 1–2 per 1000
mutation conveys antithrombin resistance by impairment of individuals. Thus, a large number of people carry more than
the thrombin–antithrombin complex formation and thereby one genetic defect and such individuals have a considerably
leads to an increased risk of thrombosis. In recent years, other increased risk of thrombosis. The FV Leiden allele is thus
mutations affecting the same amino acid residue (Arg596Gln found to be an additional genetic risk factor in certain throm-
and Arg596Trp) have been associated with venous throm- bophilic individuals with a deficiency of protein C, protein S,
boembolism and a laboratory phenotype of antithrombin or AT as well as in cases with the prothrombin mutation (Fig-
resistance. The different mutations, at the same codon, in ure 16.10).
apparently unrelated families indicate that this may represent The thrombotic tendency in individuals with inherited
a hotspot for prothrombin mutations. However, there are so genetic defects is highly variable and some individuals never
far only a handful of reports of antithrombin resistance and develop thrombosis, whereas others develop recurrent severe
the relative frequency of this hereditary risk factor has yet to thrombotic events at an early age. This depends on the par-
be determined. ticular genotype, the coexistence of other genetic defects, and
the presence of environmental risk factors such as oral con-
traceptives, trauma, surgery, and pregnancy. Thus, women
Severe thrombophilia is a multigenic with hetero- or homozygosity for the FV Leiden allele who
disease also use oral contraceptives have a considerably increased
Venous thrombosis is a typical multifactorial disease involv- risk of thrombosis. In this context it is noteworthy that the
ing one or more environmental and/or genetic risk factors. World Health Organization (WHO) recommends that com-
In Western societies, many individuals carry more than one bined hormonal contraceptives should be avoided in women
genetic risk factor because the FV Leiden allele is so common. with inherited thrombophilia.
In contrast, in countries where the FV Leiden allele is rare,
few individuals carry more than one genetic defect. This may
explain the difference in incidence of thromboembolic dis- Management of thrombophilia
ease between Japan and China on the one hand and Europe
and the USA on the other. The frequency of individuals car- Decisions about medical intervention due to the presence
rying two or more genetic defects can be calculated on the of one or more genetic defects should be based on careful
basis of the prevalence of the individual genetic defects in the consideration of the clinical picture, including the patient’s
general population. In a country where the prevalence of FV family history. The impact of the medical history on the
Leiden is 10%, combinations of protein C deficiency and FV use of oral anticoagulants is perhaps even more important
218 Molecular Hematology

in individuals with APC resistance (FV Leiden) or the gene (G20210A) is another common prothrombotic risk
prothrombin mutation than in those with the rarer defi- factor, with a prevalence of approximately 2% in the general
ciencies of protein C, protein S, or AT. The risk of bleeding population. Other less common independent genetic risk
complications due to anticoagulant therapy must always be factors include abnormalities in the genes for AT, protein
weighed against the benefits of anticoagulation, especially if C, and protein S. Families with thrombophilia present with
an oral anticoagulant is used for periods exceeding three to variable penetrance of thrombosis explained by different
six months, when the risk of thrombotic recurrence probably combinations of genetic defects and environmental risk
declines. New clinical data are continually emerging and no factors. Patients with combined genetic defects are at higher
general consensus regarding screening, prophylaxis, and the risk of thrombosis than those with single-gene defects. Thus,
treatment of symptomatic patients has yet been established. evaluation of patients with thrombosis must be performed in
When the FV Leiden allele is present in homozygous form, order to fully estimate the risk for thrombosis in each case.
or heterozygosity is combined with a second genetic defect,
prophylactic treatment with heparin or oral anticoagulants
is recommended in situations known to be associated with a Further reading
high risk of thromboembolic complications, such as surgery
or pregnancy, even if the patient has never experienced any Blood coagulation: introduction and
thrombosis or has no family history of such complications. regulation
For heterozygous asymptomatic carriers lacking a family his-
tory of thrombosis, short-term prophylaxis has been recom- Camire, R.M. (2016). Rethinking events in the haemostatic process: role
of factor V and TFPI. Haemophilia 22 (Suppl 5): 3–8.
mended in high-risk situations, but it remains to be estab-
Dahlbäck, B. (2005). Blood coagulation and its regulation by anticoagu-
lished whether prophylaxis should be given in all situations
lant pathways: genetic pathogenesis of bleeding and thrombotic dis-
associated with a risk of thrombosis. eases. J. Int. Med. 257: 209–223.
Symptomatic heterozygous patients should be managed in Dahlbäck, B. (2016). Pro- and anticoagulant properties of factor V in
the same way as any other patient with thrombotic events pathogenesis of thrombosis and bleeding disorders. Int. J. Lab. Hema-
until more specific recommendations are established. It is not tol. 38 (Suppl 1): 4–11.
known whether the presence of a genetic defect is associ- Dahlbäck, B. and Villoutreix, B.O. (2005). Regulation of blood coagu-
ated with an increased risk of recurrence, even though most lation by the protein C anticoagulant pathway: novel insights into
studies on APC resistance tend to suggest that this is indeed structure–function relationships and molecular recognition. Arte-
the case. Patients with combined defects, and probably also rioscler. Thromb. Vasc. Biol. 25: 1311–1320.
patients with single-gene defects, may be at increased risk of Esmon, C.T. (2014). Targeting factor Xa and thrombin: impact on coag-
ulation and beyond. Thromb. Haemost. 111: 625–633.
recurrence and should accordingly be given long-term anti-
Furie, B. and Furie, B.C. (2005). Thrombus formation in vivo. J. Clin.
coagulation therapy beyond six months, even after an iso-
Invest. 115: 3355–3362.
lated thromboembolic event. However, more data are needed Long, A.T., Kenne, E., Jung, R. et al. (2016). Contact system revisited: an
before these recommendations can be considered generally interface between inflammation, coagulation, and innate immunity.
applicable. J. Thromb. Haemost. 14: 427–437.
The potential benefits of general screening for APC resis- Mann, K.G., Brummel-Ziedins, K., Orfeo, T., and Butenas, S. (2006).
tance and/or the FV Leiden allele prior to thrombotic events Models of blood coagulation. Blood Cells Mol. Dis. 36: 108–117.
or in the presence of such circumstantial factors as oral con- Morrissey, J.H. and Smith, S.A. (2015). Polyphosphate as modulator of
traceptive use, pregnancy, and surgery are obvious, but more hemostasis, thrombosis and inflammation. J. Thromb. Haemost. 13
prospective data are needed, not least in terms of cost–benefit (Suppl 1): S92–S97.
ratios, before any general recommendations can be made. Rau, J.C., Beaulieu, L.M., Huntington, J.A., and Church, F.C. (2007). Ser-
pins in thrombosis, hemostasis and fibrinolysis. J. Thromb. Haemost.
5 (Suppl 1): 102–115.
Vincent, L.M., Tran, S., Livaja, R. et al. (2013). Coagulation factor V
Conclusions (A2440G) causes East Texas bleeding disorder via TFPIα. J. Clin.
Invest. 123: 3777–3787.
Inherited APC resistance, caused by the Arg506Gln muta-
tion in the FV gene (FV Leiden), is the most common genetic
risk factor for thrombosis identified to date. The mutated Molecular genetics of venous
FV has normal procoagulant properties, but the loss of the thromboembolism, APC resistance,
APC cleavage site at position 506 in FV results in impaired and FV leiden
regulation of coagulation and a hypercoagulable state. The Bertina, R.M., Koeleman, B.P., Koster, T. et al. (1994). Mutation in blood
prevalence of FV Leiden in Caucasian populations varies coagulation factor V associated with resistance to activated protein C.
between 2% and 15%. A genetic variant in the prothrombin Nature 369: 64–67.
Molecular coagulation and thrombophilia 219

Bezemer, I.D., Bare, L.A., Doggen, C.J.M. et al. (2008). Gene variants the risk of thrombosis in families with inherited antithrombin defi-
associated with deep venous thrombosis. J. Am. Med. Assoc. 299: ciency. Blood 94: 2590–2594.
1306–1314.
Castoldi, E. and Rosing, J. (2010). APC resistance: biological basis and
Protein C system and protein C deficiency
acquired influences. J. Thromb. Haemost. 8: 445–453.
Dahlbäck, B., Carlsson, M., and Svensson, P.J. (1993). Familial throm- Dahlbäck, B. and Villoutreix, B.O. (2005). Regulation of blood coagu-
bophilia due to a previously unrecognized mechanism characterized lation by the protein C anticoagulant pathway: novel insights into
by poor anticoagulant response to activated protein C: prediction of a structure–function relationships and molecular recognition. Arte-
cofactor to activated protein C. Proc. Natl. Acad. Sci. U.S.A. 90: 1004– rioscler. Thromb. Vasc. Biol. 25: 1311–1320.
1008. Esmon, C.T. (2012). Protein C anticoagulant system – anti-
Greengard, J.S., Sun, X., Xu, X. et al. (1994). Activated protein C resis- inflammatory effects. Semin. Immunopathol. 34: 127–132.
tance caused by Arg506Gln mutation in factor Va. Lancet 343: 1361– Griffin, J.H., Zlokovic, B.V., and Mosnier, L.O. (2015). Activated protein
1362. C: biased for translation. Blood 127: 2898–2907.
Griffin, J.H., Evatt, B., Wideman, C., and Fernandez, J.A. (1993). Anti- Segers, K., Dahlbäck, B., and Nicolaes, G.A. (2007). Coagulation fac-
coagulant protein C pathway defective in majority of thrombophilic tor V and thrombophilia: background and mechanisms. Thromb.
patients. Blood 82: 1989–1993. Haemost. 98: 530–542.
Heit, J.A. (2008). The epidemiology of venous thromboembolism in the
community. Arterioscler. Thromb. Vasc. Biol. 28: 370–372.
Protein S and protein S deficiency
Koster, T., Rosendaal, F.R., de Ronde, H. et al. (1993). Venous thrombo-
sis due to poor anticoagulant response to activated protein C: Leiden Dahlbäck, B. (2007). The tale of protein S and C4b-binding protein, a
Thrombophilia Study. Lancet 342: 1503–1506. story of affection. Thromb. Haemost. 98: 90–96.
Lindqvist, P.G., Svensson, P.J., Dahlbäck, B., and Marsal, K. (1998). Fac- Gandrille, S., Borgel, D., Sala, N. et al. (2000). Protein S deficiency: a data
tor V R506Q mutation (activated protein C resistance) associated base of mutations. Summary of the first update. Thromb. Haemost. 84:
with reduced intrapartum blood loss: a possible evolutionary selec- 918–934.
tion mechanism. Thromb. Haemost. 79: 69–73. Garcia de Frutos, G., Fuentes-Prior, P., Hurtado, B., and Sala, N. (2007).
Nogami, K., Shinozawa, K., Ogiwara, K. et al. (2014). Novel FV nutation Molecular basis of protein S deficiency. Thromb. Haemost. 98: 543–
(W1920R, FVNara) associated with serious deep vein thrombosis and 556.
more potent APC resistance relative to FVLeiden. Blood 123: 2420– Johansson, A.M., Hillarp, A., Säll, T. et al. (2005). Large deletions of the
2428. PROS1 gene in a large fraction of mutation negative patients with
Pezeshkpoor, B., Castoldi, E., Mahler, A. et al. (2016). Identification protein S deficiency. Thromb. Haemost. 94: 951–957.
and functional characterization of a novel F5 mutation (Ala512Val, Peraramelli, S., Rosing, J., and Hackeng, T.M. (2012). TFPI-dependent
FVBonn) associated with activated protein C resistance. J. Thromb. activities of protein S. Thromb. Res. 129 (Suppl 2): S23–S26.
Haemost. 14: 1353–1363.
Svensson, P.J. and Dahlbäck, B. (1994). Resistance to activated protein
Prothrombin gene mutations
C as a basis for venous thrombosis. N. Engl. J. Med. 330: 517–522.
Vandenbroucke, J.P., Koster, T., Briet, E. et al. (1994). Increased risk of Bulato, C., Radu, C.M., Campello, E. et al. (2016). New prothrombin
venous thrombosis in oral-contraceptive users who are carriers of mutation (Arg596Trp, prothrombin Padua 2) associated with venous
factor V Leiden mutation. Lancet 344: 1453–1457. thromboembolism. Arterioscler. Thromb. Vasc. Biol. 36: 1022–1029.
Voorberg, J., Roelse, J., Koopman, R. et al. (1994). Association of Danckwardt, S., Hartmann, K., Gehring, N.H. et al. (2006). 3′ End pro-
idiopathic venous thromboembolism with single point-mutation at cessing of the prothrombin mRNA in thrombophilia. Acta Haematol.
Arg506 of factor V. Lancet 343: 1535–1536. 115: 192–197.
Dziadosz, M. and Baxi, L.V. (2016). Global prevalence of prothrombin
Antithrombin deficiency gene mutation G20210A and implications in women’s health: a sys-
tematic review. Blood Coagul. Fibrinolysis 27: 481–489.
Lane, D.A., Bayston, T., Olds, R.J. et al. (1997). Antithrombin mutation Gehring, N.H., Frede, U., Neu-Yilik, G. et al. (2001). Increased efficiency
database: 2nd (1997) update. Thromb. Haemost. 77: 197–211. of mRNA 3′ end formation: a new genetic mechanism contributing
Perry, D.J. and Carrell, R.W. (1996). Molecular genetics of antithrombin to hereditary thrombophilia. Nat. Genet. 28: 389–392.
deficiency. Hum. Mut. 7: 7–22. Miyawaki, Y., Suzuki, A., Fujita, J. et al. (2012). Thrombosis from a
Picard, V., Nowak-Göttl, U., Biron-Andreani, C. et al. (2006). Molecular prothrombin mutation conveying antithrombin resistance. N. Engl.
bases of antithrombin deficiency: twenty-two novel mutations in the J. Med. 366: 2390–2396.
antithrombin gene. Hum. Mut. 27: 600. [Erratum in Human Mutation Poort, S.R., Rosendaal, F.R., Reitsma, P.H., and Bertina, R.M. (1996). A
2006, 27, 1160.]. common genetic variation in the 3′ -untranslated region of the pro-
Van Boven, H.H., Vandenbroucke, J.P., Briet, E., and Rosendaal, F.R. thrombin gene is associated with elevated plasma prothrombin levels
(1999). Gene–gene and gene–environment interactions determine and an increase in venous thrombosis. Blood 88: 3698–3703.
Chapter 17 The molecular basis of hemophilia
Daniel P. Hart1 & Paul L.F. Giangrande2
1 The Royal London Hospital Haemophilia Centre, Barts and The London School of Medicine and Dentistry, QMUL London, UK
2
Oxford Haemophilia and Thrombosis Centre, Churchill Hospital, Oxford, UK

Introduction: clinical features of hemophilia, 221 Inhibitor formation: etiology and clinical implications, 226
Inheritance of hemophilia, 223 Therapeutic applications of molecular biology to patient care, 228
Molecular basis of hemophilia A, 223 Conclusions, 232
Molecular basis of hemophilia B, 225 Further reading, 232

on the anterior thigh, weakness, and wasting of leg muscles


Introduction: clinical features (Figure 17.3). Bleeding from the gastrointestinal tract
of hemophilia (melena) and bleeding into the urinary tract (hematuria)
may also occur. There is also a significant risk of intracranial
The clinical severity (bleed phenotype) is critically deter-
hemorrhage in severe hemophilia, which in the past was a sig-
mined by the concentration of circulating factor VIII (or IX)
nificant cause of mortality when treatment was not so readily
in the blood, and severe hemophilia is defined by a clotting
available. Higher levels of factor VIII (or IX) above 5 IU dl−1
factor concentration below 1 IU dl−1 (Table 17.1). The
are associated with a milder form of the disease, with no
hallmark of severe hemophilia, in the absence of treatment,
spontaneous joint bleeds, but a definite risk of bleeding
is recurrent and spontaneous hemarthrosis. Typically, hinge
after even relatively minor injury or procedures (e.g. dental
joints such as the knees, elbows, and ankles are affected, but
extraction).
bleeds may also occur in the wrist or shoulder. Bleeding into
Treatment of bleeding episodes involves the intravenous
the hip joint is unusual. The acutely affected joint becomes
injection of coagulation factor concentrates (CFCs); the total
swollen and warm, and is held in a position of flexion (Fig-
dose and frequency of treatment will also be determined by
ure 17.1), with no external discoloration or bruising around
the severity and site of bleeding. Preventive infusion of CFC
the joint. It is unusual for an infant to suffer spontaneous
is the gold standard of care (prophylaxis therapy). Patients
hemarthroses in the first few months of life, and the first
regularly inject CFC intravenously (most commonly two to
joint to be affected tends to be the ankle as the child learns
four times a week) to prevent bleeds, rather than just treating
to crawl. The first sign of a hemarthrosis in an infant will
on demand after bleeds or an injury has occurred. The dose
often be obvious discomfort and distress, accompanied by
and frequency of prophylaxis infusions are determined by the
limping or reluctance to use a limb. Recurrent bleeds into a
half-life of the chosen CFC and increasingly after identify-
joint lead to synovitis and joint damage, ultimately resulting
ing the pharmacokinetics (PK) of that CFC in the candidate
in crippling arthritis (Figure 17.2). Bleeding into muscles is
patient. Prophylaxis aims to maintain a measurable plasma
also a feature of hemophilia, but this is usually a consequence
level of clotting factor activity above the patient’s baseline
of direct injury, albeit often minor (Figure 17.3). Bleeds into
(trough level) just prior to the next CFC prophylaxis dose.
certain areas are particularly dangerous because of the risk of
Patients on prophylactic therapy experience few or even no
compression of neighboring structures, manifesting then as a
spontaneous bleeds, and thus progressive joint damage and
compartment syndrome. Patients with inhibitory antibodies
arthritis can be minimized or avoided. The quality of life of
are particularly at risk in this regard, as bleeds may be more
patients on prophylaxis may be greatly enhanced, allowing
difficult to control. Bleeds in the tongue can obstruct the air-
them to lead much more independent lives.
way, and retroperitoneal bleeding within the iliopsoas muscle
Approximately 30% of patients with severe hemophilia A
may result in femoral nerve compression, causing paresthesia
can be expected to develop inhibitory antibodies to factor
VIII CFC at some stage. In contrast, inhibitor development
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben.
in hemophilia B is very rare and encountered in less than
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. 5% of patients. The development of such antibodies poses

221
222 Molecular Hematology

Table 17.1 The relation of blood levels of factor VIII (or IX) to the severity of hemorrhagic manifestations

Level (IU dl−1 ) Hemorrhagic manifestations

50–150 Normal level, no bleeding problems


25–50 No problems in day-to-day life. Tendency to bleed after major surgery
5–25 Mild hemophilia. Bleeding typically occurs only after significant injuries
2–5 Moderately severe hemophilia: spectrum of bleed phenotype – some with spontaneous bleeds and early arthropathy,
others experiencing bleeds associated with injury, albeit often relatively minor
<1 Severe hemophilia with spontaneous and recurrent bleeding into muscles and joints. Risk of intracranial hemorrhage.

Fig. 17.2 Radiograph of the knee of a patient with severe


hemophilic arthropathy. Joint replacement surgery was
Fig. 17.1 Acute hemarthrosis in severe hemophilia. This usually subsequently carried out in this case.
arises in the absence of injury. The joints most frequently involved are
the knees, elbows, and ankles. The joint is swollen, warm, and tender,
but there is no external bruising or discoloration.

considerable problems in treatment, as these immunoglob-


ulins (IgG) are capable of rapidly inactivating infused factor
VIII, and furthermore the antibody titer may rise dramat-
ically after a course of factor VIII (anamnesis). Very occa-
sionally, acquired hemophilia A may arise in a previously
normal individual due to the formation of autoantibodies
directed against factor VIII, and both males and females may
be affected. Hemarthrosis is unusual in acquired hemophilia,
and the principal manifestations are usually extensive super-
ficial purpura and muscle bleeds. Acquired hemophilia Fig. 17.3 Magnetic resonance imaging (MRI) scan showing
arises most often in the elderly, associated with underly- bilateral iliopsoas hemorrhage. This bleed was associated with a
ing malignant or autoimmune diseases in approximately complete but transient paralysis in both legs, as the femoral nerve is
half of cases, with a small, younger cohort associated with located on the anterior surface of the muscle and may be compressed
pregnancy. in such cases.
The molecular basis of hemophilia 223

Turner syndrome (XO karyotype) and androgen insensitivity


Inheritance of hemophilia syndrome (XY karyotype). A proportion of females carrying
a hemophilia gene will have factor activity levels, from that
The genes for factors VIII and IX (F8 and F9, respectively) are
corresponding gene, below recognized laboratory normal
both located at the telomeric end of the X chromosome and
ranges through a process called skewed lyonization. These
thus hemophilia is inherited as an X-linked recessive condi-
girls/women effectively have mild hemophilia and should be
tion (Figure 17.4). The daughters of affected males are obli-
identified and offered care commensurate with their levels,
gate carriers, but the sons are normal. The phenotype remains
as would be offered to boys/men with equivalent factor lev-
constant within a family, so the daughter of a man with only
els. The remaining carrier women have normal laboratory
mild hemophilia may be reassured that she will not pass on a
range factor activity in their plasma. Importantly, normal
severe form of the condition. However, up to a half of all cases
plasma factor activity cannot be used as a surrogate marker
of hemophilia arise in the absence of a previous family history
of whether a daughter of a carrier mother is a carrier her-
and are due to a new mutation. The most famous example is
self. Genetic testing will always be required to clarify true
that of Queen Victoria, who had a hemophilic son (Leopold)
carriership.
and two daughters (Alice and Beatrice) who turned out to be
carriers. Forensic genetic testing on exhumed remains of the
assassinated Russian royal family has since proven the royal
hemophilia to be severe hemophilia B. There are instances of Molecular basis of hemophilia A
hemophilia affecting females due to inheritance of the defec-
tive gene from both parents (see Figure 17.4, generation IV), Factor VIII is an essential cofactor for the activation of fac-
and there are also case reports of hemophilia in females with tor X by activated factor IX (see Chapter 16). Factor VIII

1 2
I

II 1 2 3 4 5 6

III 1 2 3 4 5 6 7 8 9

IV 1 2 3 4

Normal Female Homozygous Normal Hemophilic


female carrier female male male

Fig. 17.4 The inheritance of hemophilia. The genes for factors VIII and IX are both encoded on the X chromosome, and inheritance is thus sex
linked. The daughter of a man with hemophilia is an obligate carrier, but the son of a father living with hemophilia will not be affected.
Hemophilia may thus be transmitted to a grandson via a carrier daughter. Color blindness and Duchenne muscular dystrophy are other examples
of conditions that are X linked.
224 Molecular Hematology

X chromosome
q28

Fig. 17.5 The factor VIII gene. The factor VIII gene
(F8) was cloned in 1984 and is located toward the
G6PD telomeric end of the long arm of the X chromosome
qter 100 kb Factor VIII
(Xq28). It maps distal to the gene encoding glucose
6-phosphate dehydrogenase (G6PD), about 1 Mb
from the Xq telomere. The gene is composed of 26
10 kb
1 2 3 4 5 6 7 8 9 101112 13 14 15 16 17 18 19 20 21 22 23 24 25 26
exons, of which exon 14 is the largest. The large
intron 22 contains a nested intronless gene termed
5' 3' F8A (the function of the gene and its transcript are
unknown). There are two further copies of F8A
located about 400 kb telomeric to the factor VIII
gene. The spliced factor VIII mRNA is ∼9 kb in length
Exon 22 Exon 23
and the mature factor VIII molecule is composed of
F8A F8B 2332 amino acids.

must itself undergo proteolytic cleavage at two distinct sites encountered in as many as 45% of people with severe
through the action of thrombin before it becomes physiolog- hemophilia A in all ethnic groups (Figure 17.6). The inver-
ically active. It circulates in plasma as a large glycoprotein sion mechanism involves an intronless gene of unknown
bound non-covalently to the larger protein, von Willebrand function, designated F8A. Two copies of this gene are located
factor (VWF). The factor VIII gene (F8) was first cloned in near the tip of the X chromosome and there is another copy
1984. It is 186 kb in length and is situated on the long arm within intron 22 of the factor VIII gene itself. During meio-
of the X chromosome at Xq28 (Figure 17.5). The factor VIII sis, either of the two telomeric copies may cross over with
gene consists of 26 exons, which range in size from 69 bp the intronic copy, resulting in a division of the gene into
(exon 5) to 3.1 kb (exon 14). The factor VIII message is nearly two halves facing in opposite directions and separated by
9 kb in size and encodes a mature protein of 2332 amino approximately 500 kb. Crossover with the distal copy is much
acids. Approximately half of all cases of severe hemophilia more common than crossover with the proximal copy, and
and all cases of mild and moderate hemophilia result from accounts for approximately 80% of all inversions. It is now
heterogeneous mutations that occur throughout the F8 gene. recognized that inversion almost always forms during a male
By far the commonest single genetic defect causing meiosis. It is believed that the presence of a large region of
severe hemophilia is an inversion in intron 22, which is non-homology between the X and Y chromosomes during

3' tel 3'


FVIII mRNA3'
F8B
FVIII F8A
cen F8B
5' F8A
F8A

5' mRNA5'
q28 F8A FVIII
F8A F8A

tel tel tel


Fig. 17.6 “Flip tip inversion.” This unique inversion of the tip of the X chromosome is now recognized to be responsible for about half of all
cases of severe hemophilia A in all ethnic groups. Crossover occurs between a copy of F8A within the F8 gene and one of the two telomeric
copies. Crossover with the distal copy is more common, occurring in approximately 80% of cases where an inversion is identified.
The molecular basis of hemophilia 225

meiotic pairing may favor a misalignment, and the presence molecules devoid of any functional activity. Frameshift muta-
of a second X chromosome with a complementary region tions resulting from insertions or small deletions have also
may act as a stabilizing factor. An important clinical conse- been identified as a cause of severe hemophilia. Duplications
quence of this observation is that when an apparently new within one or more exons of the F8 gene have recently been
and spontaneous case of hemophilia is diagnosed in which identified in a small minority of patients with hemophilia
the gene inversion is identified, it is likely that the defect arose A using multiplex ligation-dependent probe amplification
in the maternal grandfather’s allele (during spermatogene- (MLPA). A full list of mutations described in association with
sis), and thus the mother can generally be assumed to be a hemophilia is outside the scope of this chapter, but addi-
carrier and at risk of having another affected male child. The tional information is provided in the Further Reading section
resulting truncated protein product is presumably unstable, or via the F8 variant database (http://www.factorviii-db.org).
resulting in severe hemophilia. The inversion is not found in Knowledge of an individual’s F8 mutation has relevance to
individuals with non-severe forms of hemophilia. The inver- predicting inhibitor risk: the more severe the mutation and
sion is easily detected and the identification of this defect the resultant absence of translated protein, the higher the
as the commonest cause of severe hemophilia has simpli- risk. However, even single amino acid differences of mis-
fied both screening for carriers and antenatal diagnosis of sense mutations can still provoke an alloimmune response,
hemophilia. The inversion may be identified with Southern resulting in clinically impactful rejection antibody formation
blotting, although many laboratories now use either inverse (inhibitor).
polymerase chain reaction (PCR) or long-range PCR for Approximately 40% of all missense mutations arise at
identification of this inversion. More recently, inversions in CG dinucleotide sites, resulting in a change to TG or
intron 1 of the F8 gene have been identified as a cause of CA sequences. It is generally believed that CG nucleotides
severe hemophilia and this abnormality appears to be respon- represent genomic hotspots. Cytosine is predominantly
sible for approximately 5% of all cases of severe hemophilia A. methylated in human DNA, but this is relatively unstable
Since approximately half of all cases of severe hemophilia are and 5-methylcytosine is prone to spontaneous deamination
associated with these two inversions, it is usual practice to to yield a GT mismatch that is inefficiently repaired. It is also
screen samples from new cases for these two abnormalities of interest that a missense mutation may be associated with
first. varying degrees of clinical severity. Thus, a C → T mutation
Developments in molecular biology have permitted more at nucleotide 1689 within exon 14 resulting in replacement
rapid identification of defects in hemophilia. Southern blot- of arginine by cysteine has been reported in association with
ting has been superseded by methods involving PCR ampli- both severe and mild clinical phenotypes.
fication of either patient DNA or material derived from
the reverse transcription of mRNA (RT-PCR). Automated
sequence analyzers enabling sequencing of the entire F8 Molecular basis of hemophilia B
gene are now more affordable. Where not available, pre-
vious methods may still be relevant to identify restricted The factor IX gene (F9) is also located on the long arm
areas of abnormal DNA in patients with hemophilia, which of the X chromosome at band Xq27, and is encoded by a
may then be targeted for specific attention. These meth- stretch of DNA spanning 33.5 kb that contains eight exons
ods include amplification and mismatch detection (AMD), (Figure 17.7). The basic structure of the gene is similar in
conformation-sensitive gel electrophoresis (CSGE), denatur- organization to that of protein C and coagulation factors
ing gradient gel electrophoresis (DGGE), high-resolution VII and X, and it is likely that they all originated in the
melting analysis (HRM), and pyrosequencing. Approxi- distant past from a common ancestral gene by duplication.
mately 4% of cases of hemophilia are the consequence of gene Factor IX mRNA comprises 2.8 kb and encodes a mature
deletions, which have been reported throughout the gene protein of 415 amino acids. This is made up of a glutamic
and which are very variable in size. As with the intron 22 acid–rich sequence (Gla domain) and two epidermal growth
inversion, most deletions are associated with a severe clin- factor (EGF)-like domains separated from the serine pro-
ical phenotype. To date, around 650 different single base tease domain by an activation region. The 12 glutamic acid
substitutions have been described, of which approximately residues in the Gla domain undergo post-translational γ-
75% predict a single amino acid change from the wild-type carboxylation, which is necessary for binding of calcium,
sequence (missense). Such mutations affect RNA processing, and exon 2 encodes a recognition site for the carboxylase.
mRNA translation, or the fine structure of factor VIII itself. Exon 1 encodes the signal peptide necessary for transport
The majority of these missense F8 mutations result in non- into the endoplasmic reticulum. Exon 6 encodes the activa-
severe forms of hemophilia A. A further 100 lead to the cre- tion peptide that is cleaved off during the activation of fac-
ation of preliminary peptide chain termination or prema- tor IX by either factor XI or a complex of tissue factor and
ture stop codons and the production of truncated factor VIII factor VII. Exons 7 and 8 encode the catalytic regions of
226 Molecular Hematology

0 10 20 30 kb

1 2 3 4 5 6 7 8

Exons F.IX gene

–46 –18 +1 39 47 85 128 145 180 415


PRE PRO GLA H EGF–type B EGF–type A Activation Catalytic F.IX protein

Fig. 17.7 The factor IX gene (F9). The factor IX gene (F9) was first cloned in 1982. It is located on the long arm of the X chromosome at band
Xq27. The gene spans 34 kb and encodes eight exons (exons are shown as colored boxes, and dotted lines between the gene and protein indicate
protein domains encoded by each exon). The signal peptide and propeptide sequences are cleaved during processing and activation of factor IX.
EGF, epidermal growth factor domains; GLA, γ-carboxyglutamic acid domain.

factor IX, which are responsible for subsequent activation of that contain binding sequences for liver-enriched transcrip-
factor X in the coagulation cascade. The gene is controlled by tion factors, which are presumably influenced by androgenic
a promoter. steroids.
The F9 gene, cloned in 1982, is considerably smaller than A gain-of-function F9 mutation R338L, F9 Padua (substi-
F8. The first defects identified in hemophilia B were gross tution of leucine for arginine at position 338), described in an
deletions, detected by Southern blotting. However, it is now Italian family after a case of juvenile thrombophilia, is para-
recognized that gene deletions account for only approxi- doxically of particular interest in gene therapy strategies for
mately 3% of all cases of hemophilia B. No equivalent of severe hemophilia B. The R338L gene sequence is being used
the F8 gene inversion has been encountered in hemophilia in early-phase clinical trials to increase the detectable levels
B and it is now clear that point mutations account for the of FIX protein function to close to the normal FIX activity
vast majority of cases of hemophilia B; over 500 have been range after in vivo gene therapy.
described from families around the world. The great major- Full details of the many genetic abnormalities associated
ity involve single base changes, which have been identified in with hemophilia B can be found at the websites listed at the
all domains of the protein. In contrast to missense F8 muta- end of this chapter.
tions causing non-severe hemophilia A, inhibitor formation
in F9-missense, non-severe hemophilia B is unheard of, the
explanation for which is unknown. The unusually high fre- Inhibitor formation: etiology and
quency of mutations at CG dinucleotide sites in hemophilia clinical implications
B probably reflects the high number of CG dinucleotides at
critical sites in the F9 gene. Exon 8 is the largest at 1.9 kb in A proportion of patients with hemophilia will develop
length and half of all mutations are found in this exon. How- alloimmune immunoglobulins directed against infused fac-
ever, about 20–30% of cases of mild hemophilia B are due tor VIII (or IX) CFC after exposure to these therapeutics for
to a small number of founder mutations. The original case treatment of bleeding episodes. The individual’s immune sys-
of Christmas disease (named after the first patient, Stephen tem perceives the infused, wild-type CFC as “foreign,” given
Christmas, described in 1952) has been identified as a G → C that the individual was not born with and is thus not toler-
mutation at nucleotide 31170, resulting in substitution of cys- ant to the “normal,” wild-type protein sequence when first
teine by serine within exon 8. exposed to the infused CFC. This is potentially very serious,
Mutations in the promoter region of the F9 gene (e.g. as individuals become refractory to conventional doses of
T → A at −20 and G → A at −6) are relatively rare and CFCs and bleeding can be very difficult to control. Inhibitory
account for around 2% of all cases. However, they are of par- antibodies interfere with the normal function of factor VIII
ticular interest as they can give rise to the unique hemophilia in a number of different ways. The most frequent site of
B Leyden phenotype, where the factor IX level rises signifi- inhibitor binding occurs within the A2 and C2 domains.
cantly after puberty, with loss of the bleeding tendency. Most Inhibitors may thereby block the ability of activated factor
of these mutations have been shown to be located in regions VIIIa to bind and activate factors IXa and X, or inhibit the
The molecular basis of hemophilia 227

binding of factor VIII to VWF or negatively charged phos- HIGH RISK


pholipid surfaces. Inhibitors may also hinder the activation
75% multidomain
of factor VIII by thrombin, or the subsequent release of factor
VIII from VWF. Proteolysis of factor VIII has recently been
identified as a novel additional mechanism of inactivation in Large
some cases. deletions light chain
Activated prothrombin complex concentrates (e.g. Nonsense
mutations
FEIBA®) and recombinant activated factor VIIa (Novo- single domain Intron 22
Seven®) are valuable therapeutic materials in controlling heavy chain inversions
bleeding in those patients with inhibitory antibodies. Recom- Non A-run C1–C2 junction
Small Missense
binant porcine factor VIII (susoctocog alpha, Obizur®) has deletions
also been developed and is emerging as an additional, 0% Non C1–C2 junction
A-run
Splice-site
licensed therapeutic agent, but only in acquired hemophilia LOW RISK mutations
A currently. The rationale is that the porcine molecule is
Fig. 17.8 Mutation types and risk of inhibitor development.
sufficiently similar (homologous) to human factor VIII to
exert a hemostatic effect, but at the same time sufficiently
different to avoid rapid neutralization by antibodies. An
alternative novel therapeutic has harnessed a biphenotypic inhibitor detection in previously treated patients (PTPs) with
monoclonal antibody (Emicizumab, Hemlibra®) to mimic severe hemophilia A, despite years, often decades, of regular
the function of the FVIII molecule, bringing activated FIX exposure to FVIII CFC.
in proximity to FX to activate it. This has recently been It is now clear that the major factor which determines
licensed for prophylaxis in chronic inhibitor hemophilia A the predisposition to inhibitor development is the underlying
patients, but is not sufficiently efficacious to treat serious, molecular defect. Certain types of gene defects in hemophilia
breakthrough bleeds or cover major surgery, at which time are undoubtedly associated with a significantly increased risk
care is needed in choice of additional hemostatic agents of inhibitor development (Figure 17.8). The risk of inhibitor
to treat bleeds, due to potential thrombotic complications. development in patients with severe molecular defects, such
This molecule may also become available for prophylaxis as large deletions, nonsense mutations, and the intron-22
in those living with severe hemophilia A without a chronic inversion, is 7–10 times higher than in patients with other
inhibitor. defects such as missense mutations, small deletions, and
Another important strategy in the management of these splice-site mutations. The overall risk of inhibitor develop-
patients who develop inhibitory antibodies is immune toler- ment in patients with the common intron-22 inversion is
ance induction (ITI), which involves the daily administration approximately 30%.
of conventional CFC over a period of some months. This usu- However, there is additional evidence from family and
ally results in the eventual disappearance of the antibody, as twin studies that other subtle genetic factors play a role,
the body becomes tolerant of the protein and inhibitor for- although no associations with specific human leukocyte
mation is suppressed. antigens (HLAs) or other linkages have been conclusively
Following the elimination of the risk of transmission of identified. Race may also influence the risk of inhibitor
viruses by CFCs, the risk of inhibitor development remains development, and several studies have shown that people of
the principal danger faced by people with hemophilia nowa- Afro-Caribbean origin are more susceptible to inhibitor for-
days. Data from the UK registry suggest that approximately mation. Links between structural variants (polymorphisms)
30% of all patients with severe hemophilia A will develop in immune-response genes have been reported. Such variants
antibodies at some time, half of which are transient and might have an impact on inhibitor development by increas-
low-titer inhibitors. The majority of inhibitors in severe ing the production and secretion of chemicals that ultimately
hemophilia A occur in previously untreated patients (PUPs) enhance the production of antibodies directed against factor
early in life, during the first 50 exposure days (EDs) to the VIII. In the case of interleukin (IL)-10, one particular poly-
therapeutic CFC, with median occurrence at approximately morphism (134-bp variant of a CA repeat microsatellite in
10–15 days in severe hemophilia A. However, in non-severe the promoter region of the IL10 gene) was found to be associ-
hemophilia A, the risk is now thought to be lifelong and per- ated with a 4.4-fold increased risk of inhibitor development.
sists after 50 exposure days. In this context, the alloantibody This same polymorphism has already been shown to influ-
has the potential to cross-react with the individual’s endoge- ence the level of antibody production in such diverse diseases
nous baseline FVIII activity level, decreasing their baseline to as myasthenia gravis, multiple myeloma, and systemic lupus
potential severe levels (<1%), with a resultant deterioration erythematosus. More recently, the same group reported that
in bleed phenotype. There is also a low-level occurrence of a particular polymorphism (C/T at −318) in the cytotoxic
228 Molecular Hematology

T-lymphocyte-associated protein (CTLA)-4 receptor of which tests need to be carried out. It may seem logical to
certain immune cells actually confers protection against initiate carrier testing to determine carrier status as soon
inhibitor development. Other than knowledge of a family as possible in girls with a family history of the condition,
history of inhibitor formation which confers increased risk, as this would facilitate management in the case of an early
it continues to be challenging to risk stratify inhibitor risk by and possibly unexpected pregnancy. There are significant
other genetic markers. It has become clear that CFC product differences in legislation as well as clinical practice among
choice may be contributory to inhibitor risk in PUPs, either healthcare professionals in various countries with regard to
between recombinant products or between a recombinant the timing of testing of children for genetic disorders. Some
(rFVIII) and plasma-derived, VWF-containing product take the view that it is unethical to test very young children
(pdFVIII). The SIPPET group described the benefit of the in order to determine carrier status for inherited disorders
latter choice of pdFVIII over rFVIII CFC to be restricted for conditions which have no immediate implications for
to the minority of individuals with less disruptive F8 their own health. In the UK, most hematologists would
mutations. generally be prepared to offer genetic confirmatory carrier
Inhibitor development in hemophilia B is a rare event, testing to girls in their early teens, with the proviso that
occurring in probably less than 3% of patients, and these the issues must be discussed with the family and the child
patients often have an underlying large gene deletion. In deemed able to understand the implications of such testing.
contrast with the immunoglobulin inhibitors in patients It is important to check FVIII or FIX plasma activity lev-
with hemophilia A, inhibitory antibodies in patients with els in obligate and possible carriers (see Figure 17.4) soon
hemophilia B are often capable of fixing complement pro- after birth. It should be emphasized that these plasma lev-
teins. The administration of CFC in such cases may there- els should not be used to determine whether a female is a
fore trigger severe, often anaphylactic, allergic reactions. The carrier of hemophilia, and that only DNA-based tests should
development of nephrotic syndrome has also been reported be used. Indirect testing of carrier status using the tracking
in patients with hemophilia B and inhibitors undergoing of restriction fragment-length polymorphisms (RFLPs) has
immune tolerance induction treatment with factor IX con- now been superseded by direct identification of the underly-
centrates. ing causative genetic abnormality, such as mutation or inver-
sion. Once the molecular defect has been identified in an
individual with either hemophilia A or B, direct screening
Therapeutic applications of molecular for that defect could be applied in subsequent generations
biology to patient care for both carrier testing and antenatal diagnosis. It is recom-
mended that hemophilia centers collate information on the
family pedigree (“family tree”) to identify potential carriers
Carrier testing
and facilitate screening.
Ideally, carriers of hemophilia should be identified before RFLP analysis may still be employed in the few cases
a pregnancy, and offered counseling. The inheritance of where, for example, a mutation has not been identified or ver-
hemophilia is X-linked, as with other disorders such as color ified in a potential carrier. This technique is also still widely
blindness and Duchenne muscular dystrophy. The daughters used in countries around the world that do not yet have
of men with hemophilia are thus obligate carriers of the access to more sophisticated technology. The method is based
condition, with a subsequent 50 : 50 chance of passing on on the fact that there are some genetic polymorphisms that
the condition to their son, and there is a similar chance that represent natural variations of the genome sequence, with-
a daughter of a carrier will also herself be a carrier of the out any adverse impact on the function of the molecule.
condition. No special genetic tests are therefore required The polymorphisms are detected by cleavage of patient DNA
to determine the carrier status of daughters of men with with restriction enzymes, which generate fragments of vary-
hemophilia (unless paternity is uncertain), although the ing size according to the presence or absence of the poly-
results of DNA-based studies are likely to be useful for morphism. Only intragenic markers should be used, because
subsequent antenatal diagnostic procedures. The phenotype the small possibility of recombination may result in erro-
of hemophilia remains constant within a family, so that the neous diagnoses when extragenic probes are used. The most
daughter of a man with only mild hemophilia may be reas- widely intragenic markers used are dinucleotide repeats in
sured that she can only transmit a similarly mild form of the introns 13 and 22; XbaI dimorphism in intron 22; and diges-
condition. However, a more common problem is to be con- tion products using BclI in intron 18. The limitations of
fronted with a woman with only a vague history of a bleeding this approach include the fact that samples have to be taken
disorder in a distant relative. National patient registers can be from several members to permit the tracking of the mutant
very helpful in establishing the type and severity of bleeding gene responsible for hemophilia in the family. Blood from an
disorder of an affected relative as a first step in determining affected family member will be required, and this may not be
The molecular basis of hemophilia 229

possible in some cases. It should also be appreciated that there Uterine wall
is ethnic variation of the allelic frequencies of the various Amniotic fluid
polymorphisms, more so with factor IX than factor VIII, and
Placenta
this may influence the choice of probes used in some family
studies. Furthermore, non-paternity may confound attempts
to track the gene in families. Chorionic villi
Germline and somatic mosaicism may complicate the pic-
ture. This needs particular consideration in cases of sporadic
hemophilia where the mother of a child with hemophilia does
not appear to carry the mutation in her own leukocyte DNA.
This has been reported to be a particular problem when the
apparently de novo mutation is a point mutation. For this rea-
son, it is recommended not to state that the mother of a child
with hemophilia is not a carrier, even when the mutation is
not identified in her somatic DNA.

Antenatal diagnosis of hemophilia


Fig. 17.9 Chorionic villus sampling. A sample of trophoblastic
As a general rule, it is the practice in the UK to perform tissue from the placental area is aspirated with a fine needle under
antenatal procedures to determine whether or not a fetus general anesthesia. The procedure is usually carried out after
has hemophilia only where a termination is being contem- 11 weeks’ gestation. DNA isolated from the fetal tissue can then be
plated. The general experience in the UK has been that only analyzed to determine fetal sex and status with regard to hemophilia.
a minority of women subsequently take up the offer of ante-
natal diagnosis with a view to termination if an affected fetus
is identified. This reflects the fact that many women with fetal gender early in the first trimester (as early as five weeks’
affected relatives appreciate the major treatment advances in gestation) by free-fetal DNA analysis (detecting a Y chro-
recent years, such as recombinant products for lifelong pro- mosome in fetal cells identified in the maternal circulation).
phylaxis, the expectation of an essentially normal life quality This result then contributes to the discussion on whether to
and life expectancy for the younger generation of people with pursue more invasive confirmatory testing for the presence
hemophilia, and the imminence of potentially curative gene of the mutated F8 or F9 gene, and generally obviates the
therapy. need for CVS in the presence of a female fetus. An imminent
Amniocentesis was the first technique employed for ante- extension to this non-invasive assay will be for full antenatal
natal diagnosis of hemophilia and other X-linked disorders diagnostics.
such as muscular dystrophy. While amniocentesis is both Direct fetal blood sampling may also be used for antena-
technically simple and safe, an important limitation is the tal diagnosis of hemophilia, but this method is usually only
fact that it may only be employed in the second trimester of offered as a last resort, either because it was not possible
pregnancy, after approximately 15 weeks’ gestation. Chori- to carry out DNA-based family studies in time or because
onic villus sampling (CVS) was first applied to antenatal such studies were carried out but did not yield results. In
diagnosis of a number of genetic disorders in the early this technique, fetal blood is taken from fetal umbilical ves-
1980s, but is now the principal method used for antenatal sels under ultrasound guidance. The procedure requires con-
diagnosis of hemophilia and several other single-gene siderable expertise and will thus not be available in all hos-
disorders. The main advantage is that the method may be pitals. It is usually carried out at a minimum of 18 weeks’
applied for antenatal diagnosis during the first trimester, gestation. The levels of factor VIII and IX in a normal
so that if termination of the pregnancy is required this is fetus at around 19 weeks’ gestation are significantly lower
easier to carry out. Furthermore, the results of the test are than those in an adult, approximately 40 and 10 IU dl−1 ,
often available within only a few days of the procedure, as respectively.
(in contrast to amniocentesis) there is no need to culture The need to abort an implanted fetus is a formidable
cells before genetic analysis. A sample is obtained by either ethical concern for many female carriers of hemophilia. Pre-
a transabdominal or transvaginal route, under ultrasound implantation diagnosis is another technique that has been
guidance (Figure 17.9). CVS should not be undertaken developed, and this may prove to be particularly attractive to
before 11 weeks of pregnancy in order to minimize the risk women who would not be prepared to undergo conventional
of inducing congenital limb abnormalities. It is now common termination of a well-established pregnancy. The female
for there to be non-invasive, prenatal determination of the partner must undergo an in vitro fertilization cycle and the
230 Molecular Hematology

fertilized ova are then biopsied. Embryos shown to be unaf- human albumin as a stabilizer. However, third- and fourth-
fected by hemophilia can then be transferred to the uterus. generation products are now available in which alternative
The initial approach was to offer determination of embry- stabilizers are used. Human albumin and all bovine pro-
onic sex alone using dual fluorescence in situ hybridization teins have also been eliminated from the culture media
of blastomere cells with labeled probes specific for the sex of these modern products, and the incorporation of spe-
chromosomes. More recently, specific mutation analysis cific viral elimination/inactivation steps further increases the
has become possible, although it must be emphasized that margin of safety. Standard half-life recombinant factor VIII
this has only been performed in a very limited number of has an essentially identical amino acid structure to natu-
cases and is still far from being a service that is available ral plasma factor VIII, although increasingly the apparently
on a routine basis even in major hospitals. Sperm sorting functionless B domain of the F8 gene is deleted or truncated
has been developed as a cheaper and easier method for to optimize production efficiency. Human cell (HEK293)-
pre-implantation sex determination, using flow cytometry to derived recombinant factor VIII became available for the
sort spermatozoa labeled with a fluorescent dye that binds to first time recently as a standard or extended half-life prod-
DNA. The resulting higher fluorescence of X chromosomes, uct (see later). Evidence exists to demonstrate different pro-
which contain more DNA than Y chromosomes, facilitates tein post-translational modifications between human- and
separation. CHO- or BHK-derived CFC products. It remains unclear
whether this translates into human cell-derived rFVIII prod-
ucts having a reduced immunogenicity compared to CHO-
Recombinant blood products
or BHK-derived products. Additional developments include
The development of plasma-derived CFCs in the early 1970s further structural modifications to increase the half-life of
dramatically improved both the longevity and quality of life the infused FVIII CFC by fusing the FVIII protein to an
of patients with hemophilia, and the demand for factor VIII immunoglobulin Fc stem, enabling apparent recycling via the
and IX has risen steadily. The burgeoning global demand for endothelial surface neonatal Fc receptor (FcR). Glycopegyla-
factor VIII can no longer be met by products derived from tion is an alternative strategy for half-life prolongation. Both
volunteer blood donors. The manufacture of recombinant strategies have increased the half-life of FVIII comparably by
coagulation factor proteins offers the promise of unlimited 1.5-fold.
supplies, albeit at increased cost. However, the most impor- Recombinant factor IX is also available. Recombinant
tant advantage of recombinant products is safety with regard factor IX is identical in amino acid sequence to the Ala148
to the transmission of human pathogens. Many patients with (as opposed to the less common Thr148) human polymor-
hemophilia were infected with HIV and/or hepatitis C before phic variant. Plasma factor IX is synthesized in the liver and
the introduction of physical methods of viral inactivation undergoes post-translational glycosylation of a number of
of plasma-derived CFCs in 1985. More recently, there has glutamic acid residues. Vitamin K is a vital cofactor in this
been concern about the possibility of transmission of vari- process, which is essential for its activity, but recombinant
ant Creutzfeldt–Jakob disease (vCJD) via blood products, as factor IX is not as effectively carboxylated. The post-infusion
the prions believed to be the cause of this neurological dis- recovery does appear to be reduced when compared with
order are extremely resistant to the usual viral inactivation plasma-derived products, although the plasma half-life
procedures, such as heat treatment and exposure to a sol- is identical. It is a smaller molecule than factor VIII and
vent/detergent mixture. Many clinicians now regard recom- requires no albumin, or other material, to be added to
binant products as the treatment of choice for all patients with the final product as a stabilizer. The cell line is grown in
hemophilia, as they offer the best possible protection from media that contain no animal or human-derived proteins,
the transmission of blood-borne pathogens. However, the but the product is subjected to nanofiltration to enhance
increased cost with regard to conventional plasma-derived its safety profile. There is no suggestion of an increased
products has limited the availability of these products in risk of inhibitor development associated with the use of
many parts of the world. recombinant factor IX.
Recombinant CFCs are manufactured by insertion of the Factor IX has more complex pharmacokinetics than FVIII,
most common H1 or H2 F8 human haplotype gene into as a smaller protein that is distributed extravascularly and
mammalian cell lines – such as Chinese hamster ovary also binds to collagen IV. Consensus on the true half-life of
(CHO) cells or baby hamster kidney (BHK) cells – which FIX is debatable, but it is likely longer than the previously esti-
are then grown in culture on an industrial scale. Factor mated 18 hours due to this apparent complex multicompart-
VIII (or IX) is secreted into the growth medium, from mental pharmacokinetics. Strategies to prolong FIX half-life
which it is subsequently extracted by monoclonal or other in the intravascular space have proved successful with either
immunoaffinity chromatography (Figure 17.10). The origi- glycopegylation, fusion to albumin or the immunoglobulin
nal recombinant factor VIII products all contained added Fc molecule. Although prolonging detectable FIX activity in
The molecular basis of hemophilia 231

FVIII
vWF

(Circle of bacterial DNA that replicates)


(a) Plasmid vector (b) Isolate human gene sequence and
splice into a suitable vector
FVIII
vWF

Nucleus

Cells in
nutrient media

Vial of cells is “cultured” in


media, cells multiply into
CHO cell Insert recombinant
millions of cells, each
plasmid into host cell
expressing FVIII into the
media
(c) Chinese hamster ovary (CHO) cell (d) Bioreactor
FVIII
Impurities

Impurities pass
Column is washed
through column
and rFVIII protein
and rFVIII binds
is released
to antibodies

Impurities
rFVIII
Protein
(e) FVIII is purified from media using a monoclonal
Ab column and ion-exchange columns
Fig. 17.10 Manufacture of recombinant factor VIII. The gene for human factor VIII is incorporated into a bacterial vector (a, b). Inclusion of
the von Willebrand factor gene also enhances production of factor VIII. The vector is then inserted into a mammalian host cell (c). The cells grow
and multiply in a nutrient medium, and then secrete factor VIII (d). Factor VIII can be extracted and purified by immunoaffinity chromatography (e).
The final product also contains no von Willebrand factor.

the intravascular space three- to fivefold, the impact of each Gene therapy for hemophilia
of these novel technologies on extravascular distribution of
Gene therapy (see also Chapter 23) poses a number of ethi-
FIX is incompletely understood.
cal problems, particularly since effective and safe treatment
Another useful recombinant product is recombinant acti-
with recombinant coagulation factors is already available for
vated factor VII (NovoSeven; Novo Nordisk). It is now recog-
patients with both hemophilia A and B. Gene therapy does
nized that factor VII plays a key role in initiation of the coag-
offer the prospect of a cure for hemophilia in the long term,
ulation cascade through contact with tissue factor released
but it must be emphasized that although clinical trials in
from damaged tissues, to form activated factor VII (see also
both hemophilia A and B are progressing well into later
Chapter 16). Recombinant activated factor VII is very useful
phases, there is still a need for long-term surveillance for
in the clinical management of patients with either hemophilia
potential side effects. Trials are all currently in adults and it
A or B and inhibitory antibodies, as well as those with
will be some time before incrementally younger children are
acquired hemophilia.
232 Molecular Hematology

included in trials. It should also be noted that gene therapy predominantly affected, but some female carriers will
does not correct the germ line F8 or F9 mutation. also be symptomatic. Approximately half of cases arise in
There are two basic approaches to gene delivery into cells. families with no previous family history, and represent new
The first technique involves the direct injection of transduc- mutations. The typical features of severe hemophilia include
ing vector into the bloodstream or target tissue, with sub- spontaneous bleeding into joints and resultant disability,
sequent in vivo transformation of the cells which take up but in the absence of treatment more serious complica-
the gene, namely hepatocytes in hemophilia. Alternatively, tions (such as intracranial hemorrhage) will lead to early
target cells may be modified by removal of cells from a death.
patient, with subsequent modification ex vivo of these cells The first products used for the treatment of hemophilia
followed by reinfusion. The first strategy has been favored in were derived from human plasma, but unfortunately the use
hemophilia trials, utilizing non-integrating, non-replicative, of pooled plasma products before 1985 resulted in the trans-
hepatotropic adeno-associated viruses (AAVs). Wild-type mission of serious viral infections such as HIV and hepatitis
AAVs are communally acquired, intercurrent viral infections C to many patients. The development of recombinant blood
of little consequence to an immunocompetent host. AAVs products has eliminated the risk of transmission of these
exist in multiple serotypes, some more homologous than oth- infections, and also offers the prospect of a secure supply
ers, and are less immunogenic than adenovirus vectors in the chain and half-life–extended products. The life expectancy
gene therapy setting. Patients may have pre-existing immu- of the younger generation of hemophiliacs now approaches
nity to a given AAV serotype, currently making them ineligi- that of the normal population.
ble for gene therapy. A dose of AAV gene therapy does gen- The commonest molecular defect in severe hemophilia
erate high-titer antibodies against the specific AAV serotype A is an inversion in intron 22 of the factor VIII gene
vector used which may cross-react with other serotypes, on the X chromosome, which accounts for approximately
making AAV gene therapy a single opportunity. Some, but half of all cases. Genetic testing documents the genetic
not all, serotypes trialed to date provoke a viral capsid- defect in each family, and identifies carriers within fam-
specific T-cell response thought, at least in part, to be contrib- ilies. Antenatal diagnosis is readily available, although in
utory to a complicating rise in liver-specific enzymes requir- countries with sustainable hemophilia care, recourse to ter-
ing intervention with steroids. mination of an affected pregnancy is rare. Recent trials
Despite the recent success of non-integrative AAV strate- of gene therapy for hemophilia have yielded encouraging
gies, other gene therapy, gene editing, or base editing plat- results.
forms are also in development. Integration events provoke
concerns of oncogenesis. However, “safe-haven” gene edit-
ing may circumvent this issue, as is now being trialed in Hemophilia resources on the internet
hemophilia B, precisely inserting the F9 gene downstream of
Factor VIII Variant Database. Available at http://www.
the albumin promoter in hepatocytes using zinc finger nucle-
factorviii-db.org
ase (ZFN).
World Federation of Haemophilia. Available at www.wfh
The smaller F9 gene made it easier to package into the
.org
modified AAV vectors for early study, yielding durable low-
United Kingdom Haemophilia Centre Doctors’ Organisa-
level yet effective FIX expression in humans. Implementa-
tion (UKHCDO). Available at www.ukhcdo.org
tion of the aforementioned R338L gain of function F9 Padua
European Association for Haemophilia and Allied Disor-
polymorphism has further improved expression in subse-
ders (EAHAD). Available at www.eahad.org
quent hemophilia B trials. Despite the larger F8 gene with
the anticipated problems of vector packaging, the first-in-
man hemophilia A gene therapy trial has normalized FVIII
activity levels in early participants, durable for at least a year. Further reading
For the first time, curative gene therapy seems to be a distinct
possibility. General
Darby, S.C., Kan, S.W., Spooner, R.J. et al. (2007). Mortality rates, life
expectancy, and causes of death in people with hemophilia A or B
in the United Kingdom who were not infected with HIV. Blood 110:
Conclusions 815–825.
Lee, C.A., Berntorp, E., and Hoots, W.K. (eds.) (2005). Textbook of
Hemophilia is an inherited disorder of coagulation, asso- Hemophilia. Oxford: Blackwell Publishing.
ciated with congenital deficiency of factor VIII (or IX). Zeepvat, C. (1998). Prince Leopold: The Untold Story of Queen Victoria’s
It is inherited in a X-linked fashion, so that males are Youngest Son. Stroud: Sutton Publishing.
The molecular basis of hemophilia 233

Hemophilia A Astermark, J., Oldenburg, J., Escobar, M. et al. (2005). The Malmö Inter-
national Brother Study (MIBS). Genetic defects and inhibitor devel-
Antonarakis, S.E., Rossiter, J.P., Young, M. et al. (1995). Factor VIII gene opment in siblings with severe hemophilia A. Haematologica 90: 924–
inversions in severe hemophilia A: results of an international consor- 931.
tium study. Blood 86: 2206–2212. Astermark, J., Oldenburg, J., Pavlova, A. et al. (2006). Polymorphisms
Bagnall, R.D., Waseem, N., Green, P.M., and Giannelli, F. (2002). Recur- in the IL10 but not in the IL1beta and IL4 genes are associated with
rent inversion breaking intron 1 of the factor VIIII gene is a frequent inhibitor development in patients with hemophilia A. Blood 107:
cause of severe hemophilia A. Blood 99: 168–174. 3167–3172.
Davidson, C.J., Tuddenham, E.G., and McVey, J.H. (2003). 450 million Collins, P.W., Chalmers, E., Hart, D.P. et al., United Kingdom
years of hemostasis. J. Thromb. Haemost. 1 (7): 1487–1494. Haemophilia Centre Doctors. (2013). Diagnosis and treatment of fac-
Keeney, S., Mitchell, M., and Goodeve, A. (2005). The molecular anal- tor VIII and IX inhibitors in congenital haemophilia (4th edition).
ysis of haemophilia A: a guideline from the UK Haemophilia Centre UK Haemophilia Centre Doctors Organization. Br. J. Haematol.
Doctors’ Organization Haemophilia Genetics Laboratory Network. 160 (2): 153–170.
Haemophilia 11: 387–397. Collins, P.W., Palmer, B.P., Chalmers, E.A. et al., United Kingdom
Leuer, M., Oldenburg, J., Lavergne, J.M. et al. (2001). Somatic mosaicism Haemophilia Centre Doctors’ Organization. (2014). Factor VIII
in hemophilia A: a fairly common event. Am. J. Hum. Genet. 69: 75– brand and the incidence of factor VIII inhibitors in previously
87. untreated UK children with severe hemophilia A, 2000-2011. Blood
Rost, S., Löffler, S., Pavlova, A. et al. (2008). Detection of large dupli- 124 (23): 3389–3397.
cations within the factor VIII gene by MLPA. J. Thromb. Haemost. 6: Eckhardt, C.L., Loomans, J.I., van Velzen, A.S. et al., INSIGHT Study
1996–1999. Group. (2015). Inhibitor development and mortality in non-severe
Wood, W.I., Capon, D.J., Simonsen, C.C. et al. (1984). Expression of hemophilia A. J. Thromb. Haemost. 13 (7): 1217–1225.
active human factor VIII from recombinant DNA clones. Nature 312 Eckhardt, C.L., van Velzen, A.S., Peters, M. et al., INSIGHT Study
(5992): 330–337. Group. (2013). Factor VIII gene (F8) mutation and risk of inhibitor
development in nonsevere hemophilia A. Blood 122 (11): 1954–1962.
Huth-Kühne, A., Baudo, F., Collins, P. et al. (2009). International recom-
Hemophilia B mendations on the diagnosis and treatment of patients with acquired
Choo, K.H., Gould, K.G., Rees, D.J., and Brownlee, G.G. (1982). Molec- hemophilia A. Haematologica 94: 566–575.
ular cloning of the gene for human anti-haemophilic factor IX. Nature Oldenburg, J. and Pavlova, A. (2006). Genetic risk factors for inhibitors
299 (5879): 178–180. to factor VIII and IX. Haemophilia 12 (Suppl 6): 15–22.
Giangrande, P.L.F. (2005). Haemophilia B: Christmas disease. Expert Peyvandi, F., Mannucci, P.M., Garagiola, I. et al. (2016). A randomized
Opin. Pharmacother. 6: 1517–1524. trial of factor VIII and neutralizing antibodies in hemophilia A. N.
Mitchell, M., Keeney, S., and Goodeve, A. (2005). The molecular anal- Engl. J. Med. 374 (21): 2054–2064.
ysis of haemophilia B: a guideline from the UK Haemophilia Centre Saint Remy, J.M. (2008). How to get rid of inhibitors. Haemophilia
Doctors’ Organization Haemophilia Genetics Laboratory Network. 14 (Suppl 3): 33–35.
Haemophilia 11: 398–404. Saint Remy, J.M., Lacroix-Desmazes, S., and Oldenburg, J. (2004).
Reijnen, M.J., Peerlinck, K., Maasdam, D. et al. (1993). Hemophilia B Inhibitors in haemophilia: pathophysiology. Haemophilia 10 (Suppl
Leyden: substitution of thymine for guanine at position −21 results 4): 146–151.
in a disruption of a hepatocyte nuclear factor 4 binding site in the Shepherd, A.J., Skelton, S., Sansom, C.E. et al. (2015). A large-scale com-
factor IX promoter. Blood 82: 151–158. putational study of inhibitor risk in non-severe haemophilia A. Br. J.
Rogaev, E.I., Grigorenko, A.P., Faskhutdinova, G. et al. (2009). Geno- Haematol. 168 (3): 413–420.
type analysis identifies the cause of the “royal disease”. Science van Velzen, A.S., Eckhardt, C.L., Hart, D.P. et al., INSIGHT study group.
326 (5954): 817. (2015). Inhibitors in nonsevere haemophilia A: outcome and eradi-
Simioni, P., Tormene, D., Tognin, G. et al. (2009). X-linked throm- cation strategies. Thromb. Haemost. 114 (1): 46–55.
bophilia with a mutant factor IX (factor IX Padua). N. Engl. J. Med.
361: 1671–1675. Carrier testing and antenatal diagnosis
Yoshitake, S., Schach, B., Foster, D.C. et al. (1985). Nucleotide sequenc-
Dunn, N.F., Miller, R., Griffioen, A., and Lee, C.A. (2008). Carrier test-
ing of the gene for human factor IX (antihemophilic factor B). Bio-
ing in haemophilia A and B: adult carriers’ and their partners’ expe-
chemistry 24: 3736–3750.
riences and their views on the testing of young females. Haemophilia
14: 584–592.
Inhibitors Finning, K.M. and Chitty, L.S. (2008). Non-invasive fetal sex determi-
nation: impact on clinical practice. Semin. Fetal Neonatal. Med. 13:
Astermark, J., Donfield, S.M., Gomperts, E.D. et al., Hemophilia 69–75.
Inhibitor Genetics Study (HIGS) Combined Cohort. (2013). The Lavery, S. (2009). Preimplantation genetic diagnosis of haemophilia. Br.
polygenic nature of inhibitors in hemophilia A: results from the J. Haematol. 144: 303–307.
Hemophilia Inhibitor Genetics Study (HIGS) Combined Cohort. Ludlam, C.A., Pasi, K.J., Bolton-Maggs, P. et al. (2005). A framework for
Blood 121 (8): 1446–1454. https://doi.org/10.1182/blood-2012-06- genetic service provision for haemophilia and other inherited bleed-
434803. ing disorders. Haemophilia 11: 145–163.
234 Molecular Hematology

Street, A.M., Ljung, R., and Lavery, S. (2008). Management of carriers Pasi, K.J., Rangarajan, S., Georgiev, P. et al. (2017). Targeting of
and babies with haemophilia. Haemophilia 14 (Suppl 3): 181–187. antithrombin in hemophilia A or B with RNAi therapy. N. Engl. J.
Med. 377 (9): 819–828.
Peters, R. and Harris, T. (2018). Advances and innovations in
Hemophilia therapeutics: current haemophilia treatment. Nat. Rev. Drug Discov. 17 (7): 493–
and future 508.
Oldenburg, J., Mahlangu, J.N., Kim, B. et al. (2017). Emicizumab pro-
phylaxis in hemophilia A with inhibitors. N. Engl. J. Med. 377 (9):
809–818.
Chapter 18 The molecular basis of von
Willebrand disease
Luciano Baronciani
Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy

Introduction, 235 Genetic defects in VWD, 238


Function of VWF in primary hemostasis, 235 Treatment of VWD, 244
Function of VWF in blood coagulation, 236 Conclusion, 246
Gene organization, synthesis, and multimeric structure of VWF, 236 Further reading, 247
von Willebrand disease and its classification, 236

of platelets in hemostasis is to become irreversibly attached at


Introduction sites of injury. The primary physical factor that affects platelet
binding to the vessel wall is the rate of blood flow in the
von Willebrand disease (VWD) is a common inherited bleed-
vessel, which is faster at the center and slower close to the
ing disorder. Precise data regarding its prevalence are not
wall. These variations in velocity create a shearing effect, or
available due to the extreme variability in clinical symptoms
shear stress, between layers of fluid. Disruption of the vascu-
of mild VWD. However, population-based studies give an
lar endothelial surface leads to exposure of the subendothe-
estimate of clinically significant VWD with a prevalence of
lium and results in an alteration of the rate of blood flow
at least 100 per million. VWD is caused by the deficiency
and an increase in shear stress. Plasma VWF binds rapidly
or dysfunction of a multimeric plasma glycoprotein, von
and tightly to subendothelial collagen. VWF does not con-
Willebrand factor (VWF). Because of its ability to bind to
stitutively bind platelets, but in high blood-flow conditions
a number of ligands, VWF is involved in hemostasis via
is capable of tethering platelets and exposing thrombogenic
a number of mechanisms, but can be considered as hav-
surfaces through the interaction of its A1 domain with the
ing two main roles: in primary hemostasis and in intrin-
platelet receptor glycoprotein (GP)Ib. However, the VWF–
sic blood coagulation. VWF is directly involved in platelet
GPIb interaction does not provide irreversible platelet adhe-
binding to the subendothelium and in platelet–platelet inter-
sion because of the fast dissociation rate, and platelets teth-
actions, and also acts as the carrier of procoagulant fac-
ered to the vessel wall still move constantly in the direction
tor VIII (FVIII). Mutations at the VWF locus can affect
of the flow, but at a much slower rate.
VWF synthesis, its complex biosynthetic assembly, stability
A second molecule on the platelet surface is required
in the circulation, and its binding interactions with specific
to obtain firm platelet adhesion: the integrin GPIIb-IIIa
ligands.
(αIIbβ3). This molecule is responsible for the platelet–
platelet interaction, which is mediated by VWF and, under
slow-flow conditions, by fibrinogen. αIIbβ3 does not appear
to be involved in the first events of platelet adhesion, proba-
Function of VWF in primary bly because its rate of binding to VWF is too slow to mediate
hemostasis the initial platelet attachment to the vessel wall under high-
flow conditions. However, when platelets become activated
At the time of a hemostatic challenge, VWF acts as a bridge as a consequence of the VWF–GPIb interaction, αIIbβ3
between platelets and the subendothelium of blood vessels, increases its affinity for its ligand VWF. This event, together
and is involved in the formation of the platelet plug. The role with the slow motion of platelets due to the VWF–GPIb inter-
action, allows αIIbβ3 to bind platelets irreversibly to the ves-
sel wall (Figure 18.1). In all vessels with a high shear rate,
VWF is the primary mediator of platelet binding to the vessel
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben.
wall and of platelet aggregation.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.

235
236 Molecular Hematology

Initial tethering Translocation Firm adhesion


Non-activated Activated
Fluid flow Activation
Torque

GPlbα A1 domain RGDS


Non-activated αllbβ3
Activated αllbβ3 von Willebrand factor

Fig. 18.1 Schematic representation of platelet adhesion to immobilized von Willebrand factor (VWF). At first, platelets are tethered to
the subendothelium through the interaction of their glycoprotein (GP)Ibα with VWF (A1 domain), while the inactivated αIIbβ3 do not bind to the
VWF’s RGDS sequence. Due to the torque imposed by the flowing fluid, platelets begin to roll. New bonds are formed as different regions of the
membrane of the rolling platelets come in contact with the surface, and the translocation continues until the αIIbβ3 becomes activated and binds
firmly to the RGDS of the VWF.
Source: Adapted with permission from Ruggeri Z.M. (1997). von Willebrand Factor. Journal of Clinical Investigation 99: 559–564.

The biosynthesis of VWF is a complex process, involv-


Function of VWF in blood ing post-translational processing of the protein prior to stor-
coagulation age or release into the circulation. The initial dimeriza-
tion occurs by disulfide bonding between cysteine residues
VWF circulates in plasma as a complex associated with FVIII.
in the cystine knot (CK) carboxyl-terminal region of the
The binding of VWF to FVIII is required to stabilize FVIII in
monomer (tail-to-tail dimerization). The signal peptide is
the circulation, preventing cleavage by activated protein C or
cleaved before it enters the Golgi apparatus. The tail-to-tail
factor Xa. This interaction with VWF is crucial in prolong-
glycosylated dimers are then transported to the Golgi appara-
ing the half-life of FVIII and concentrating FVIII at the point
tus, where multimerization and further glycosylation occur.
of bleeding. When patients with severe VWD (type 3) are
The propeptide of VWF mediates the assembly of VWF mul-
treated with purified FVIII, this is cleared with a half-life of
timers. This process requires the presence of the D1 and
less than 2 hours, whereas the VWF–FVIII complex infused
D2 domains (propeptide) and the D′ and D3 domains of
in the same patient has a half-life of about 12–14 hours. VWF
the mature polypeptide. This is followed by cleavage of the
binds to FVIII via regions within the first 272 residues of the
propeptide sequence (Figure 18.3) and secretion of both the
mature polypeptide, in the D′ and D3 domains of VWF.
mature VWF and the propeptide into the circulation, or stor-
age within the Weibel–Palade bodies of endothelial cells or
the α-granules of megakaryocytes/platelets. VWF in these
Gene organization, synthesis, and storage organelles presents very large multimers (ultra-large
multimeric structure of VWF VWF) that exceed the size of multimers found in plasma.
However, when these molecules are released into the circu-
The gene encoding VWF has been mapped in chromosome lation, these forms persist only for a limited time, because
12, has a length of approximately 178 kb with 52 exons, and a metalloprotease, ADAMTS-13 (a disintegrin and metal-
transcribes a messenger RNA of about 8.2 kb. Analysis of loprotease with a thrombospondin type 1 motif, member
the VWF gene is complicated by the existence of a partial 13), physiologically cleaves the ultra-large VWF, reducing its
unprocessed pseudogene in chromosome 22. The pseudo- molecular weight.
gene extends from exon 23 to exon 34 and shares a high
degree of homology with the gene (97%). VWF is synthe-
sized in megakaryocytes and endothelial cells as a precur- von Willebrand disease and its
sor of 2813 amino acids, the pre-pro-VWF. It is composed classification
of a 22-residue signal peptide, a 741-residue propeptide and a
2050-residue mature subunit. More than 95% of the sequence The most common symptoms of mild VWD are mucosal
accounts for structural domains arranged in the following bleeding (epistaxis, gingival bleeding, menorrhagia) and pro-
order: D1-D2-D′ -D3-A1-A2-A3-D4-C1-C2-C3-C4-C5-C6- longed bleeding after surgical procedures and dental extrac-
CK (Figure 18.2). tions. Hemarthroses and soft-tissue hematomas are rare, but
The molecular basis of von Willebrand disease 237

Pseudogene chromosome 22
Gene chromosome 12 23 34
51 introns 178 kb

1 11 28 38 52
5’ 3’

2A* 2A* 2A 2A 2A
(IIC) (IIC) 2N*2N*(IIE) 2B 2M (IIA) 2M (IID)
SP

NH2 D1 D2 D’ D3 A1 A2 A3 D4 C1 C2 C3 C4 C5 C6 CK COOH
ADAMTS
GPlbα 13
multimer collagen GPIIb/IIIa dimer
FVIII heparin 2813 a.a
S-S type I & III (αIIbβ3) S-S
collagen
pro-peptide type IV VWF mature polypeptide
Fig. 18.2 Structure of the von Willebrand factor (VWF) gene, pseudogene, and protein precursor. The schematic structure of the
pre-pro-VWF is reported along with the homologous repeated domain. Locations of intersubunit disulfide bonds involved in dimerization and
multimerization are shown together with the binding sites for several ligands. Arrows point out the position of von Willebrand disease (VWD) type
2 mutations. Recessive VWD type 2 variants are indicated by an *.

mRNA
5’ 3’ 8.5 Kb
Translation
pre-pro-VWF glycosylation
N C 2813 a.a. ( 260 Kda )
sp VW Ag II VWF

C-terminal Rough endoplasmic


SS
dimerization reticulum
520 kDa
CC

N N

Carbohydrate N-terminal
processing multimerization
pro-VWF multimer SS SS

SS SS SS
CC CC

Golgi apparatus

N N N N N N

up to 20000 kDa

Propeptide cleavage
N
VWF multimer N
N N

SS SS SS SS

SS
Golgi storage body
SS SS SS SS
CC CC CC CC or secretion
N N N N N N N N N N

Fig. 18.3 Processing steps of the von Willebrand factor (VWF) multimer synthesis and subcellular localization of the
post-translational modification events. The propeptide is shown in blue, the mature subunit in red. The N- and C- prefixes represent,
respectively, the amino and carboxyl terminal ends of the protein. Monomers are linked together at the C-termini by disulfide bonds to form
dimers (endoplasmic reticulum), which further multimerize in the Golgi apparatus. The cleavage of the propeptide occurs prior to secretion.
238 Molecular Hematology

they occur in severely affected individuals (type 3). The diag-


Table 18.1 Current classification of von Willebrand disease
nosis of VWD is suspected in individuals with these symp-
toms and a family history of bleeding. Type 1 Partial quantitative deficiency of von Willebrand factor
Several VWF assays are used in the diagnosis and classi- (VWF)
fication of VWD. The initial laboratory assessments evalu- Type 2 Qualitative deficiency of VWF
ate plasma levels of FVIII coagulant activity (FVIII:C), VWF 2A Decreased VWF-dependent platelet adhesion with lack
antigen (VWF:Ag), and VWF-GPIb binding activity, mea- of high-molecular-weight VWF multimers
sured usually using the antibiotic ristocetin and formalin 2B Increased affinity for platelet glycoprotein Ib
fix or lyophilized platelets (VWF ristocetin cofactor activity, 2M Decreased VWF-dependent platelet adhesion with
VWF:RCo). Recently, several new assays, not using platelets normal multimeric structure
2N Markedly decreased VWF binding affinity for factor VIII
with or without ristocetin, have been developed to measure
Type 3 Complete deficiency of VWF
the VWF–GPIb binding activity, allowing many laboratories
to evaluate this important VWF function. Additional tests are
necessary for the classification of VWD types, most of which
are not yet standardized and are performed only in special- is characterized by the partial quantitative deficiency of VWF.
ized laboratories. Type 2 is characterized by qualitative abnormalities of VWF
VWF collagen-binding activity (VWF:CB) measures the and is further categorized into subgroups (2A, 2B, 2M, and
capacity of VWF to bind collagen (typically type 1 or type 3). 2N) depending on the nature of the qualitative defect (see
A VWF:CB value that is disproportionately low compared later). Type 3 is the most severe form of the disease, character-
with VWF:Ag reflects the loss of high-molecular-weight ized by the complete absence of VWF in plasma and platelets.
VWF multimers, or can reflect a specific VWF collagen- Transmission of type 1 VWD is usually dominant, type 2 is
binding deficiency, as in the case of mutations in the dominant in the majority of cases, whereas transmission of
A3 domain. VWF multimer analysis evaluates the dis- type 3 is recessive (Figure 18.4).
tribution of VWF oligomers in the plasma through an
SDS-agarose electrophoresis (western blot technique). Using
a non-reducing low-resolution (1.2% agarose/0.1% sodium Genetic defects in VWD
dodecylsulfate) gel, it is possible to appreciate the lack
of high- and intermediate-molecular-weight multimers In the last two decades, many molecular defects of the VWF
or the presence of ultra-large VWF multimers. Using a gene have been identified in VWD patients, with more than
non-reducing intermediate-resolution (1.6% agarose/0.1% 900 different mutations reported. At first, most of the muta-
sodium dodecylsulfate) gel, individual VWF oligomers from tions were identified in patients with the functional variants
normal plasma appear as a triplet, consisting of a main (type 2A, 2B, 2M, and 2N), then in the rarer type 3, and
band flanked by two satellite bands, being the products of last in the most common type 1 VWD. The unique pheno-
proteolysis by ADAMTS-13. Measurement of VWF binding types of the type 2 variants made it possible to restrict the
capacity to FVIII (VWF:FVIIIB) is crucial to diagnose type genetic analysis to the exons encoding for specific structural
2N VWD patients. Ristocetin-induced platelet agglutination domains, such as the A1 domain for the variants 2B and most
(RIPA) assay evaluates the VWF affinity to platelet GPIb. The 2M, A2 for the more common variant 2A, and D′ –D3 for the
assay is performed using an aggregometer, evaluating patient variant 2N. On the other hand, characterization of molecular
platelet-rich plasma agglutination at different concentrations defects in type 1 and 3 VWD requires extensive screening,
of ristocetin. This test is important to identify type 2B since mutations are not restricted to specific regions and are
variants and in particularly to differentiate type 2B from usually scattered throughout the VWF gene.
type 2A VWD. The evaluation of von Willebrand factor In addition, whereas the identification of a nonsense muta-
propeptide (VWFpp) plasma level is useful to establish the tion or a small deletion, more common in type 3 VWD, can
VWF clearance rate by determining the steady-state VWFpp undoubtedly be considered the cause of the disease, the iden-
to VWF:Ag ratio. Evaluation of VWF platelet content, using tification of missense mutations, more common in type 1
VWF:Ag and VWF:RCo, might be useful to discriminate VWD, requires supplementary studies in order to confirm
among different VWD types. their relationship with the disease, causing further complex-
This large number of measurements reflects the fact that ity in the molecular characterization of the most common
none of them is by itself sensitive and specific enough for a variant. In most type 2 cases the mutations consist in amino
diagnosis of VWD. VWD is a highly heterogeneous disease in acid substitutions (missense mutations), although small in-
which there are quantitative and qualitative abnormalities of frame deletions or insertions have been reported. In type 3
VWF, caused so far only by mutations at the VWF locus. The VWD the majority of molecular defects identified are respon-
classification identifies three basic types (Table 18.1). Type 1 sible for null alleles (nonsense, splice-site mutations, small
The molecular basis of von Willebrand disease 239

(a)

Ch12 Ch12 Ch12 Ch12

Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12

Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12

(b)

Ch12 Ch12 Ch12 Ch12

Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12

Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12 Ch12
Fig. 18.4 (a) A family pedigree with an autosomal dominant segregation, as in von Willebrand disease (VWD) type 1, 2A(IIA), 2A(IID), 2A(IIE), 2B,
and 2M. (b) A family pedigree with an autosomal recessive segregation, as in VWD type 2A(IIC), 2N, and 3.
240 Molecular Hematology

deletions, small insertions, and, more rarely, large gene dele- VWD type 2, the missense mutations are found throughout
tions). Nevertheless, numerous missense mutations have also the entire VWF gene.
been identified. In type 1 the opposite seems to be the case, Patients with type 1 VWD could be divided roughly into
with a majority of missense mutations and a minority of three groups: (i) patients with a fully penetrant dominant
splice-site mutations, small deletion and insertion (some- mutation with low VWF levels; (ii) patients with an incom-
times in-frame), and more rarely nonsense mutations. plete penetrant mutation, where not all the family members
Nowadays, the diffusion of new molecular techniques, like with the mutation have low VWF levels; and (iii) patients
multiplex ligation-dependent probe amplification (MLPA) with no apparent mutations in the VWF gene. The first
and in particular next-generation sequencing (NGS), along group is characterized by penetrant missense mutations like
with the use of online databases and in silico analysis, has p.Cys1130Phe and p.Cys1149Arg. An in vitro study has clar-
improved patients’ molecular characterization. MLPA anal- ified the molecular mechanism of these dominant negative
ysis allows the identification of large deletions or duplica- mutations found in the D3 domain, showing that intracellu-
tions in the whole VWF coding region in a single assess- lar retention of mutant VWF is the predominant responsi-
ment. NGS enables investigation of the entire VWF coding ble mechanism. In this group also, accelerated clearance can
region in just few days, making the molecular characteriza- cause type 1 VWD, like the case of mutation p.Arg1205His
tion of VWD types 1 and 3 as straightforward as type 2. The (Vicenza) being in the D3 domain. This variant has been
use of online databases reporting previously identified VWD found in several populations (all large type 1 VWD stud-
mutations, along with databases containing single nucleotide ies) and is always characterized by a low level of VWF:Ag,
variants and their frequency in different population groups, normal platelet VWF, and ultra-large plasma VWF multi-
helps to confirm, sustain, or exclude the pathogenicity of an mers. Compared with healthy controls, the half-life of VWF
identified variant. In addition, different kinds of software are Vicenza is reduced 4.4-fold after desmopressin, suggesting
now available online to evaluate (in silico analysis) the poten- that rapid clearance accounts for the moderately severe VWF
tial negative impact of candidate missense, synonymous, or deficiency. In addition, rapid clearance decreases the time
splicing mutations. during which a large, circulating VWF multimer can be
cleaved by ADAMTS-13, resulting in partial preservation of
ultra-large multimers. Mutation p.Ser2179Phe is also charac-
Type 1 VWD
terized by a short VWF half-life after desmopressin, although
This is the most common form, comprising approximately not to the same extent as mutation p.Arg1205His. Also this
70% of all cases of VWD. Patients have mild to moderate mutation has been reported by several population studies on
bleeding symptoms, a normal or variably prolonged bleed- type 1 VWD patients.
ing time, decreased levels of VWF in plasma, and a normal In the second group, incomplete penetrant mutations, like
multimeric structure, although some cases show a slightly p.Tyr1584Cys, have been identified. In the heterozygous state
decreased level of high-molecular-weight multimers. Type 1 this mutation does not always result in low VWF levels and
VWD may be difficult to diagnose due to natural variations bleeding symptoms. Apparently, only the coinheritance of
in circulating VWF levels related to the influence of environ- blood group O, which has been linked with increased VWF
mental factors, such as exercise, thyroid hormone, estrogens, proteolysis, results in type 1 VWD in individuals with muta-
and ABO blood type. VWF levels are lower by as much as tion p.Tyr1584Cys. Although p.Tyr1584Cys is the mutation
30% in individuals with blood group O compared with indi- that has been found more often among the VWD type 1
viduals with other blood groups. patients investigated, there is still some debate regarding the
In the last decade, a few molecular epidemiological studies pathological role of this variant.
of about 350 index cases with type 1 VWD have significantly The third group of patients, with no identified mutations,
advanced our knowledge, in particular of the genetic basis remains to be clarified. It cannot be excluded that mutations
of the disorder. Direct sequencing analysis of the whole cod- in genes other than VWF engaged in VWF biosynthesis and
ing region and exon/intron boundaries, along with the pro- processing could be the cause of the reduced plasma levels of
moter region of the VWF gene, has allowed the identification VWF. However, in this group there is the highest percent-
of mutations in about 60–65% of VWD type 1 patients. Most age of subjects with blood group O, and these are usually
of these were missense mutations (70%), with the remain- patients with low borderline VWF levels, who should per-
der being splice-site mutations (12%), small insertions and haps not be classified as type 1 VWD patients. Indeed, in
deletions (8%; one quarter in-frame), nonsense mutations the National Heart, Lung, and Blood Institute, NIH guide-
(2%), and promoter changes (8%). Only 16% of the muta- lines, patients are classified as type 1 VWD only if their
tions cause null alleles. Therefore, haploinsufficiency result- VWF:Ag and VWF:RCo values are lower than 30 IU dl−1 .
ing from a heterozygous null allele results in reduced VWF Quite recently, three molecular epidemiological studies have
expression in a small proportion of cases. At variance with been published, each reporting a cohort of VWD patients
The molecular basis of von Willebrand disease 241

(type 1, 2, and 3) from a different country (Spain, France, and 3. In these cases the distinction between VWD type 3 (see
Portugal). In all three studies only type 1 VWD patients with later) and type 1 might be difficult, and the evaluation of the
VWF levels <30 IU dL−1 were included. Due to this selec- VWFpp level, detectable in type 1 but not in type 3 patients,
tion, the percentage of VWF mutations identified in type 1 could be helpful to establish the correct diagnosis.
patients was higher (89.2%, 96.4%, and 100%, respectively, in
the three populations) than that reported in previous studies
(about 65%). The VWD patients from the French study were Type 2 VWD
molecularly characterized using Sanger sequence and MLPA Type 2 VWD accounts for approximately 20–25% of all VWD
analysis, whereas both Spanish and Portuguese patients were cases, is caused by a variety of mechanisms, and is associated
characterized also using NGS analysis. Among the 161 type with a variety of specific defects, reflecting the multifunc-
1 patients molecularly characterized in the French study, half tional role of VWF and the complicated post-translational
were found to carry null alleles (49% truncating sequence processing of this moiety.
variations versus 51% of missense mutations), a percentage
much higher than that reported by previous population stud-
ies in type 1 VWD. For the authors, this finding might sug- Type 2A
gest that the penetrance of some truncating mutations qual- Type 2A is characterized by decreased VWF-dependent
ified as “recessive” may be variable, like in the case of the platelet adhesion due to the reduction or absence of high- and
exon 4–5 deletion identified recurrently in patients of British intermediate-molecular-weight multimers (Figure 18.5a).
white origin with either type 3 (in the homozygous or com- The bleeding diathesis is caused by a lack of the most bio-
pound heterozygous state) or type 1 (in the heterozygous logically active high-molecular-weight multimers. In the cur-
state) phenotype. However, further evaluations of these trun- rent classification of VWD, several different phenotypes, with
cating sequence variations (one-fourth of which are poten- these features, are designated VWD type 2A. However, these
tial splice-site mutations) are needed, along with the investi- distinct phenotypes, described in the previous classification
gation of patients’ family members, to better understand the of VWD as IIA, IIC, IID, and IIE, are due to different molec-
role of these mutations in type 1 VWD. ular pathomechanisms.
The term “severe” VWD has been used occasionally for The most common form of type 2A (originally designated
VWD type 1 characterized by very low VWF levels. However, VWD type IIA) is due to mutations in the A2 domain.
VWD type 1 due to dominant heterozygous mutations, with This variant is characterized by a lack of the VWF high-
few exceptions (e.g. p.Arg1205His), is rarely associated with and intermediate-molecular-weight multimers, markedly
VWF levels as low as 10 IU dl−1 . More often these “severe” pronounced satellite bands (Figure 18.5b), and dominant
VWD type 1 are due to two different mutations (homozygous inheritance. A similar pattern can also be seen in type 2B
or compound heterozygous state), similar to VWD type VWD patients, and the use of RIPA assay is required to

(a) (b)

HMW
multimer

N 2M 2A 2B N N 2A 2B 2A 2A N
(IIA) (IIC) (IIE)
Fig. 18.5 (a) Comparison, in non-reducing low-resolution gel, of plasma von Willebrand factor (VWF) multimeric structure from von Willebrand
disease (VWD) patients and normal subjects. Lanes 3 (VWD type 2A/IIA) and 4 (VWD type 2B) show the lack of high-molecular-weight (HMW)
multimers. Lane 2 (VWD type 2M) shows a normal multimeric pattern. Lanes 1 and 5 show a normal multimeric structure from healthy controls.
(b) Comparison, in non-reducing intermediate-resolution gel, of plasma VWF multimeric structure from VWD patients and normal subjects. Lanes
1 and 6 show a normal multimeric structure from healthy controls, with the typical triplet structure (brackets); the satellite bands are indicated by
the arrows. Lanes 2 (VWD type 2A/IIA) and 3 (VWD type 2B) show a markedly increased triplet structure, with enhanced satellite bands due to the
increased susceptibility of these variants to ADAMTS-13 proteolysis. Lane 4 (VWD type 2A/IIC) shows the absence of a triplet structure (only the
main band is present), whereas lane 5 (VWD type 2A/IIE) shows markedly reduced satellite bands.
242 Molecular Hematology

discriminate between these two variants. In type 2A patients, variant is characterized by a lack of high-molecular-weight
platelet-rich plasma agglutinates at a higher concentration multimers, absence of the triplet structure (only the main
of ristocetin (>1.2 mg ml−1 ; normal range 0.8–1.2 mg ml−1 ), band is present; Figure 18.5b), and recessive inheritance.
whereas in type 2B patients platelet-rich plasma agglutinates The VWF propeptide is required for the formation of VWF
at a lower concentration of ristocetin (<0.7 mg ml−1 ). multimers and the sorting of VWF to storage granules. The
Two mechanisms have been report to explain phenotype presence of these mutations in the propeptide compromises
2A, subclassified as group 1 and group 2. Group 1 mutations the multimerization process, and hence the lack of high-
lead to a VWF subunit that is improperly folded, retained molecular-weight multimers. Only a few mutations have
in the endoplasmic reticulum, and subsequently degraded. been identified, mainly in the D2 domain (e.g. p.Gly550Arg,
Multimers formed by the interactions between normal and p.Asn528Ser, and p.Cys623Trp). In the case of mutation
mutant subunits are retained in the cell. Therefore, the largest p.Asn528Ser, in vitro studies have also shown its role in pre-
multimers are more efficiently retained, accounting for the venting the intracellular transport of VWF multimer into the
characteristic type 2A multimer pattern. The defect of group storage organelles.
2 does not interfere with VWF secretion, but instead renders A rare type 2A variant (phenotype IID) is caused by muta-
the mutant subunit more susceptible to proteolytic cleavage tions localized in the CK domain at the carboxy-terminal
(between amino acids Y1605 and M1606 of the A2 domain) of VWF. This variant is characterized by a decreased level
by ADAMTS-13. This enhanced plasma proteolysis results of high-molecular-weight multimers, the presence of extra
in the selective loss of the largest VWF multimers typical of intervening bands between individual VWF oligomers, and
type 2A. However, more recently, in vitro expression stud- dominant inheritance. These mutations compromise the
ies have shown that group 1 mutations in the A2 domain pro-VWF dimerization process, therefore multimer analy-
also confer increased susceptibility to ADAMTS-13 cleavage, sis shows additional bands between the individual VWF
as confirmed by the pronounced satellite bands that these oligomers, representing multimers with an odd number of
patients shown in their plasma VWF multimers. Over 50 dis- monomers. The mutations mainly involve cysteine residue
tinct mutations have been reported in the A2 VWF struc- (p.Cys2771Tyr, p.Cys2771Ser, and p.Cys2773Arg), although
tural domain, the most frequent being p.Arg1597Trp and an out-of-frame deletion has also been identified in exon 51.
p.Ile1628Thr.
The second most common type 2A variant (pheno-
type IIE) is caused by mutations in the D3 domain. This
Type 2B
variant is characterized by a decreased level, more rarely
a lack, of high-molecular-weight multimers, reduced or The VWF 2B variant shows increased affinity for platelet
absent satellite bands (Figure 18.5b), and dominant inher- GPIb, therefore the diagnosis of type 2B VWD patients
itance. A large group of VWD type 2A (IIE) patients from requires the use of RIPA assay for these patients’ results to
Germany has been characterized on the basis of their plasma be enhanced (see earlier). Type 2B VWF is usually associ-
VWF multimeric structure, and most of the mutations identi- ated with the absence of high-molecular-weight multimers in
fied involve a cysteine residue (p.Cys1130Arg, p.Cys1130Trp, circulating plasma but not in platelets (Figure 18.5a). There-
p.Tyr1146Cys, p.Cys1153Tyr, p.Cys1173Phe). The majority fore, type 2B mutations do not impair the assembly of large
of these mutations correlate with a moderate to severe secre- VWF multimers, but after secretion the VWF binds spon-
tion defect and compromised the VWF multimerization pro- taneously to platelets, leading to low plasma levels. In addi-
cess, hence the lack/decreased level of high-molecular-weight tion, the spontaneous interaction of type 2B VWF with GPIb
multimers. In addition, due to the position of the muta- accelerates the cleavage of VWF by ADAMTS-13, resulting
tions in the D3 domain, some of these variants were also in the further depletion of the high-molecular-weight mul-
found associated with modestly reduced VWF:FVIIIB. The timers along with the enhancement of satellite bands (Fig-
important proportion of type 2A (IIE) variants among type ure 18.5b). The variable thrombocytopenia which may be
2A patients has been recently underlined also by the inves- present in these patients is believed to be secondary to the for-
tigation of the French VWD cohort (42 type 2A/IIE cases mation of platelet–VWF aggregates. However, in vitro stud-
out of 122 type 2A patients). The mutation spectrum of ies have shown that the presence of type 2B VWF influences
type 2A (IIE) partly overlapped with VWD type 1 (see ear- megakaryocyte maturation, causing the formation of giant
lier), which points to the problem of interindividual phe- platelets, platelets with altered morphology, and reducing the
notypic differences in spite of the same VWF genotype, or number of platelet produced, suggesting also a direct role of
problems in the performance of non-standardized multimer type 2B VWF in megakaryocytopoiesis.
analysis. Type 2B is inherited as a dominant trait. Mutations respon-
A rarer type 2A variant (phenotype IIC) is due to muta- sible for this subtype are mainly in the A1 domain (or at the
tions in the VWF propeptide (D1 and D2 domains). This D3–A1 junction), which binds to the GPIb receptor. With
The molecular basis of von Willebrand disease 243

a few exceptions, all mutations result in amino acid substi- type 2M VWD due to mutations in the A1 domain, and they
tutions, and a few of them (p.Arg1306Trp, p.Arg1308Cys, seem also to have milder bleeding symptoms.
p.Val1316Met, and p.Arg1341Gln) account for about 80% of
the reported cases.
Type 2N
Type 2B VWD has sometimes been misdiagnosed and
treated as autoimmune thrombocytopenia. Thrombocytope- Type 2N VWD refers to all the qualitative variants character-
nia and the absence of the larger multimers are not con- ized by decreased affinity for FVIII. The first description of
stantly present in type 2B VWD, however. For instance, this type of variant was of a patient from Normandy (hence
mutation p.Pro1266Leu causes an increased GPIb binding the N). This subtype is characterized by low levels of FVIII;
with no apparent loss of large multimers and an absence however, VWF levels are usually normal and have an intact
of thrombocytopenia before and after desmopressin, lead- VWF multimeric pattern. It is difficult to distinguish type 2N
ing to earlier classification among type 1 variants (type 1 from mild hemophilia A. Hemophilia is an X-linked disease,
New York or type 1 Malmo). Patients carrying this muta- whereas type 2N VWD is an autosomal recessive disease. A
tion have usually mild bleeding symptoms and in some cases definite diagnosis can be made by demonstrating the reduced
may have none. Pseudo-VWD, also known as platelet-type binding of FVIII to VWF with assays exploring this VWF
VWD, is a rare platelet disorder due to gain-of-function property (i.e. VWF:FVIIIB). Since the majority of mutations
mutations in the platelet receptor GPIb (e.g. p.Gly233Val and are located in the N-terminus of the mature VWF subunit
p.Met239Val). These mutations cause platelet GPIb to bind (D′ –D3), diagnosis can be confirmed by screening for muta-
spontaneously to VWF, resulting, as for VWD type 2B, in tions from exons 18–25. Three mutations (p.Thr791Met,
enhanced RIPA. Either platelet/plasma mixing studies (per- p.Arg816Trp, and p.Arg854Gln) account for 90% of the type
forming RIPA assay also using patient plasma with normal 2N mutations.
platelets and vice versa) or genetic analysis of VWF and
GPIBA genes are necessary to perform the differential diag-
Type 3 VWD
nosis.
Type 3 VWD is an autosomal recessive, clinically severe
disorder characterized by the complete or nearly complete
Type 2M
absence of VWF (<5 IU dl−1 ) in both plasma and platelets.
Type 2M VWD refers to qualitative variants with decreased As in type 2N VWD, there is a secondary deficiency of FVIII,
VWF-dependent platelet adhesion not caused by the absence and patients therefore present with a double defect of pri-
of the high-molecular-weight multimers (Figure 18.5a). In mary hemostasis and intrinsic coagulation. Type 3 VWD is
type 2M VWF a functional defect is caused by mutations that rare and accounts for less than 5% of all VWD cases, with
compromise VWF binding to platelets or to subendothelium. a prevalence in the general population of 0.5–1 per million.
In the first case, type 2M is characterized by a VWF:RCo The parents of type 3 patients are obligatory heterozygotes
that is disproportionately low compared with VWF:Ag, and in most cases are asymptomatic.
in spite of the presence of a normal multimer distribution Because of the large size of the gene, the presence of
(hence M for multimer). This variant is inherited as a domi- the pseudogene, the low prevalence of the disease, and the
nant trait and the mutations are identified in the A1 domain. absence of a specific location of the mutations, the charac-
These defects, which in most cases are missense mutations terization of the molecular defects in type 3 has been rela-
(e.g. p.Arg1315Cys, p.Tyr1321Cys, and p.Gly1324Ser), seem tively slow. At first, only large gene deletions were identified
to downregulate the binding of the A1 domain to the GPIb using Southern blot analysis. Subsequently, the use of screen-
platelet receptor. Therefore, in RIPA assay, platelet-rich ing methods such as single-stranded conformational poly-
plasma of these patients agglutinates at a concentration of morphisms, chemical cleavage mismatch analysis, and con-
ristocetin >1.2 mg ml−1 (normal range 0.8–1.2 mg ml−1 ), formational sensitive gel electrophoresis, along with DNA
similar to type 2A patients. Mutations p.Arg1374Cys and sequence analysis, allowed the identification of mutations in
p.Arg1374His have been reported often as type 2M variants, most patients.
but their classification as type 2M has been controversial. The majority of the defects identified in type 3 VWD cause
In the second case, type 2M VWD is characterized by a dis- null alleles. Among these, nonsense mutations are the most
proportionately low VWF:CB compared with VWF:Ag and a common, followed by small deletions, splice-site mutations,
normal multimer distribution. Also this variant is inherited small insertions, and large deletions. Nevertheless, almost
as a dominant trait and the mutations causing VWF colla- a quarter of the mutations identified in these patients were
gen binding defect have been found mainly in the A3 domain missense. Among the nonsense mutations, a few hotspot
(e.g. p.Ser1731Thr and p.Met1761Lys). Patients with these mutations at the arginine codons (p.Arg365*, p.Arg1659*,
mutations have been reported more rarely than patients with p.Arg1853*, and p.Arg2535*) have been found repeatedly in
244 Molecular Hematology

different populations. A single cytosine deletion, in a stretch The advantage of this compound is that it is relatively
of 6 cytosines in exon 18, appears to be particularly com- inexpensive and carries no risk of transmitting blood-
mon in patients from Sweden and Germany. In the Hungar- borne infectious agents. When infused intravenously over
ian population, a large deletion, including exons 1, 2, and 30 minutes at a dose of 0.2–0.3 μg kg−1 , desmopressin is
3, was found in about 25% of the unrelated type 3 VWD expected to increase plasma FVIII and VWF two- to four-
patients investigated. Among a group of 42 type 3 patients fold above basal levels. In general, high FVIII/VWF con-
from Spain, the mutation p.Gln1311*, derived from a poten- centrations last in plasma for at least 8–10 hours and infu-
tial gene conversion, was significantly recurrent (6 patients sions can be repeated every 12–24 hours, depending on the
out 42). The same mutation was also identified among type 3 type and severity of the bleeding episode. At the time of
patients from Portugal (5 out of 15 families). In both studies diagnosis, a DDAVP infusion test should be performed to
this mutation was identified among Spanish Romani fami- evaluate individual response patterns. Patients with baseline
lies. Patients with type 3 VWD may develop allo-antibodies plasma levels of FVIII/VWF measurements in the range of
to VWF, which render replacement therapy ineffective. This 10–20 IU dl−1 or more are those who are more likely to reach
complication is strongly associated with the presence of post-desmopressin levels sufficient to attain hemostasis, tak-
homozygous large gene deletions, although a few cases have ing into account variables such as the type and severity of the
been reported due to nonsense mutations. bleeding episode and the levels of FVIII/VWF that must be
attained and maintained to secure hemostasis. Even though
most patients with mild hemophilia A treated repeatedly with
desmopressin become less responsive to therapy, this prob-
Treatment of VWD
lem is less frequent and prominent in patients with type
1 VWD. The drug is also available in concentrated forms
The aim of therapy in VWD is to correct both hemosta-
for subcutaneous and intranasal administration (at doses of
sis defects: the decreased VWF-dependent platelet adhesion
0.3 μg kg−1 and 150–300 μg, respectively), which can be con-
(primary hemostasis) and the reduced FVIII level due to
venient for home treatment.
the VWF deficiency or dysfunction (intrinsic coagulation).
Side effects of desmopressin are usually mild tachycar-
The main treatment options in order to stop spontaneous
dia, headache, and facial flushing. Hyponatremia and volume
bleeding or to prevent bleeding at the time of surgical pro-
overload due to the antidiuretic effect of desmopressin are
cedures are autologous replacement therapy with desmo-
relatively rare if fluid intake is not excessive during treatment.
pressin (1-deamino-8-D-arginine vasopressin, DDAVP) and
Even though no thrombotic episodes have been reported in
allogeneic replacement therapy with plasma-derived concen-
VWD patients treated with desmopressin, this compound
trates FVIII/VWF or recombinant VWF. Ancillary forms
should be used with caution in elderly patients with cardio-
of treatment are platelet concentrates, synthetic fibrinoly-
vascular disease, because a few cases of myocardial infarction
sis inhibitors, and combined estrogen/progestogen prepa-
and stroke have occurred in treated patients with hemophilia
rations, which in some clinical situations are adjunctive or
and uremia.
sometimes alternative to the main treatments.
Desmopressin is particularly effective in patients with type
1 VWD. A prospective clinical study reported that in 77
patients with type 1, a complete response was observed in
Desmopressin
83%, partial response in 13%, and no response in only 4% of
Desmopressin is a synthetic analogue of the antidiuretic patients. In other VWD types, responsiveness is less certain
hormone vasopressin that, when administered to healthy (Table 18.2). In fact, in a different prospective trial only 18%
volunteers or patients with mild hemophilia and VWD, tran- of the type 2 VWD patients were responsive. In this study,
siently increases FVIII and VWF by releasing these moieties
from storage sites into plasma. Endothelial cell Weibel– Table 18.2 Indications for desmopressin in different types of von
Palade bodies appear to be the source of both VWF and Willebrand disease
FVIII. However, VWF is constitutively present in Weibel–
Palade bodies (the generation of these organelles is strictly Type Response
VWF dependent), whereas FVIII (which is mainly synthe-
sized in the liver) seems to be present only in a select subset of 1 Usually effective
endothelial cells (lung, heart, intestine, skin, and pulmonary 2A Usually ineffective
2B May be contraindicated
artery). Desmopressin induces VWF release into plasma by
2M Usually ineffective
binding to the vasopressin V2 receptor and thereby activat-
2N Rarely effective
ing cyclic AMP-mediated signaling in vascular endothelial 3 Ineffective
cells.
The molecular basis of von Willebrand disease 245

only one out of 15 type 2A patients was responsive to DDAVP. pasteurization. Another largely diffused product has
A similar result was obtained in type 2M, where DDAVP was the same VWF:RCo/VWF:Ag ratio (0.8–0.9), but is different
effective in only 3 patients out of 21. In type 2N FVIII:C lev- because it also contains similar relative amounts of VWF and
els increase following desmopressin, but released FVIII cir- FVIII (VWF:RCo/FVIII:C ratio 1.2). Two virucidal meth-
culates for a relatively short time in patients’ plasma, because ods, solvent/detergent and heating at high temperatures,
the stabilizing effect of VWF on FVIII is impaired. Therefore, are included in the manufacturing stage of this product.
plasma concentrates containing FVIII and VWF are usually Indeed, most of the plasma-derived products adopted these
preferable. In type 2B patients, desmopressin releases VWF methods with the goal of inactivating both enveloped and
from the endothelial cells. However, there is only a transient non-enveloped viruses.
increased of VWF plasma level, because the ultra-large VWF One unique product is a chromatography-purified VWF
multimers of this gain-of-function variant spontaneously concentrate with a very low FVIII content (VWF:RCo/
agglutinate platelets, worsening or causing patients’ throm- FVIII:C ratio 60). As many as three viral inactivation meth-
bocytopenia. Therefore, it is unclear if the use of desmo- ods are used in the production of this concentrate (sol-
pressin can provide clinical benefit in type 2B patients. How- vent/detergent, heating, and nanofiltration). The recently
ever, mild type 2B patients who present a full set of multi- commercialized product containing human recombinant
mers, such as those carrying mutation p.Pro1266Leu, have VWF is obtained by a genetically engineered cell line,
been treated with desmopressin without any complications. therefore there is virtually no risk of blood-borne virus
Patients with type 3 VWD are unresponsive to desmopressin, transmission. Recombinant VWF, similar to endothelial
because they lack releasable stores of VWF. cell VWF, presents ultra-large multimers, whereas most
plasma-derived products are deficient in high-molecular-
weight multimers. The presence of ultra-large multimers,
Transfusional therapies
along with its high purity, confer a higher specific activity
If desmopressin therapy is not effective or its use is con- (VWF:RCo/VWF:Ag >1) and a stronger hemostatic poten-
traindicated, transfusion therapy with FVIII/VWF concen- tial to this product in comparison to plasma-derived concen-
trates is the treatment of choice to prevent or stop bleed- trates, so it could be used in crucial bleeding situations (e.g.
ing. FVIII and VWF may be infused as fresh frozen plasma VWD associated with gastrointestinal angiodysplasia) where
(FFP), but the large volumes required severely limit its traditional FVIII/VWF products seem to be less effective.
use. Cryoprecipitate contains 5–10 times more FVIII and The dosages recommended for the control or prevention
VWF than FFP (each bag contains approximately 80–100 of bleeding are summarized in Table 18.3. Since in VWD
IU). Early studies indicated that cryoprecipitate adminis- patients FVIII has a longer half-life than in patients with
tered every 12–24 hours normalized plasma FVIII levels hemophilia A (20–24 vs. 12–14 hours), the infusion of one
and stopped or prevented bleeding in VWD. On the basis daily dose is sufficient to reach and maintain adequate plasma
of these observations, cryoprecipitate has been the main-
stay of treatment for many years. However, virucidal meth-
ods cannot be applied to cryoprecipitate produced by blood Table 18.3 Dosages of VWF:RCo/FVIII:C recommended in patients
banks, so that this product carries a small risk of transmitting with von Willebrand disease treated with FVIII/VWF concentrates
blood-borne infectious agents. Therefore, virus-inactivated
FVIII/VWF concentrates, originally developed for the treat- Type of Dose Number of Target plasma
ment of hemophilia A, were perceived as safer and were bleeding (IU kg−1 ) infusions VWF:RCo/FVIII:C level
preferred in the management of VWD patients. Nowadays,
several plasma-derived products are licensed for the treat- Major surgery 40–60 Every 12 hours 50–100 IU dl−1 ,
initially then maintain level until
ment of VWD. Moreover, quite recently, a recombinant VWF
once a day healing is complete
product has been licensed in the USA for the treatment of
(3–10 days)
adult VWD patients. All products licensed for VWD treat- Minor surgery 30–50 Once a day >30 IU dl−1 until
ment have been evaluated for safety and efficacy in prospec- healing is complete
tive clinical trials. However, some of these products have been (may require
more extensively evaluated than others and some present 1–3 days)
unique characteristics. Dental 20–30 Single dose >30 IU dl−1 for
One of the most widely used concentrates in the treatment extractions prior to >12 hours
of VWD contains relatively larger amounts of VWF than procedure
of FVIII (VWF:RCo/FVIII:C ratio 2.4) and has been used Spontaneous 20–60 Once a day > 30 IU dl−1 until
in treatment for the last 25 years. The VWF:RCo/VWF:Ag or traumatic bleeding stops
bleeding
ratio is 0.8 and the virucidal method adopted is
246 Molecular Hematology

levels for the treatment of spontaneous bleeding episodes


Table 18.4 Summary of management of different types and sub-
and to prevent excessive bleeding. The doses of this concen- types of von Willebrand disease (VWD)
trate recommended for their demonstrated efficacy in large,
prospective clinical trials are 40–60 IU kg−1 of VWF:RCo Treatment of Alternative or
(50–75 IU kg−1 in children because of the lower in vivo recov- choice adjunctive therapy
ery), which usually results in VWF:RCo plasma levels of 80–
120 IU dl−1 or higher. Even though the plasma half-life of Type 1 Desmopressin Antifibrinolytic amino
VWF:RCo is much shorter than that of FVIII:C (8–10 vs. (factor VIII/VWF acids
concentrates)
20–24 hours), usually these doses do not need to be repeated
Type 2A Factor VIII/VWF As above
more often than every 24 hours, although sometimes treat-
concentrates
ment intervals must be tailored to the clinical situation. (Desmopressina )
It is usually not necessary to carry out laboratory tests to Type 2B Factor VIII/VWF As above
monitor replacement therapy in patients with spontaneous concentrates
bleeding episodes. For surgical procedures we recommend Type 2M Factor VIII/VWF As above
measuring FVIII:C and VWF:RCo every 12 hours on the concentrates
operation day and then every 24 hours. Major surgical pro- (Desmopressina )
cedures are successfully carried out and spontaneous bleed- Type 2N Factor VIII/VWF As above
ing episodes controlled following the infusion of FVIII/VWF concentrates
concentrates. In the relatively rare instances when bleeding is (Desmopressina )
Type 3 Factor VIII/VWF Antifibrinolytic amino
not controlled, platelet concentrates (given immediately after
concentrates acids, platelet
FVIII/VWF-containing preparations, at doses of 4–5 × 1011
concentrates
platelets) are effective, particularly in patients with type 3 Type 3 Recombinant Recombinant activated
VWD. Platelets from type 3 VWD patients completely lack complicated by factor VIII factor VII
VWF and there is no uptake of the protein from plasma allo-antibodies
after the infusion of concentrates. The hemostatic effective-
a 1-deamino-8-D-arginine vasopressin (DDAVP) should also be trialed
ness of the transfusion of normal platelets is likely to be due
to the fact that these cells transport and localize ultra-large for types 2A, 2M, and 2N VWD patients, even though DDAVP will
likely be insufficiently effective for most of these cases and therefore
VWF at sites of vascular injury. From a practical standpoint,
FVIII/VWF concentrates will be required.
it must be emphasized that in one of the largest prospec-
tive study carried out in VWD patients, platelet concen-
trates became necessary to prevent or stop bleeding in one
case only. phenotypically, because the choice of treatment must be
To avoid the risk of a life-threatening anaphylactic reac- tailored to the different types of the disease. Such character-
tion, type 3 VWD patients who develop allo-antibodies ization is not simple, so in the majority of clinical centers it
against VWF must not be treated with products contain- is probably not worth setting up relatively complicated tests
ing VWF. Instead, treatment can be done using continuous such as multimer analysis assay, when samples can be sent
intravenous infusion of recombinant FVIII or using recom- for analysis to more expert laboratories that have become
binant activated FVII. Venous thromboembolic episodes sel- proficient during the study of large series of patients.
dom occur during repeated FVIII/VWF concentrate infu-
sions. As mentioned earlier, therapy should be monitored
von Willebrand disease resources on the
daily, also to avoid reaching very high FVIII plasma levels,
internet
known to be a risk factor for venous thromboembolism.
International Society on Thrombosis and Haemostasis-
Scientific and Standardisation Committee of von Willebrand
Conclusion Factor (ISTH-SSC). von Willebrand Factor Online Database
(VWFdb). Available at: http://www.vwf.group.shef.ac.uk/
The different options currently available for the management vwd.html
of VWD are summarized in Table 18.4. Treatment of spon- European Association for Haemophilia and Allied Dis-
taneous bleeding episodes and their prevention at the time orders (EAHAD) Coagulation Factor Variant Databases.
of invasive procedures are relatively simple and can certainly Available at: https://grenada.lumc.nl/LOVD2/VWF/home.
be tackled by the average clinical hematologist with access php?select_db=VWF
to a minimum of laboratory testing (FVIII:C and VWF:RCo OMIMTM Online Mendelian Inheritance in Man. Avail-
assays). However, patients need to be well characterized able at: http://omim.org/entry/193400
The molecular basis of von Willebrand disease 247

Sadler, J.E. (1994). A revised classification of von Willebrand disease.


Further reading Thromb. Haemost. 71: 520–525.
Sadler, J.E., Budde, U., Eikenboom, J.C.J. et al. (2006). Update on
General the pathophysiology and classification of von Willebrand disease: a
report of the subcommittee on von Willebrand factor. J. Thromb.
Nichols, W.L., Hultin, M.B., James, A.H. et al. (2008). von Willebrand
Haemost. 4: 1–12.
disease (VWD): evidence-based diagnosis and management guide-
lines, the National, Heart, Lung and Blood Institute (NHLBI) Expert
Panel report (USA). Haemophilia 14: 171–232.
Sadler, J.E., Mannucci, P.M., Berntorp, E. et al. (2000). Impact, diagnosis
Genetic defects in VWD
and treatment of von Willebrand disease. Thromb. Haemost. 84: 160– Acquila, M., Bottini, F., DI Duca, M. et al. (2009). Multiplex ligation-
174. dependent probe amplification to detect a large deletion within the
von Willebrand gene. Haemophilia 15: 1346–1348.
von Willebrand factor Baronciani, L., Federici, A.B., Castaman, G. et al. (2008). Prevalence
of type 2B “Malmo/New York” von Willebrand disease in Italy: the
Dong, Z., Thoma, R.S., Crimmins, D.L. et al. (1994). Disulfide bonds role of von Willebrand factor gene conversion. J. Thromb. Haemost.
required to assemble functional von Willebrand factor multimers. J. 6: 887–890.
Biol. Chem. 269: 6753–6758. Baronciani, L., Federici, A.B., and Eikenboom, J.C.J. (2011). Clinical,
Fujikawa, K., Suzuki, H., McMullen, B., and Chung, D. (2001). Purifica- laboratory, and molecular markers of type 3 von Willebrand disease.
tion of human von Willebrand factor cleaving protease and its iden- In: Von Willebrand disease: basic and clinical aspects (eds. A.B. Fed-
tification as a new member of the metalloproteinase family. Blood 98: erici, C.A. Lee, E. Berntop, et al.), 148–165. Wiley-Blackwell.
1662–1666. Batlle, J., Pérez-Rodrı́guez, A., Corrales, I. et al. (2015). Molecular and
Ginsburg, D., Handin, R.I., Bonthron, D.T. et al. (1985). Human von clinical profile of von Willebrand disease in Spain (PCM–EVW–ES):
Willebrand factor (vWF): isolation of complementary DNA (cDNA) proposal for a new diagnostic paradigm. Thromb. Haemost. 2: 1–11.
clones and chromosomal localization. Science 228: 1401–1406. Bodo, I., Katsumi, A., Tuley, E.A. et al. (2001). Type 1 von Wille-
Kroll, M.H., Hellums, J.D., McIntire, L.V. et al. (1996). Platelets and brand disease mutation Cys 1149Arg causes intracellular retention
shear stress. Blood 88: 1525–1541. and degradation of heterodimers: a possible general mechanism for
Lenting, P.J., Christophe, O.D., and Denis, C.V. (2015). von Willebrand dominant mutations of oligomeric proteins. Blood 98: 2973–2979.
factor biosynthesis, secretion, and clearance: connecting the far ends. Bowen, D.J. and Collins, P.W. (2006). Insights into von Willebrand factor
Blood 125: 2019–2028. proteolysis: clinical implications. Br. J. Haematol. 133: 457–467.
Mancuso, D.J., Tuley, E.A., Westfield, L.A. et al. (1989). Structure of the Corrales, I., Catarino, S., Ayats, J. et al. (2012). High-throughput molec-
gene for human von Willebrand factor. J. Biol. Chem. 264: 19514– ular diagnosis of von Willebrand disease by next generation sequenc-
19527. ing methods. Haematologica 97: 1003–1007.
Mancuso, D.J., Tuley, E.A., Westfield, L.A. et al. (1991). Human von Cumming, A., Grundy, P., Keeney, S. et al. (2006). An investigation of
Willebrand factor gene and pseudogene: structural analysis and dif- the von Willebrand factor genotype in UK patients diagnosed to have
ferentiation by polymerase chain reaction. Biochemistry 30: 253–269. type 1 von Willebrand disease. Thromb. Haemost. 96: 630–641.
Shelton-Inloes, B.B., Titani, K., and Sadler, J.E. (1986). cDNA sequences Eikenboom, J., Hilbert, L., Ribba, A.S. et al. (2009). Expression of 14
for human von Willebrand factor reveal five types of repeated von Willebrand factor mutations identified in patients with type 1
domains and five possible protein sequence polymorphisms. Bio- von Willebrand disease from the MCMDM-1VWD study. J. Thromb.
chemistry 25: 3164–3171. Haemost. 7: 1304–1312.
Wagner, D.D. and Marder, V.J. (1984). Biosynthesis of von Willebrand Eikenboom, J.C.J., Matsushita, T., Reitsma, P.H. et al. (1996). Dominant
protein by human endothelial cells: processing steps and their intra- type I von Willebrand disease caused by mutated cysteine residues in
cellular localization. J. Cell Biol. 99: 2123–2130. the D3 domain of von Willebrand factor. Blood 88: 2433–2441.
Weiss, H.J., Sussman, I.I., and Hoyer, L.W. (1977). Stabilization of factor Enayat, M.S., Guilliatt, A.M., Surdhar, G.K. et al. (2001). Aberrant
VIII in plasma by the von Willebrand factor. Studies on posttransfu- dimerization of von Willebrand factor as the result of mutations in
sion and dissociated factor VIII and in patients with von Willebrand’s the carboxy-terminal region: identification of 3 mutations in mem-
disease. J. Clin. Invest. 60: 390–404. bers of 3 different families with type 2A (phenotype IID) von Wille-
Zhou, Y.F., Eng, E.T., Zhu, J. et al. (2012). Sequence and structure rela- brand disease. Blood 98: 674–680.
tionships within von Willebrand factor. Blood 120: 449–458. Federici, A.B., Mannucci, P.M., Castaman, G. et al. (2009). Clinical and
molecular predictors of thrombocytopenia and risk of bleeding in
von Willebrand disease and its patients with von Willebrand disease type 2B: a cohort study of 67
classification patients. Blood 113: 526–534.
Fidalgo, T., Salvado, R., Corrales, I. et al. (2016). Genotype–phenotype
De Jong, A. and Eikenboom, J. (2016). Developments in the diagnostic correlation in a cohort of Portuguese patients comprising the entire
procedures for von Willebrand disease. J. Thromb. Haemost. 14: 449– spectrum of VWD types: impact of NGS. Thromb. Haemost. 116: 17–
460. 31.
Federici, A.B. (1998). Diagnosis of von Willebrand disease. Haemophilia Gadisseur, A., van der Planken, M., Schroyens, W. et al. (2009). Domi-
4: 654–660. nant von Willebrand disease type 2M and 2U are variable expressions
248 Molecular Hematology

of one distinct disease entity caused by loss-of-function mutations in classification. High frequency in the northwest of Spain (Galicia).
the A1 domain of the von Willebrand factor gene. Acta Haematol. Am. J. Hematol. 80: 188–196.
121: 145–153. Richards, S., Aziz, N., Bale, S. et al. (2015). ACMG Standards and Guide-
Gezsi, A., Budde, U., Deak, I. et al. (2010). Accelerated clearance alone lines. Standards and guidelines for the interpretation of sequence
explains ultra-large multimers in von Willebrand disease Vicenza. J. variants: a joint consensus recommendation of the American College
Thromb. Haemost. 8: 1273–1280. of Medical Genetics and Genomics and the Association for Molecular
Goodeve, A., Eikenboom, J.C.J., Castaman, G. et al. (2007). Phenotype Pathology. Genet. Med. 17: 405–424.
and genotype of a cohort of families historically diagnosed with type Ruggeri, Z.M., Pareti, F.I., Mannucci, P.M. et al. (1980). Heightened
1 von Willebrand disease in the European study, Molecular and Clin- interaction between platelets and factor VIII/von Willebrand factor
ical Markers for the Diagnosis and Management of Type 1 von Wille- in a new subtype of von Willebrand disease. N. Engl. J. Med. 302 (19):
brand disease (MCMDM-1VWD). Blood 109: 112–121. 1047–1051.
Haberichter, S.L., Balistreri, M., Christopherson, P. et al. (2006). Assay Schneppenheim, R., Michiels, J.J., Obser, T. et al. (2010). A cluster of
of the von Willebrand factor (VWF) propeptide to identify patients mutations in the D3 domain of von Willebrand factor correlates with
with type 1 von Willebrand disease with decreased VWF survival. a distinct subgroup of von Willebrand disease: type 2A/IIE. Blood
Blood 108: 3344–3351. 115: 4894–4901.
Haberichter, S.L., Budde, U., Obser, T. et al. (2010). The mutation N528S Sutherland, M.S., Cumming, A.M., Bowman, M. et al. (2009). A novel
in the von Willebrand factor (VWF) propeptide causes defective mul- deletion mutation is recurrent in von Willebrand disease types 1 and
timerization and storage of VWF. Blood 115: 4580–4587. 3. Blood 114: 1091–1098.
Hassenpflug, W.A., Budde, U., Obser, T. et al. (2006). Impact of muta- Veyradier, A., Boisseau, P., Fressinaud, E. et al. (2016). A laboratory
tions in the von Willebrand factor A2 domain on ADAMTS13- phenotype/genotype correlation of 1167 French patients from 670
dependent proteolysis. Blood 107: 2339–2345. families with von Willebrand disease: a new epidemiologic picture.
James, P.D., Notley, C., Hegadom, C. et al. (2007). The mutational spec- Medicine (Baltimore) 95: e3038.
trum of type 1 von Willebrand disease: results from a Canadian Yadegari, H., Driesen, J., Pavlova, A. et al. (2012). Mutation distribution
cohort study. Blood 109: 145–154. in the von Willebrand factor gene related to the different von Wille-
Keeling, D., Beavis, J., Marr, R. et al. (2012). A family with type 2M VWD brand disease (VWD) types in a cohort of VWD patients. Thromb.
with normal VWF:RCo but reduced VWF:CB and a M1761K muta- Haemost. 108: 662–671.
tion in the A3 domain. Haemophilia 18: e33–e2516. Zimmerman, T.S., Dent, J.A., Ruggeri, Z.M., and Nannini, L.H. (1986).
Lyons, S.E., Bruck, M.E., Bowie, E.J.W., and Ginsburg, D. (1992). Subunit composition of plasma von Willebrand factor. Cleavage is
Impaired intracellular transport produced by a subset of type IIA von present in normal individuals, increased in IIA and IIB von Wille-
Willebrand disease mutations. J. Biolumin. Chemilumin. 267: 4424– brand disease, but minimal in variants with aberrant structure of
4430. individual oligomers (types IIC, IID, and IIE). J. Clin. Invest. 77 (3):
Mannucci, P.M., Bloom, A.L., Larrieu, M.J. et al. (1984). Atherosclerosis 947–951.
and von Willebrand factor. I. Prevalence of severe von Willebrand’s
disease in western Europe and Israel. Br. J. Haematol. 57: 163–169. Treatment of VWD
Mannucci, P.M., Lombardi, R., Castaman, G. et al. (1988). von Wille-
brand disease “Vicenza” with larger-than-normal (supranormal) von Berntorp, E., Archey, W., Auerswald, G. et al. (2008). A systematic
Willebrand factor multimers. Blood 71: 65–70. overview of the first pasteurised VWF/FVIII medicinal product, Hae-
Mazurier, C. and Meyer, D. (1996). Factor VIII binding assay of von mate P/Humate-P: history and clinical performance. Eur. J. Haema-
Willebrand factor and the diagnosis of type 2N von Willebrand dis- tol. Suppl. 80 (Suppl.70): 3–35.
ease – results of an international survey. On behalf of the subcom- Berntorp, E., Windyga, J., and European Wilate Study Group (2009).
mittee on von Willebrand Factor of the Scientific and Standardization Treatment and prevention of acute bleedings in von Willebrand dis-
Committee of the ISTH. Thromb. Haemost. 76: 270–274. ease – efficacy and safety of Wilate, a new generation von Willebrand
Meyer, D., Fressinaud, E., Gaucher, C. et al. (1997). Gene defects in factor/factor VIII concentrate. Haemophilia 15: 122–130.
150 unrelated French cases with Type 2 von Willebrand disease: from Bond, L. and Bevan, D. (1988). Myocardial infarction in a patient with
patient to the gene. Thromb. Haemost. 78: 451–456. hemophilia treated with DDAVP (letter). N. Engl. J. Med. 318: 121.
Nurden, P., Gobbi, G., Nurden, A. et al. (2010). Abnormal VWF mod- Borel-Derlon, A., Federici, A.B., Roussel-Robert, V. et al. (2007). Treat-
ifies megakaryocytopoiesis: studies of platelets and megakaryocyte ment of severe von Willebrand disease with a high-purity von
cultures from patients with von Willebrand disease type 2B. Blood Willebrand factor concentrate (Wilfactin): a prospective study of 50
115: 2649–2656. patients. J. Thromb. Haemost. 5: 1115–1124.
O’Brien, L.A., James, P.D., Othman, M. et al. (2003). Founder von Wille- Brooker, M. (2012). Registry of clotting factor concentrates, 9e. Montréal:
brand factor haplotype associated with type 1 von Willebrand disease. World Federation of Hemophilia available from: www.wfh.org.
Blood 102: 549–557. Castaman, G., Coppola, A., Zanon, E. et al. (2013). Efficacy and safety
Othman, M. and Emsley, J. (2014). Platelet-type von Willebrand disease: during formulation switch of a pasteurized VWF/FVIII concentrate:
toward an improved understanding of the “sticky situation.”. Semin. results from an Italian prospective observational study in patients
Thromb. Hemost. 40: 146–150. with von Willebrand disease. Haemophilia 19: 82–88.
Penas, N., Perez-Rodriguez, A., Torea, J.H. et al. (2005). Von Wille- Castaman, G., Lethagen, S., Federici, A.B. et al. (2008). Response to
brand disease R1374C: type 2A or 2M? A challenge to the revised desmopressin is influenced by the genotype and phenotype in type
The molecular basis of von Willebrand disease 249

1 von Willebrand disease (VWD): results from the European Study Mannucci, P.M. (2004). Treatment of von Willebrand disease. N. Engl.
MCMDM-1VWD. Blood 111: 3531–3539. J. Med. 351: 683–694.
Castillo, R., Monteagudo, J., Escolar, G. et al. (1991). Hemostatic effect of Mannucci, P.M., Bettega, D., and Cattaneo, M. (1992). Patterns of devel-
normal platelet transfusion in severe von Willebrand disease patients. opment of tachyphylaxis in patients with haemophilia and von Wille-
Blood 77: 1901–1905. brand disease after repeated doses of desmopressin (DDAVP). Br. J.
Chang, A.C., Rick, M.E., Ross, P.L., and Weinstein, M.J. (1998). Sum- Haematol. 82: 87–93.
mary of a workshop on potency and dosage of von Willebrand factor Mannucci, P.M., Chediak, J., Hanna, W. et al. (2002). Treatment of von
concentrates. Haemophilia 4 (Suppl 3): 1–6. Willebrand’s disease with a high-purity factor VIII/von Willebrand
Coppola, A., Franchini, M., Makris, M. et al. (2012). Thrombotic factor concentrate: a prospective, multicenter study. Blood 99: 450–
adverse events to coagulation factor concentrates for treatment of 456.
patients with haemophilia and von Willebrand disease: a systematic Mannucci, P.M., Franchini, M., and Castaman, G., Federici AB; Italian
review of prospective studies. Haemophilia 18: e173–e187. Association of Hemophilia Centers (2009). Evidence-based recom-
Favaloro, E.J., Franchini, M., and Lippi, G. (2012). Biological therapies mendations on the treatment of von Willebrand disease in Italy. Blood
for von Willebrand disease. Expert Opin. Biol. Ther. 12 (5): 551–564. Transfus. 7 (2): 117–126.
Federici, A.B. (2008). The use of desmopressin in von Willebrand dis- Mannucci, P.M., Ruggeri, Z.M., Pareti, F.I., and Capitanio, A. (1977).
ease: the experience of the first 30 years (1977–2007). Haemophilia 14 1-deamino-8-D-arginine vasopressin: a new pharmacological
(Suppl 1): 5–14. approach to the management of haemophilia and von Willebrand
Federici, A.B., Baudo, F., Caracciolo, C. et al. (2002). Clinical efficacy of disease. Lancet 1: 869–872.
highly purified, doubly virus-inactivated factor VIII/von Willebrand Mannucci, P.M., Tenconi, P.M., Castaman, G., and Rodeghiero, F.
factor concentrate (Fanhdi) in the treatment of von Willebrand dis- (1992). Comparison of four virus-inactivated plasma concentrates for
ease: a retrospective clinical study. Haemophilia 8: 761–767. treatment of severe von Willebrand disease: a cross-over randomized
Federici, A.B., Mazurier, C., Berntorp, E. et al. (2004). Biologic response trial. Blood 79: 3130–3137.
to desmopressin in patients with severe type 1 and type 2 von Wille- Mazurier, C., De Romeuf, C., Parquet-Gernez, A., and Goudemand,
brand disease: results of a multicenter European study. Blood 103: M. (1989). In vitro and in vivo characterization of a high-purity,
2032–2038. solvent/detergent-treated factor VIII concentrate: evidence for its
Franchini, M. and Mannucci, P.M. (2014). Gastrointestinal angiodyspla- therapeutic efficacy in von Willebrand’s disease. Eur. J. Haematol. 43:
sia and bleeding in von Willebrand disease. Thromb. Haemost. 112: 7–14.
427–431. Mazurier, C., Gaucher, C., Jorieux, S., and Goudeman, M. (1994).
Franchini, M. and Mannucci, P.M. (2016). Von Willebrand factor (Von- Biological effect of demopressin in eight patients with type 2N
vendi®): the first recombinant product licensed for the treatment of (“Normandy”) von Willebrand disease. Br. J. Haematol. 88: 849–
von Willebrand disease. Expert Rev. Hematol. 9: 825–830. 854.
Goudemand, J., Scharrer, I., Berntorp, E. et al. (2005). Pharmacoki- Pasi, K.J., Williams, M.D., Enayat, M.S., and Hill, F.G.H. (1990). Clin-
netic studies on Wilfactin, a von Willebrand factor concentrate with a ical and laboratory evaluation of the treatment of von Willebrand’s
low factor VIII content treated with three virus-inactivation/removal disease patients with heat-treated factor VIII concentrate (BPL8Y).
methods. J. Thromb. Haemost. 3: 2219–2227. Br. J. Haematol. 75: 228–233.
Holmberg, L., Nilsson, I.M., Borge, L. et al. (1983). Platelet aggregation Perkins, H.A. (1967). Correction of the hemostatic defects in von Wille-
induced by 1-desamino-8-D-arginine vasopressin (DDAVP) in type brand disease. Blood 30: 375–380.
IIB von Willebrand’s disease. N. Engl. J. Med. 309: 816–821. Rivard, G.E., Aledort, L., and Alphanate Surgical Investigators (2008).
James, P.D., Lillicrap, D., and Mannucci, P.M. (2013). Alloantibodies in Efficacy of factor VIII/von Willebrand factor concentrate Alphanate
von Willebrand disease. Blood 122: 636–640. in preventing excessive bleeding during surgery in subjects with von
Kaufmann, J.E., Oksche, A., Wollheim, C.B. et al. (2000). Vasopressin- Willebrand disease. Haemophilia 14: 271–275.
induced von Willebrand factor secretion from endothelial cells Shahani, T., Lavend’homme, R., Luttun, A. et al. (2010). Activation
involves V2 receptors and cAMP. J. Clin. Invest. 106: 107–116. of human endothelial cells from specific vascular beds induces the
Kohler, M., Hellstern, P., and Wenzel, E. (1985). The use of heat-treated release of a FVIII storage pool. Blood 115 (23): 4902–4909.
Factor VIII-concentrates in von Willebrand’s disease. Blut 50: 25–27. Smith, T.J., Gill, J.C., Ambroso, D.R., and Hathaway, W.E. (1989).
Mannucci, P.M. (2002). Venous thromboembolism in von Willebrand Hyponatremia and seizures in young children given DDAVP. Am. J.
disease. Thromb. Haemost. 88: 378–379. Hematol. 31: 199–202.
Chapter 19 Platelet disorders
Kenneth J. Clemetson
Department of Haematology, Inselspital, University of Berne, Berne, Switzerland

Introduction, 251 Platelet secretion defects (storage pool disease), 258


Normal platelet function, 251 Procoagulant regulation defects, 259
Diagnosis of platelet defects in bleeding disorders, 252 Giant platelet syndromes and cytoskeletal defects, 260
Platelet adhesion disorders, 253 Transcription factor defects, 260
Platelet aggregation defects: Glanzmann thrombasthenia, 256 Platelet protein polymorphisms and tendency to thrombosis, 260
Agonist receptor defects, 257 Further reading, 261
Defects in intracellular signaling pathways, 257

human disease. Although these are mostly bleeding disor-


Introduction ders, genetic differences leading to an enhanced tendency to
thrombosis are also increasingly suspected.
Over the past few decades a large number of disorders
caused by genetic mutations in platelet components have
been diagnosed at a molecular level. These include defects in
platelet receptors, signaling pathways, and cytoskeletal pro- Normal platelet function
teins. Generally, these were originally detected in bleeding
disorders, varying from severe through moderate to mild. Following damage to the endothelium that protects the vas-
Platelets have a major role in hemostasis, particularly pri- cular system, which may simply be caused by the loss of “old”
mary hemostasis, where platelets adhere to sites of vascular endothelial cells due to the high shear stress in the arterial
damage, become activated, and bind further platelets by system, platelets adhere to the subendothelium via glyco-
aggregation to form a thrombus. The platelet plug formed protein (GP)Ib on the platelet and von Willebrand factor
is then stabilized by secondary hemostasis, which involves (VWF) attached to extracellular matrix proteins, particularly
coagulation factors that assemble on the surface of the acti- collagen (Figure 19.1). This interaction alone is sufficient to
vated platelets, becoming activated in turn and leading to for- start platelet activation, but following initial platelet adhe-
mation of thrombin and cleavage of fibrinogen to form fibrin. sion other receptors such as the collagen receptor GPVI are
Many components have active roles in the overall process, recruited. GPVI comes into contact with collagen and signals
from first platelet contact to stable platelet plug. Most large via its Fcγ subunit to enhance platelet activation and recruit
hospitals see over 100 patients annually, each with mild to integrins, which increase the binding of adherent platelets
moderate bleeding disorders that are not caused by coagula- and provide attachment for further aggregating platelets.
tion factor problems and can be ascribed to uncharacterized Activation of αIIbβ3 allows platelet–platelet aggregation via
congenital platelet defects. While major defects in known ligands such as fibrinogen. Activated platelets release adeno-
receptors may lead to severe bleeding and are relatively easy sine diphosphate (ADP), adenosine triphosphate (ATP),
to diagnose, others are often very difficult to diagnose at a serotonin, and Ca2+ from dense granules and a wide range of
molecular level, and each represents a challenging research proteins from α-granules that contribute to platelet activa-
project; most remain effectively undiagnosed. However, new tion and aggregation, including fibrinogen, high-multimer
methods are beginning to become available which could VWF, and thrombospondin, as well as growth factors and
make such diagnoses more straightforward. This chapter chemokines that affect endothelial cells and leukocytes
deals with genetic defects in platelet components that cause involved in wound repair. The major collagen adhesive
receptor α2β1 is also activated, increasing platelet adhesion
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. to the exposed collagen of the subendothelium. Both these
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. integrins use disulfide bond-reshuffling mechanisms via

251
252 Molecular Hematology

Tethering Adhesion Activation Aggregation

= VWF = GPIb-V-IX = Activated α2β1


= Collagen = GPVI = Activated αIIbβ3
= Fibrinogen
= αIIbβ3
Fig. 19.1 Platelet thrombus formation. Following vessel wall injury, the matrix of the subendothelium is exposed. Under high-shear conditions,
platelets adhere and become tethered via GPIb–V–IX on platelets binding to von Willebrand factor (VWF) in the matrix. This interaction starts
platelet activation, but also brings platelets into contact with the subendothelium, allowing other receptors, such as GPVI, to interact with collagen
and enhance activation. Under static or low-shear conditions, the GPVI–collagen interaction is mainly responsible for the initial activation. Platelet
activation brings additional receptor–ligand interactions into play, such as α2β1–collagen, since this receptor does not bind in the resting state.
Platelet activation also leads to shape change, release of granule contents, activation of other receptors, such as αIIbβ3, and exposure of
negatively charged phospholipids, critical for procoagulant activity. Binding of VWF and fibrinogen to GPIb–V–IX and αIIbβ3 on adjacent platelets
leads to platelet aggregation and the formation of a thrombus.

thiol isomerase to change disulfide bond patterns in the β screened for coagulation defects; if none is detected, exami-
subunits to control the resting or active state. nation for platelet problems is the next logical step. Normally,
Activated platelets also show surface changes, with a complete blood count is done to exclude thrombocytopenia
exposure of negatively charged phospholipids that act, and a peripheral blood smear is examined to check platelet
together with surface receptors such as GPIb, to assemble morphology.
and activate coagulation factors, leading to conversion of A number of whole-blood techniques are now avail-
prothrombin to thrombin. The major effect of thrombin is able to check platelet function, including PFA-100 and
to convert fibrinogen to fibrin, stabilizing and solidifying the Impact technologies. These should indicate the presence of a
thrombus. However, thrombin also feeds back to platelets platelet defect and its likely origin. Platelet aggregation tests
via receptors including GPIb and the seven-transmembrane with a battery of agonists, including ADP, collagen, throm-
PAR1 and PAR4, activating them further. When platelets bin receptor activation peptide (TRAP), arachidonic acid,
are strongly activated by a combination of agonists such as epinephrine, and ristocetin, should also help to narrow down
thrombin/collagen, that part of the activated platelet popu- which responses are defective. Flow cytometry or Western
lation with strong Ca2+ fluxes shows enhanced procoagulant blotting with specific monoclonal antibodies may be used
properties though ballooning via water intake to present an to check for the absence of receptors or to determine the
increased surface and by covalent binding of coagulation amounts present. Flow cytometry may also be used to exam-
factors. When platelets are treated with Ca2+ -ionophores, ine the expression of P-selectin, the activation of αIIbβ3, or
all platelets fall in this hyperactive category. It is not yet the exposure of negatively charged phospholipids in response
clear why with thrombin/collagen activation only part of the to the classic agonists. All these techniques can provide valu-
platelet population is hyperactivated. able information to lead to a molecular diagnosis.
If a defect can be ascribed to a given platelet molecule,
coding regions can be amplified from genomic DNA and
Diagnosis of platelet defects in sequenced. In rare cases it may also be necessary to sequence
bleeding disorders non-coding regions to establish a genetic reason for a par-
ticular platelet protein deficiency. Regardless of whether a
Most patients with clinically relevant platelet defects seek disorder is homozygous or compound heterozygous in reces-
medical assistance because of bleeding problems, ranging sive disease, or present in only one allele in dominant disease,
from easy or excessive bruising, epistaxis, gingival bleed- it is extremely useful to establish the familial inheritance
ing, and menorrhagia to excessive bleeding following tooth by analyzing DNA from other family members. Modern
extraction or other surgery. Such patients are normally “next-generation” sequencing methods are increasingly
Platelet disorders 253

being used routinely for molecular diagnosis of bleeding BSS is characterized by thrombocytopenia, giant platelets
disorders of platelet origin. These are much more effective (up to 20 μm diameter), decreased platelet adhesion, reduced
when applied to patients with a clear phenotype where platelet survival, and abnormal prothrombin consumption.
likely genes can be targeted. However, problems arise where BSS platelets do not aggregate in response to ristocetin or
the phenotype is simply mild bleeding and it is difficult botrocetin and show weaker and slower responses to throm-
to ascribe the problem to poorly defined mutations/single bin due to the lack/deficiency of GPIb as a thrombin receptor
nucleotide polymorphisms (SNPs) in genes for unclearly that accelerates responses. As well as interacting with VWF
related proteins. In those cases where this has been effective, and thrombin, GPIb is a receptor for a wide range of lig-
there have generally been sufficient family members with ands with various physiological roles, including P-selectin,
and without the phenotype to show a clear relationship. In thrombospondin 1, factors XI and XII, αMβ2, and high-
some cases it has been possible to establish mouse models of molecular-weight kininogen; its absence in BSS may there-
these disorders, confirming and extending the diagnosis. fore contribute to other aspects of this disorder.
Four separate genes, GPIBA (chromosome 17), GPIBB
(chromosome 22), and GP5 and GP9 (chromosome 3), code
Platelet adhesion disorders for the subunits of the GPIb complex, which are expressed
relatively late in megakaryocyte maturation. All four subunits
belong to the leucine-rich repeat (LRR) family of proteins,
Bernard–soulier syndrome
with 8, 2, 16, and 2 repeats present, respectively. GPIbα and
Bernard–Soulier syndrome (BSS) is a rare bleeding disorder GPIbβ are linked covalently via disulfide bonds, probably in
caused by defective expression or function of the GPIb–V– a 1 : 2 ratio, while GPIX and GPV associate non-covalently
IX receptor complex (Figure 19.2). With a few still unclear with GPIb, probably in 1 : 1 and 2 : 1 ratios, respectively (Fig-
exceptions, it is inherited in an autosomal recessive way, and ure 19.3). While GPIbβ and GPIX are certainly critical for the
homozygous cases are often associated with consanguinity. function of the complex, with structural and signaling roles,

GPlb complex
Bernard-Soulier syndrome
Platelet-type von Willibrand disease

Collagen-receptor defects
αllbβ3
Glanzmann thrombasthenia
α2β1
GPVl

7 Transmembrane
receptor defects
e.g. TXA2 receptor

CLEC2 α5β1
α6β1
Fig. 19.2 Diagram of a resting platelet indicating the major receptors implicated in inherited bleeding disorders.
254 Molecular Hematology

VWF-binding
site Thrombin-binding
sites
GPIbα

GPIb–V–IX GPV
complex
Highly O-glycosylated
2GPIbβ
domain
Fig. 19.3 Structure of the GPIb–V–IX complex.
GPIbα is linked via disulfide bridges to two GPIbβ
GPIX
molecules and this complex is non-covalently
associated with GPIX and GPV in the ratio of
Calmodulin 2 : 4 : 2 : 1. Mutations and deletions leading to
Bernard–Soulier syndrome and platelet-type von
Willebrand disease are described in the text.
14-3-3ζ Mutations affecting cytoskeletal molecules such as
Actin Filamin
filaments filamin-1 and myosin heavy chain IIA, which
interact directly or indirectly with GPIb complex,
Myosin heavy chain IIA can also lead to giant platelet syndromes.

the function of GPV remains controversial; it has been sug- functional GPIb are often still expressed, enough to support
gested that it has a role in the thrombin response (it contains basic levels of hemostasis if other aspects, such as coagula-
a thrombin-binding and -cleavage site) as well as in platelet tion factors, are normal. The Asn45Ser mutation in GPIX
collagen binding. is the commonest cause of BSS among northern Europeans,
Mutations and other genetic defects causing BSS have now accounting for the majority of cases among these popula-
been collected on a website (www.bernardsoulier.org) and tions. Because this mutation generally does not cause such a
will therefore not be listed in detail here. Many of the muta- marked phenotype as many of the GPIbα and GPIbβ muta-
tions found to cause BSS lie in the LRR domains and appear tions, patients have not been easily detected and diagnosis is
to destabilize their folding. The subunits also contain typi- only now catching up. This mutation has also been found in
cal disulfide bridge patterns, and thus mutation of the con- patients of Turkish origin. Other mutations in GPIX, leading
stituent cysteines or of other amino acids close to cysteine to its failure to express, produce similar phenotypes.
has deleterious effects on folding, leading to lack of expres- Mice models of BSS have been produced through targeted
sion of the affected subunit and hence decrease in expression knockout of GPIbα or GPIbβ, but so far not GPIX (most
of the rest of the complex. likely because the rest of the complex is partially expressed).
No cases of BSS due to defects in GPV have been described The phenotype is essentially the same as in humans. Research
and mice lacking GPV express GPIb–IX normally. On the on megakaryocytes from these mice indicates that proplatelet
other hand, expression of GPIb–IX appears to be essential formation and microtubule coil assembly are defective. As
for stable expression of GPV on platelets, possibly by protect- already noted, GPV knockout mice do not show major differ-
ing it from proteolysis. Most mutations in GPIbα either cause ences in phenotype using the present selection criteria and, in
folding problems or are nonsense mutations leading to pre- particular, do not have characteristics of BSS.
mature termination. Rare BSS variants have been described
as having dominant inheritance and lead to expression of
Platelet-type von Willebrand disease
non-functional GPIb, although only one allele is affected.
While the molecular mechanism involved is not understood, Platelet-type von Willebrand disease (VWD) is an inherited
it has been suggested that the defective GPIbα molecule dominant bleeding disorder resembling VWD type IIB, but
prevents the normal one from functioning. This class of caused by mutations in GPIbα rather than in the A1 domain
mutation includes Leu57Phe, Leu129Pro, Ala156Val (known of VWF (see Figures 19.2 and 19.3). Unlike BSS, platelet-type
as BSS Bolzano), and Leu179del (known as BSS Nancy). VWD shows a gain of function, with spontaneous binding
Recently, a monoallelic Val31Leu mutation was described in of VWF to platelets. In platelet-type VWD, platelets are
an Indian patient with BSS symptoms. Mutations in GPIbβ often enlarged like those in BSS, although the reasons for
generally have the same effect as those in GPIbα concern- this are not yet clear. It may be due to enhanced binding
ing expression. Mutations in GPIX, while causing many of of VWF to GPIb, which blocks other GPIb functions. The
the classic symptoms of BSS, generally do not produce such platelet-type VWD mutations that have been described
a severe phenotype because trace amounts (up to 10%) of include Gly233Val/Ser and Met239Val, and a 27 bp deletion
Platelet disorders 255

in the region of the gene coding for the macroglycopeptide (Figure 19.4). Most of the defects have not been diagnosed
domain of GPIbα has been reported to produce a similar on a molecular level. There are two recent reports of bleed-
effect. Recently, two related patients were diagnosed with ing disorders caused by compound heterozygous mutations
platelet-type VWD due to a heterozygous 27 bp inframe in GPVI. One patient with a lifelong history of bleeding
deletion in GP1BA that removes amino acids 459–467 and problems had structurally normal platelets, but a functional
therefore overlaps the deletion already mentioned. Studies platelet defect. Platelet aggregation was normal except for
on mutant proteins suggest that mutations in other amino an absent response to collagen, convulxin, and the collagen-
acids of the flexible β-loop structure of GPIbα can also related peptide. ATP-dense granule secretion was normal
produce this phenotype. with ADP, but defective with collagen. Thrombus forma-
tion on a collagen surface in flowing blood was reduced,
Collagen receptor defects but more single platelets were attached. PFA-100 analysis
showed a shortened collagen/ADP closure time. Flow cytom-
α2β1 integrin etry showed absence of GPVI expression, while immunoblot-
A few patients have been reported with bleeding problems ting showed strongly reduced levels of GPVI. The patient
related to an α2β1 integrin deficiency, showing defective is compound heterozygous for an out-of-frame 16 bp dele-
platelet adhesion to collagen while responses to other ago- tion and a missense mutation S175 N in a highly conserved
nists were normal. Both patients were female and became residue of the second immunoglobulin-like GPVI domain.
normal following the menopause, suggesting a hormonal role The parents, who do not have clinical bleeding problems, are
in the disorder. Neither α2−/− nor β1−/− mice have major heterozygous carriers. The mother carries the S175 N muta-
hemostatic problems, but defects were reported in aggre- tion and has a mild platelet functional defect. In vitro studies
gation to fibrillar collagen and adhesion to soluble colla- showed reduced membrane expression and convulxin bind-
gen. Differences in occlusion times in thrombosis models in ing in the S175 N mutant compared with the wild-type GPVI
α2−/− or β1−/− mice seem to be dependent on the model receptor.
used and remain controversial. The other patient, a 10-year-old girl, had a tendency to
bruising since infancy, a prolonged bleeding time despite
a normal platelet count, and no antiplatelet antibodies.
GPVI
Collagen-induced platelet activation was null, although
Several patients have been described with mild bleed- there was an incomplete deficiency of GPVI detected by
ing disorders related to low levels or deficiency of GPVI flow cytometry. Immunoblotting showed abnormal residual

R38C
Collagen

GPVI C2-1
C2-2

N-glycosylation

S175N
Polymorphism sites
O-glycosylation

Metalloprotease Salt bridge


cleavage site
R
DD

ITAM-domains

Fig. 19.4 Structure of the GPVI complex. GPVI is linked via a salt bridge to FcγR, which provides most of the signaling to the platelet
cytoplasm. The positions of mutations leading to decreased expression and of polymorphisms thought to affect function are indicated. Activation
of GPVI by autoantibodies can lead to coactivation of matrix metalloproteases and loss of GPVI by cleavage.
256 Molecular Hematology

GPVI, and no FcγR defect. DNA sequencing revealed an Ligand-


αIIb β3
R38C mutation in exon 3 of one allele of GPVI and an inser- binding site
tion of five nucleotides in exon 4 of the other allele, leading
β-Propeller βA
to a premature nonsense codon and absence of the corre-
Ca2+-binding Disulfide-rich
sponding mRNA. Expression of the R38C mutation gave sites domains
an abnormal protein migration and loss of collagen bind-
ing. This composite genetic GPVI defect leads to absence of Hybrid
Thigh PSI
platelet responses to collagen and a mild bleeding phenotype.
Platelets from GPVI−/− mice do not adhere to colla- EGF-1
gen under static conditions, probably because the α2β1 EGF-2
integrin is not activated. GPVI is probably important for Calf-1
platelet activation on subendothelium under low-shear con- EGF-3
ditions. Under high-shear conditions in GPVI−/− mice,
α2β1 is probably activated via GPIb/VWF interactions, EGF-4
with VWF bound to collagen. Stable adhesion and spread- Calf-2
ing are strongly affected in GPVI−/− platelets, indicating βTD
that GPVI has an important role in downstream signaling
to molecules critical for thrombus formation. Tail bleeding
Membrane
times in GPVI−/− mice are only slightly prolonged compared
with wild type. Thrombosis models using both GPVI−/− and
Talin
Fcγ−/− (the GPVI signaling subunit) mice are controversial, Kindlin-3
because the type of vascular wall damage involved is vari-
able. Thus, laser-induced injuries did not implicate GPVI as a Fig. 19.5 Structure of the αIIbβ3 complex. This major platelet
major factor in thrombosis, whereas FeCl3 -induced injuries integrin is shown in the activated state. The various structural domains
are labeled. Cytoskeletal proteins associated with the cytoplasmic
suggested a more substantial role. Other research pointed
domain of β3 in the activated state are shown.
to thrombin production as a major factor in overcoming
GPVI/Fcγ deficiencies in mice thrombosis models. There is
a clear need for research involving mouse thrombosis mod-
els that are more closely related to human pathology, such as are traditionally designated as type I, those with a moderate
in the presence of fragile vascular plaque, before any defini- deficiency (10–50% of normal) as type II, and others as vari-
tive conclusions can be reached about collagen receptors in ants. The latter includes patients who express non-functional
human thrombosis. Increased amounts of soluble GPVI in αIIbβ3 on their platelets. There is a very wide range of molec-
plasma seem to be indicative of raised platelet activation in ular defects in αIIb or β3 and as a consequence the clini-
vivo, for whatever reason. This may include vascular prob- cal symptoms are highly variable. However, even the same
lems, inflammation, cirrhosis, or latent cancer. defect may show some variability in symptoms in different
patients, depending on other factors such as levels of coagu-
lation factors. Typical symptoms include purpura, epistaxis,
Platelet aggregation defects: gum bleeding, and menorrhagia, but gastrointestinal and
Glanzmann thrombasthenia interjoint bleeding and hematuria are rare. Problems gener-
ally follow trauma and are rarely spontaneous. Diagnostic cri-
Glanzmann thrombasthenia (GT) is an autosomal recessive teria are prolonged bleeding time and defective clot retrac-
bleeding disorder caused by defects in the αIIbβ3 integrin tion. Platelet aggregation to all agonists except ristocetin is
or its signaling (see Figures 19.2 and 19.5). If this receptor absent or defective. Molecular diagnosis of GT includes flow
is absent or defective, it is unable to interact with its ligands, cytometry and/or Western blotting to establish whether both
including fibrinogen and VWF, but also fibronectin and vit- subunits are absent or present in reduced amounts.
ronectin. Thus, platelet aggregates are not formed or are lim- Mutations and other genetic defects causing GT have
ited in extent, preventing efficient recruitment of platelets to a now been collected on a database (http://sinaicentral.mssm.
damaged vessel site. In addition, αIIbβ3– fibrinogen binding edu/intranet/research/glanzmann) and are therefore not
has an important role in clot retraction, the process by which listed in detail here. GT can be caused by mutations in either
wound edges are drawn together to help seal the wound. subunit αIIb or β3 and may show some distinct differences.
Hence, GT patients form more scar tissue than normal Since fibrinogen is not synthesized in the megakaryocyte
because wounds tend to reopen. Patients with platelets lack- but in the liver and transported to the α-granules from
ing, or with a severe deficiency (<5% of normal) in αIIbβ3, the plasma by αIIbβ3-dependent endocytosis, GT patients
Platelet disorders 257

often lack or are deficient in platelet α-granule fibrinogen, release Ca2+ from the dense tubular system and cytoskele-
a contributory factor to their poor hemostatic function. tal changes lead to shape change. P2Y12 signals to αIIbβ3,
In patients lacking β3, who as a consequence lack the leading to platelet aggregation. Thus, platelet defects in P2Y12
vitronectin receptor αvβ3 as well as αIIbβ3, the platelet were first thought to be GT variants. Platelets from rare
α-granules, in addition to fibrinogen deficiency, also contain patients in both France and Italy showed decreased and
up to five times the normal amounts of vitronectin. Vit- reversible platelet aggregation to ADP, but normal shape
ronectin is synthesized in megakaryocytes, so these results change and calcium mobilization. This was due to an auto-
suggest a possible role for αvβ3 in transporting vitronectin somal recessive hereditary disease affecting one allele of
from α-granules out of platelets. Both αIIb and β3 have the P2Y12 gene. No patients with P2Y1 defects have been
a complex gene structure, with the αIIb gene (ITGA2B) described so far, but knockout mice lacking this receptor have
composed of 30 exons and spanning 17 kb, while the β3 been prepared. P2X1 is the platelet receptor for ATP. Three
gene (ITGB3) consists of 15 exons and spans 46 kb. Both molecules form a calcium channel that is regulated when
are located on chromosome 17q21–23. Genetic defects are platelets are activated, to control intracellular calcium levels.
distributed over the entire region. One case of a young girl with a bleeding syndrome caused by
Mutations/deletions may prevent subunit biosynthesis deletion of a single amino acid in P2X1 has been described.
or transport of precursors from the endoplasmic reticulum That this is a dominant inherited disease is presumably due to
to the Golgi or plasma membrane. When mutations do not the defective molecule preventing the active calcium channel
affect folding too seriously, expression of lower levels of the from forming.
complex may occur with some functions retained. Several
GT mutations lead to expressed dysfunctional receptor.
Other primary agonist receptor defects
Thus, an Asp119Tyr mutation in β3 affects the key RGD
binding site, while Ser752Pro removes a key phosphory- Rare examples of an Arg60Leu mutation in the thromboxane
lation site involved in outside–in signaling. A stop codon (TX)A2 receptor have been reported in some Japanese fami-
truncating β3 (Arg724tTer), so that only 8 of the normal 47 lies that result in defective signaling. Mutations in other ago-
amino acids of the cytoplasmic domain are present, removes nist receptors have been reported, but it is not clear that these
binding sites for the cytoskeletal proteins talin and kindlin-2. affect platelet function. Common variants in the PAR4 gene
Cys560Arg and Cys598Tyr mutations in β3 produce platelets (F2RL3) have been described, but only recently were effects
that spontaneously bind fibrinogen, because the β3 in their of these on platelet activation observed, particularly in the
αIIbβ3 has its disulfide bridges blocked in the activated presence of clinically used PAR1 inhibitors. So far there have
pattern, and because the disulfide isomerase activity that been no reports of bleeding disorders linked to defects in the
normally switches between resting and activated and vice PAR1 thrombin receptors.
versa is unable to do so. A heterozygous cytoplasmic domain
mutation in the αIIb subunit Arg995Gln within the critical
GFFKR sequence reduces expression of the complex and Defects in intracellular signaling
produces mild “thrombasthenia-like” symptoms. There is pathways
some evidence for cases of GT caused by replacement of
DNA segments lying outside coding regions and affect- A wide range of defects in signaling molecules are known,
ing mRNA stability. A more general syndrome affecting often affecting other cells as well as platelets (Figure 19.6).
inside–out activation of several classes of integrins present These include defects in phospholipase C (PLC) activation,
in platelets, neutrophils, and lymphocytes has been shown to calcium mobilization, and plekstrin phosphorylation. Stud-
be due to mutations in the CalDAG-GEFI gene, preventing ies in one of these patients with impaired PLC activation
signaling to the small GTPase Rap1. indicated a decrease only in PLC-β2, while other isoforms
Recent studies on the linkage between ITGA2B and ITGB3 were normal. Coding sequence was normal, but mRNA for
gene variants in patients with GT show disequilibrium, indi- PLCβ2 was decreased, suggesting sequence changes in the
cating that most disease-causing mutations are recent. gene outside the coding region affecting mRNA stability.
Eight patients were described with bleeding problems and
abnormal aggregation and secretion in response to several
Agonist receptor defects different agonists. Receptor-mediated calcium mobiliza-
tion and/or plekstrin phosphorylation were abnormal in
seven of these patients. Several patients have been reported
ADP and ATP receptor defects
with platelet cyclooxygenase deficiency, a milder bleeding
Platelets interact with ADP via the purinergic seven- disorder, and impaired platelet aggregation responses.
transmembrane receptors P2Y1 and P2Y12 . P2Y1 signals to Thromboxane synthase deficiency has been described in two
258 Molecular Hematology

Giant platelet syndromes


Myosin heavy chain llA
Filamin 1

α-granule syndromes
Metabolic defects Gray platelet syndrome
ATP synthesis α, δ-deficiency
Quebec syndrome

Enzyme defects
Cyclooxygenase
Thromboxane synthase
Signaling defects Dense granule syndromes
Lipoxygenase
Kinases/phosphatases Chédiak–Higashi syndrome
G-proteins, Ca2+ fluxes Hermansky–Pudlak syndrome
Phospholipases/PKC Griscelli syndrome
Fig. 19.6 Diagram of a resting platelet indicating the major internal organelles and other structures implicated in inherited bleeding disorders.

patients. In other patients a deficiency of the PLC-γ2 isoform and CD63, synthesized in megakaryocytes that translocate
or a specific decrease in platelet Gαq has been reported. to the plasma membrane during platelet activation and
Defects in signaling molecules probably account for many secretion and are good markers for these processes. Inher-
of the undiagnosed patients with mild to moderate bleeding ited deficiencies of plasma proteins that are taken up into
problems. Techniques to diagnose these on a molecular basis α-granules will be reflected in their content.
are only starting to be developed, and even proteomics by
itself cannot be expected to detect all of these. There is still
a large unmet need here. Gray platelet syndrome
Gray platelet syndrome is characterized by the absence
of platelet contents and has a mostly autosomal recessive
Platelet secretion defects (storage inheritance. Packaging or storage of proteins during platelet
pool disease) production in megakaryocytes is defective and the pro-
teins are directly released from the megakaryocytes. The
This is a large and heterogeneous group of inherited disor- platelets are often larger than normal and the patients are
ders caused by intracellular defects of platelets (Figure 19.6). somewhat thrombocytopenic. Platelet aggregation responses
Since secretion is a function of many cell types, it is clear that are variously affected and differ between patients. Tissue
the phenotype may extend beyond hemostasis, depending on inhibitors of metalloproteinases (TIMPs) are still present
which molecule is affected. in gray platelet syndrome and may account for some of this
variability. In 2011, three groups applied next-generation
sequencing to study the genetic defect underlying gray
Defects of α-granules
platelet syndrome and showed mutations in NBEAL2 in
Proteins in α-granules are either synthesized in megakary- all patients. NBEAL2 is a member of the LYST (mutated in
ocytes or endocytosed from plasma. Membranes of Chesiak–Higashi syndrome) gene family. Following these
α-granules also contain specific receptors, such as P-selectin results, the production of NBEAL2−/− mice has extended
Platelet disorders 259

phenotype studies in gray platelet syndrome. After injury, Griscelli syndrome


tissue regeneration was affected. Megakaryocyte maturation
Patients with Griscelli syndrome are partially albino with
was delayed and few α-granules observed. Unusual direct
silver hair. Neurological defects and/or immunodeficiency
secretion of VWF was also seen.
are linked to lymphocyte cytotoxicity defects. Clinical man-
ifestations include fatal complications caused by lymphoid
Quebec platelet disorder cell activation and cytokine release. Griscelli syndrome
is associated with mutations in the genes for myosin Va,
As the name implies, this autosomal dominant bleeding
Rab27a (a small GTPase), or melanophilin. A mouse
disorder was described in this region of Canada. Many
model for this disease has recently been characterized with a
α-granule proteins are degraded proteolytically, including
mutation in Rab27a, a variant of the ashen mouse with a phe-
membrane receptors such as P-selectin. Defective platelet
notype resembling Hermansky–Pudlak syndrome. Rab27a
aggregation is particularly strong, with epinephrine as
deficiency in humans was identified in a young patient with
agonist. The problem is caused by overexpression of uroki-
psychomotor retardation. The patient’s platelets lacked dense
nase plasminogen activator, a protease normally stored in
granules, but had normal α-granules; epinephrine-induced
α-granules and released on platelet activation.
aggregation was defective.

Dense (δ) granule defects


Wiskott–aldrich syndrome
Platelet-dense granules are storage sites for a number
of small molecules, including ADP, ATP, serotonin, and Platelets in Wiskott–Aldrich syndrome (WAS) aggregate
calcium. Defects in these granules therefore affect platelet poorly and have few granules. This is an X-linked recessive
aggregation responses (Figure 19.6). Granule deficiencies disease characterized by thrombocytopenia, small platelets,
are variable and disorders are known in which α-granules eczema, immunodeficiency, and increased autoimmunity
are also affected, in which common pathway molecules are and malignancy problems. Hereditary X-linked thrombocy-
defective (αδ-storage pool deficiency). Since similar types of topenia is a milder form lacking the immune problems. T
organelles exist in other cells, these are often disorders that lymphocytes are the other major blood cells affected. The
affect pigmentation of the skin and hair, such as Hermansky– 502-amino-acid WAS protein is encoded by the large WAS
Pudlak, Chédiak–Higashi, and Griscelli syndromes. Mouse gene, which consists of 12 exons. Mutations in exons 1 and
models exist for several of these. 2 mostly lead to the mild form. WAS protein regulates actin
polymerization in hematopoietic cells and its deficiency leads
to premature proplatelet formation in the bone marrow. In
Hermansky–pudlak syndrome mice, profilin-1 deficiency in megakaryocytes was shown to
Hermansky–Pudlak syndrome manifests as albinism and lead to microtubule instability and a large increase in acety-
absence of ceroid-lipofuscin storage in the reticuloendothe- lated tubulin, as well as affecting platelet production and
lial system. The syndrome is common in Puerto Rico and is causing a WAS-like platelet defect.
caused by a frameshift due to a 16 bp duplication in exon 15
of the HPS-1 gene, which normally encodes a 79 kDa protein
with two membrane-spanning domains. Defects in at least Procoagulant regulation defects
eight genes cause distinct subtypes of Hermansky–Pudlak
syndrome in humans.
Scott syndrome
Scott syndrome is a very rare disorder found in a few humans
Chédiak–higashi syndrome
and some dogs. So far there is no mouse model. When nor-
Severe immunological defects and progressive neurological mal platelets are activated, one of the changes produced is
dysfunction accompany bleeding problems. The major diag- the exposure of negatively charged phospholipids on the
nostic criterion is the presence of giant inclusion bodies in surface, critical for procoagulant activity. This is absent in
a variety of cells with granules, including platelets. The gene Scott syndrome platelets and as a consequence less thrombin
responsible (LYST) has been cloned and a series of frameshift and fibrin are formed in the platelet plug, leading to bleeding
and nonsense mutations identified that produce a truncated complications. This defect also affects membrane vesicu-
LYST protein. A milder form of the disease is thought to be lation, suggesting a common mechanism. Under in vitro
due to rare missense mutations. LYST protein is large and conditions, this phenomenon requires platelet activation by
complex, with domains suggesting that it regulates organelle thrombin and collagen (or a GPVI agonist such as convulxin
protein trafficking and membrane–membrane interactions. or collagen-related peptide). The molecular defect in Scott
260 Molecular Hematology

syndrome was recently attributed to mutations in the gene in dogs. It is important to distinguish these large platelet syn-
for anoctamin 6 (TMEM16F), a Ca2+-activated Cl− channel. dromes (including BSS), particularly when accompanied by
thrombocytopenia, from idiopathic thrombocytopenic pur-
pura, and to avoid treatments such as splenectomy, which are
Stormorken syndrome
not useful in these disorders.
Stormorken syndrome is an even rarer disorder confined so
far to one family, implying a dominant inheritance. Unlike
Scott syndrome, the platelets are constitutively activated,
Transcription factor defects
with surface exposure of negatively charged phospholipids
and raised levels of microvesicles even under resting con-
Autosomal dominant Paris–Trousseau syndrome is a typi-
ditions. The molecular basis of Stormorken syndrome was
cal example of this class of disorder, with decreased platelet
recently shown to be a dominant mutation in STIM1. Gain-
production and a mild bleeding tendency associated with a
of-function mutations in STIM1 are responsible for a CRAC
deletion at 11q23. Platelets are often larger than normal with
channel defect in York platelet syndrome characterized by
giant α-granules. The defect leads to a hemizygous loss of the
thrombocytopenia, ultrastructural granule abnormalities
FLI1 gene coding for Fli1 transcription factor. A subpopula-
including giant dense granules and massive, multilayered
tion of megakaryocyte precursors fails to mature adequately
target bodies, and lack of Ca2+ -storage in δ-granules.
to produce platelets. Other disorders including thrombo-
cytopenia are caused by mutations in the GATA-1 gene.
Platelets may be larger than normal, with poor response to
Giant platelet syndromes and
collagen. The phenotype depends on whether the mutations
cytoskeletal defects
affect GATA-1 interactions with DNA or FOG-1. GATA-1-
dependent platelet molecules include members of the GPIb
Although this group is diverse, many of the patients have
complex and α-granule components. Defects in RUNX1 lead
mutations or other defects in the MYH9 gene coding for non-
to platelets with a deficiency in protein kinase C-θ and lack
muscle myosin heavy chain IIA (see Figures 19.3 and 19.6).
of phosphorylation of its downstream targets, but also an
Since this is expressed in a variety of cells, not only platelets
increased tendency to develop leukemia.
are affected. Macrothrombocytopenia (variable, up to leuko-
cyte size) is accompanied by variable phenotypes in other
cells/tissues. Döhle bodies in leukocytes with distinctive
distribution of myosin IIA detected by immunofluorescence Platelet protein polymorphisms and
is a major diagnostic criterion. Hearing loss caused by defec- tendency to thrombosis
tive cilia, nephritis, and cataracts are all possible symptoms
in later life. Characteristic mutation zones in the protein Platelet activation responses vary widely in the population,
seem linked to particular symptoms: mutations in the head but are reproducible for individuals and show a strong genetic
region can affect folding of the entire molecule and can component. Following the demonstration of major polymor-
yield more serious symptoms, whereas mutations toward phisms in coagulation factors affecting thrombotic tenden-
the C-terminus have less dramatic effects, possibly because cies, there has been a major effort to identify polymorphisms
function is partly maintained and there is less tendency to in platelet proteins such as receptors that could explain the
form denatured protein clusters. While non-muscle myosin interindividual variation. There is also considerable interest
has an important role in many cells/tissues, the presence of in polymorphisms that affect patient responses to antithrom-
the IIB and IIC forms may compensate in some of these. botic treatments, because this could be an important way to
Giant platelets have also been reported in patients with improve treatment and reduce mortality.
mutations in the FLNA gene coding for filamin A. As Common polymorphisms in αIIbβ3 were among the
well as platelets, neuronal migration was abnormal in these first targets for statistical investigation. Each of the sub-
patients. In platelets, filamin A is a major attachment site units contains a common amino acid dimorphism: β3 has
for the GPIb complex, stabilizing the connection between two isoforms, Leu33/Pro33, that lead to conformational
the membrane and the cytoskeleton, and anchoring GPIb differences in the proteins; the Ile843/Ser843 dimorphism
in platelets when they attach during adhesion under high in αIIb has also been intensively investigated. Most of the
shear. Myosin IIA may play a role in this process, as well studies were controversial, with only minor effects detected.
providing a rationale for the giant platelets in these disor- The situation in GPIbα is more complicated, with two
ders as well as BSS. Recently, a number of patients have been major polymorphic systems present: (i) a Thr145/Met145
described with mutations in the β-tubulin gene, also leading dimorphism and (ii) several versions of a tandem repeat
to macrothrombocytopenia, and similar defects were found structure of 13 amino acids within the macroglycopeptide
Platelet disorders 261

domain; these have the nomenclature VLTR-A, -B, -C, and Prevost, N., Woulfe, D., Tognolini, M., and Brass, L.F. (2003). Contact-
-D with four, three, two, and one copies, respectively, of the dependent signaling during the late events of platelet activation. J.
tandem repeat. The incidence varies widely depending on Thromb. Haemost. 1: 1613–1627.
ethnic origins between Europe and East Asia. Again, there Ren, Q., Ye, S., and Whiteheart, S.W. (2008). The platelet release reac-
has been no consensus reached about a role in thrombosis tion: just when you thought platelet secretion was simple. Curr. Opin.
Hematol. 15: 537–541.
susceptibility and many studies have given contradictory
Rivera, J., Lozano, M.L., Navarro-Nunez, L., and Vicente, V. (2009).
results. Polymorphisms in collagen receptors α2β1 and Platelet receptors and signaling in the dynamics of thrombus forma-
GPVI have also been implicated in thrombosis susceptibility, tion. Haematologica 94: 700–711.
but these studies remain controversial. Ruggeri, Z.M. and Mendolicchio, G.L. (2007). Adhesion mechanisms in
Perhaps if one considers that many individual genetic platelet function. Circ. Res. 100: 1673–1685.
differences may contribute to the overall thrombosis sus-
ceptibility, it is not so surprising that these single differences
do not have more pronounced effects. However, several Laboratory assessment of platelet
newly discovered polymorphisms, particularly in signaling disorders
molecules, are still under investigation. One area which is Harrison, P. (2009). Assessment of platelet function in the laboratory.
less controversial is the effect of cytochrome P450 polymor- Hamostaseologie 29: 25–31.
phisms on activation of clopidogrel, and therefore on its Hayward, C.P. and Favaloro, E.J. (2009). Diagnostic evaluation of
ability to inhibit the ADP receptor P2Y12 . Among persons platelet disorders: the past, the present, and the future. Semin.
treated with clopidogrel, carriers of a reduced-function Thromb. Hemost. 35: 127–130.
CYP2C19 allele had significantly lower levels of the active Simeoni, I., Stephens, J.C., Hu, F. et al. (2016). A high-throughput
metabolite of clopidogrel, diminished platelet inhibition, sequencing test for diagnosing inherited bleeding, thrombotic, and
and a higher rate of major adverse cardiovascular events, platelet disorders. Blood 127: 2791–2803.
including stent thrombosis, than did non-carriers.
Overview of inherited platelet disorders
Johnson, B., Lowe, G.C., Futterer, J. et al. (2016). Whole exome sequenc-
ing identifies genetic variants in inherited thrombocytopenia with
Further reading secondary qualitative function defects. Haematologica 101 (10):
1170–1179.
Overview of platelet function Kunicki, T.J., Williams, S.A., and Nugent, D.J. (2012). Genetic variants
that affect platelet function. Curr. Opin. Hematol. 19: 371–379.
Clemetson, K.J. (2007). A short history of platelet glycoprotein Ib com-
Lanza, F. (2007). Murine models of platelet diseases. Transfus. Clin. Biol.
plex. Thromb. Haemost. 98: 63–68.
14: 35–40.
Clemetson, K.J. (2012). Platelets and primary haemostasis. Thromb. Res.
Nurden, P. and Nurden, A.T. (2008). Congenital disorders associated
129: 220–224.
with platelet dysfunctions. Thromb. Haemost. 99: 253–263.
Clemetson, K.J. and Clemetson, J.M. (2001). Platelet collagen receptors.
Nurden, A.T. and Nurden, P. (2015). Inherited disorders of platelet func-
Thromb. Haemost. 86: 189–197.
tion: selected updates. J. Thromb. Haemost. 13 (Suppl. 1): S2–S9.
Coller, B.S. and Shattil, S.J. (2008). The GPIIb/IIIa (integrin alphaIIb-
Rao, A.K., Jalagadugula, G., and Sun, L. (2004). Inherited defects in
beta3) odyssey: a technology-driven saga of a receptor with twists,
platelet signaling mechanisms. Semin. Thromb. Hemost. 30: 525–535.
turns, and even a bend. Blood 112: 3011–3025.
Salles, I.I., Feys, H.B., Iserbyt, B.F. et al. (2008). Inherited traits affecting
Farndale, R.W., Slatter, D.A., Siljander, P.R., and Jarvis, G.E. (2007).
platelet function. Blood Rev. 22: 155–172.
Platelet receptor recognition and cross-talk in collagen-induced acti-
vation of platelets. J. Thromb. Haemost. 5: 220–229.
Hartwig, J.H. and Italiano, J.E. Jr. (2006). Cytoskeletal mechanisms for Bernard–soulier syndrome overview
platelet production. Blood Cell Mol. Dis. 36: 99–103.
Jackson, S.P. (2007). The growing complexity of platelet aggregation. Dumas, J.J., Kumar, R., McDonagh, T. et al. (2004). Crystal structure of
Blood 109: 5087–5095. the wild-type von Willebrand factor A1–glycoprotein Ibα complex
Lau, T.L., Kim, C., Ginsberg, M.H., and Ulmer, T.S. (2009). The struc- reveals conformation differences with a complex bearing von Wille-
ture of the integrin αIIbβ3 transmembrane complex explains inte- brand disease mutations. J. Biol. Chem. 279: 23327–23334.
grin transmembrane signalling. EMBO J. 28: 1351–1361. Lanza, F. (2006). Bernard–Soulier syndrome (hemorrhagiparous
Ozaki, Y., Asazuma, N., Suzuki-Inoue, K., and Berndt, M.C. (2005). thrombocytic dystrophy). Orphanet J. Rare Dis. 1: 46.
Platelet GPIb-IX-V-dependent signaling. J. Thromb. Haemost. 3: Pham, A. and Wang, J. (2007). Bernard–Soulier syndrome: an inherited
1745–1751. platelet disorder. Arch. Pathol. Lab. Med. 131: 1834–1836.
Plow, E.F., Qin, J., and Byzova, T. (2009). Kindling the flame of integrin Savoia, A., Kunishima, S., De Rocco, D. et al. (2014). Spectrum of
activation and function with kindlins. Curr. Opin. Hematol. 16: 323– the mutations in Bernard-Soulier syndrome. Hum. Mutat. 35: 1033–
328. 1045.
262 Molecular Hematology

Uff, S., Clemetson, J.M., Harrison, T. et al. (2002). Crystal structure of Platelet-type von Willebrand disease
the platelet glycoprotein Ibα N-terminal domain reveals an unmask-
ing mechanism for receptor activation. J. Biol. Chem. 277: 35657– Matsubara, Y., Murata, M., Sugita, K., and Ikeda, Y. (2003). Identification
35663. of a novel point mutation in platelet glycoprotein Ibα Gly to Ser at
residue 233, in a Japanese family with platelet-type von Willebrand
disease. J. Thromb. Haemost. 1: 2198–2205.
New GPIbα mutations Nurden, P., Lanza, F., Bonnafous-Faurie, C., and Nurden, A. (2007).
A second report of platelet-type von Willebrand disease with a
Imai, C., Kunishima, S., Takachi, T. et al. (2009). A novel homozygous 8- Gly233Ser mutation in the GPIBA gene. Thromb. Haemost. 97: 319–
base pair deletion mutation in the glycoprotein Ibα gene in a patient 321.
with Bernard–Soulier syndrome. Blood Coagul. Fibrinolysis 20: 470– Othman, M., Notley, C., Lavender, F.L. et al. (2005). Identification
474. and functional characterization of a novel 27-bp deletion in the
Noris, P., Perrotta, S., Bottega, R. et al. (2012). Clinical and laboratory macroglycopeptide-coding region of the GPIBA gene resulting in
features of 103 patients from 42 Italian families with inherited throm- platelet-type von Willebrand disease. Blood 105: 4330–4336.
bocytopenia derived from the monoallelic Ala156Val mutation of Suva, L.J., Hartman, E., Dilley, J.D. et al. (2008). Platelet dysfunction and
GPIbα (Bolzano mutation). Haematologica 97: 82–88. a high bone mass phenotype in a murine model of platelet-type von
Rosenberg, N., Lalezari, S., Landau, M. et al. (2007). Trp207Gly in Willebrand disease. Am. J. Pathol. 172: 430–439.
platelet glycoprotein Ibα is a novel mutation that disrupts the connec-
tion between the leucine-rich repeat domain and the disulfide loop
structure and causes Bernard–Soulier syndrome. J. Thromb. Haemost. Collagen receptor defects α2β1 integrin
5: 378–386. Joutsi-Korhonen, L., Smethurst, P.A., Rankin, A. et al. (2003). The low-
Vettore, S., Scandellari, R., Moro, S. et al. (2008). Novel point mutation frequency allele of the platelet collagen signaling receptor glycopro-
in a leucine-rich repeat of the GPIbα chain of the platelet von Wille- tein VI is associated with reduced functional responses and expres-
brand factor receptor, GPIb/IX/V, resulting in an inherited dominant sion. Blood 101: 4372–4379.
form of Bernard–Soulier syndrome affecting two unrelated families: Sarratt, K.L., Chen, H., Zutter, M.M. et al. (2005). GPVI and α2β1 play
the N41H variant. Haematologica 93: 1743–1747. independent critical roles during platelet adhesion and aggregate for-
mation to collagen under flow. Blood 106: 1268–1277.

New GPIbβ mutations


GPVI
Hadjkacem, B., Elleuch, H., Gargouri, J., and Gargouri, A. (2009).
Bernard–Soulier syndrome: novel nonsense mutation in GPIbβ gene Arthur, J.F., Dunkley, S., and Andrews, R.K. (2007). Platelet glycopro-
affecting GPIb–IX complex expression. Ann. Hematol. 88: 465–472. tein VI-related clinical defects. Br. J. Haematol. 139: 363–372.
Dumont, B., Lasne, D., Rothschild, C. et al. (2009). Absence of collagen-
induced platelet activation caused by compound heterozygous GPVI
mutations. Blood 114: 1900–1903.
Velocardiofacial syndrome
Hermans, C., Wittevrongel, C., Thys, C. et al. (2009). A compound het-
Liang, H.P., Morel-Kopp, M.C., Curtin, J. et al. (2007). Heterozygous loss erozygous mutation in glycoprotein VI in a patient with a bleeding
of platelet glycoprotein (GP)Ib-V-IX variably affects platelet function disorder. J. Thromb. Haemost. 7: 1356–1363.
in velocardiofacial syndrome (VCFS) patients. Thromb. Haemost. 98: Hughan, S.C., Senis, Y., Best, D. et al. (2005). Selective impairment of
1298–1308. platelet activation to collagen in the absence of GATA1. Blood 105:
4369–4376.

New GPIX mutations Glanzmann thrombasthenia overview


Kunishima, S., Yamada, T., Hamaguchi, M., and Saito, H. (2006). Fang, J., Hodivala-Dilke, K., Johnson, B.D. et al. (2005). Therapeu-
Bernard–Soulier syndrome due to GPIX W127X mutation in Japan tic expression of the platelet-specific integrin, αIIbβ3, in a murine
is frequently misdiagnosed as idiopathic thrombocytopenic purpura. model for Glanzmann thrombasthenia. Blood 106: 2671–2679.
Int. J. Hematol. 83: 366–367. French, D.L. (1998). The molecular genetics of Glanzmann’s thrombas-
thenia. Platelets 9: 5–20.
Jayandharan, G., Nelson, E.J., Baidya, S. et al. (2007). A new multi-
Variant forms plex PCR and conformation-sensitive gel electrophoresis strategy for
mutation detection in the platelet glycoprotein αIIb and β3 genes. J.
Balduini, A., Malara, A., Pecci, A. et al. (2009). Proplatelet formation Thromb. Haemost. 5: 206–209.
in heterozygous Bernard–Soulier syndrome type Bolzano. J. Thromb. Manickam, N., Sun, X., Li, M. et al. (2008). Protein disulphide isomerase
Haemost. 7: 478–484. in platelet function. Br. J. Haematol. 140: 223–229.
Vettore, S., Scandellari, R., Scapin, M. et al. (2008). A case of Bernard– Nurden, A.T., Pillois, X., Fiore, M. et al. (2015). Expanding the mutation
Soulier syndrome due to a homozygous four bases deletion (TGAG) spectrum affecting αIIbβ3 Integrin in Glanzmann thrombasthenia:
of GPIbα gene: lack of GPIbα but absence of bleeding. Platelets 19: screening of the ITGA2B and ITGB3 genes in a large international
388–391. cohort. Hum. Mutat. 36: 548–561.
Platelet disorders 263

αIIb defects ADP and ATP receptor defects


Losonczy, G., Rosenberg, N., Boda, Z. et al. (2007). Three novel muta- Cattaneo, M. (2006). Disorders of platelet function. Abnormalities of
tions in the glycoprotein IIb gene in a patient with type II Glanzmann the platelet P2 receptors. Pathophysiol. Haemost. Thromb. 35: 10–14.
thrombasthenia. Haematologica 92: 698–701. Daly, M.E., Dawood, B.B., Lester, W.A. et al. (2009). Identification and
Vijapurkar, M., Ghosh, K., and Shetty, S. (2009). Novel mutations in characterization of a novel P2Y12 variant in a patient diagnosed with
GPIIb gene in Glanzmann’s thrombasthenia from India. Platelets 20: type 1 von Willebrand disease in the European MCMDM-1VWD
35–40. study. Blood 113: 4110–4113.
Leon, C., Hechler, B., Freund, M. et al. (1999). Defective platelet aggre-
gation and increased resistance to thrombosis in purinergic P2Y1
β3 defects receptor-null mice. J. Clin. Investig. 104: 1731–1737.
Oury, C., Toth-Zsamboki, E., Vermylen, J., and Hoylaerts, M.F. (2002).
Chen, P., Melchior, C., Brons, N.H. et al. (2001). Probing conformational
Does the P2X1del variant lacking 17 amino acids in its extracellular
changes in the I-like domain and the cysteine-rich repeat of human
domain represent a relevant functional ion channel in platelets? Blood
α3 integrins following disulfide bond disruption by cysteine muta-
99: 2275–2277.
tions: identification of cysteine 598 involved in αIIbβ3 activation. J.
Oury, C., Toth-Zsamboki, E., Vermylen, J., and Hoylaerts, M.F. (2006).
Biol. Chem. 276: 38628–38635.
The platelet ATP and ADP receptors. Curr. Pharm. Des. 12: 859–875.
D’Andrea, G., Bafunno, V., Del Vecchio, L. et al. (2008). A α3
Remijn, J.A., Ijsseldijk, M.J., Strunk, A.L. et al. (2007). Novel molecular
Asp217αVal substitution in a patient with variant Glanzmann throm-
defect in the platelet ADP receptor P2Y12 of a patient with haemor-
basthenia severely affects integrin αIIbβ3 functions. Blood Coagul.
rhagic diathesis. Clin. Chem. Lab. Med. 45: 187–189.
Fibrinolysis 19: 657–662.
Gonzalez-Manchon, C., Butta, N., Larrucea, S. et al. (2004). A
variant thrombasthenic phenotype associated with compound
Other primary agonist receptor defects
heterozygosity of integrin α3-subunit: (Met124Val)α3 alters the
subunit dimerization rendering a decreased number of con- Edelstein, L.C., Simon, L.M., Lindsay, C.R. et al. (2014). Common vari-
stitutive active αIIbβ3 receptors. Thromb. Haemost. 92: 1377– ants in the human platelet PAR4 thrombin receptor alter platelet
1386. function and differ by race. Blood 124: 3450–3458.
Liu, J., Jackson, C.W., Gruppo, R.A. et al. (2005). The α3 subunit of the Hirata, T., Kakizuka, A., Ushikubi, F. et al. (1994). Arg60 to Leu mutation
integrin αIIbβ3 regulates αIIb-mediated outsidein signaling. Blood of the human thromboxane A2 receptor in a dominantly inherited
105: 4345–4352. bleeding disorder. J. Clin. Investig. 94: 1662–1667.
Mor-Cohen, R., Rosenberg, N., Peretz, H. et al. (2007). Disulfide bond
disruption by a α3-Cys549Arg mutation in six Jordanian fami-
lies with Glanzmann thrombasthenia causes diminished produc- Intracellular signaling pathway defects
tion of constitutively active αIIbβ3. Thromb. Haemost. 98: 1257–
1265. Bellucci, S., Huisse, M.G., Boval, B. et al. (2005). Defective collagen-
Nurden, P., Poujol, C., Winckler, J. et al. (2002). A Ser752βPro substi- induced platelet activation in two patients with malignant
tution in the cytoplasmic domain of α3 in a Glanzmann thrombas- haemopathies is related to a defect in the GPVI-coupled signalling
thenia variant fails to prevent interactions between the αIIbβ3 inte- pathway. Thromb. Haemost. 93: 130–138.
grin and the platelet granule pool of fibrinogen. Br. J. Haematol. 118: Dunkley, S., Arthur, J.F., Evans, S. et al. (2007). A familial platelet func-
1143–1151. tion disorder associated with abnormal signalling through the glyco-
Ruiz, C., Liu, C.Y., Sun, Q.H. et al. (2001). A point mutation in the protein VI pathway. Br. J. Haematol. 137: 569–577.
cysteine-rich domain of glycoprotein (GP) IIIa results in the expres- Mao, G.F., Vaidyula, V.R., Kunapuli, S.P., and Rao, A.K. (2002). Lineage-
sion of a GPIIb-IIIa (αIIbβ3) integrin receptor locked in a high- specific defect in gene expression in human platelet phospholipase
affinity state and a Glanzmann thrombasthenia-like phenotype. Blood C-α2 deficiency. Blood 99: 905–911.
98: 2432–2441. Patel, Y.M., Patel, K., Rahman, S. et al. (2003). Evidence for a role
Wang, R., Shattil, S.J., Ambruso, D.R., and Newman, P.J. (1997). Trunca- for Gαi1 in mediating weak agonist-induced platelet aggregation in
tion of the cytoplasmic domain of α3 in a variant form of Glanzmann human platelets: reduced Gαi1 expression and defective Gi signaling
thrombasthenia abrogates signaling through the integrin αIIbβ3 in the platelets of a patient with a chronic bleeding disorder. Blood
complex. J. Clin. Investig. 100: 2393–2403. 101: 4828–4835.

Kindlins and leukocyte adhesion defect Platelet secretion defects (storage pool
disease): gray platelet syndrome
Pasvolsky, R., Feigelson, S.W., Kilic, S.S. et al. (2007). A LAD-III
syndrome is associated with defective expression of the Rap-1 Albers, C.A., Cvejic, A., Favier, R. et al. (2011). Exome sequencing iden-
activator CalDAG-GEFI in lymphocytes, neutrophils, and platelets. tifies NBEAL2 as the causative gene for gray platelet syndrome. Nat.
J. Exp. Med. 204: 1571–1582. Genet. 43: 735–737.
Rognoni, E., Ruppert, R., and Fässler, R. (2016). The kindlin family: Deppermann, C., Cherpokova, D., Nurden, P. et al. (2013). Gray platelet
functions, signaling properties and implications for human disease. syndrome and defective thrombo-inflammation in Nbeal2-deficient
J. Cell Sci. 129: 17–27. mice. J. Clin. Investig. 123: 3331–3342.
264 Molecular Hematology

Drouin, A., Favier, R., Masse, J.M. et al. (2001). Newly recognized cellu- Wiskott–aldrich syndrome
lar abnormalities in the gray platelet syndrome. Blood 98: 1382–1391.
Gunay-Aygun, M., Falik-Zaccai, T.C., Vilboux, T. et al. (2011). NBEAL2 Imai, K., Nonoyama, S., and Ochs, H.D. (2003). WASP (Wiskott–
is mutated in gray platelet syndrome and is required for biogenesis of Aldrich syndrome protein) gene mutations and phenotype. Curr.
platelet α-granules. Nat. Genet. 43: 732–734. Opin. Allergy Clin. Immunol. 3: 427–436.
Kahr, W.H., Hinckley, J., Li, L. et al. (2011). Mutations in NBEAL2, Lutskiy, M.I., Shcherbina, A., Bachli, E.T. et al. (2007). WASP localizes
encoding a BEACH protein, cause gray platelet syndrome. Nat. Genet. to the membrane skeleton of platelets. Br. J. Haematol. 139: 98–105.
43: 738–740. Ochs, H.D., Filipovich, A.H., Veys, P. et al. (2008). Wiskott–Aldrich syn-
Nurden, A.T. and Nurden, P. (2007). The gray platelet syndrome: clinical drome: diagnosis, clinical and laboratory manifestations, and treat-
spectrum of the disease. Blood Rev. 21: 21–36. ment. Biol. Blood Marrow Transplant. 15 (1 Suppl): 84–90.
Nurden, P., Jandrot-Perrus, M., Combrie, R. et al. (2004). Severe defi- Sabri, S., Foudi, A., Boukour, S. et al. (2006). Deficiency in the Wiskott–
ciency of glycoprotein VI in a patient with gray platelet syndrome. Aldrich protein induces premature proplatelet formation and platelet
Blood 104: 107–114. production in the bone marrow compartment. Blood 108: 134–
140.

Quebec platelet disorder


Procoagulant regulation defects
Diamandis, M., Veljkovic, D.K., Maurer-Spurej, E. et al. (2008). Quebec
platelet disorder: features, pathogenesis and treatment. Blood Coagul. Solum, N.O. (1999). Procoagulant expression in platelets and defects
Fibrinolysis 19: 109–119. leading to clinical disorders. Arterioscler. Thromb. Vasc. Biol. 19:
Veljkovic, D.K., Rivard, G.E., Diamandis, M. et al. (2009). Increased 2841–2846.
expression of urokinase plasminogen activator in Quebec platelet dis- Weiss, H.J. (2009). Impaired platelet procoagulant mechanisms in
order is linked to megakaryocyte differentiation. Blood 113: 1535– patients with bleeding disorders. Semin. Thromb. Hemost. 35: 233–
1542. 241.

Hermansky–pudlak syndrome Scott syndrome


Gautam, R., Novak, E.K., Tan, J. et al. (2006). Interaction of Hermansky– Bettache, N., Gaffet, P., Allegre, N. et al. (1998). Impaired redistribu-
Pudlak syndrome genes in the regulation of lysosome-related tion of aminophospholipids with distinctive cell shape change dur-
organelles. Traffic 7: 779–792. ing Ca2 + −induced activation of platelets from a patient with Scott
Huizing, M., Parkes, J.M., Helip-Wooley, A. et al. (2007). Platelet α- syndrome. Br. J. Haematol. 101: 50–58.
granules in BLOC-2 and BLOC-3 subtypes of Hermansky–Pudlak Castoldi, E., Collins, P.W., Williamson, P.L., and Bevers, E.M. (2011).
syndrome. Platelets 18: 150–157. Compound heterozygosity for 2 novel TMEM16F mutations in a
Spritz, R.A. (1998). Molecular genetics of the Hermansky–Pudlak and patient with Scott syndrome. Blood 117: 4399–4400.
Chediak–Higashi syndromes. Platelets 9: 21–29. van Geffen, J.P., Swieringa, F., and Heemskerk, J.W. (2016). Platelets
Walker, M., Payne, J., Wagner, B., and Vora, A. (2007). Hermansky– and coagulation in thrombus formation: aberrations in the Scott syn-
Pudlak syndrome. Br. J. Haematol. 138: 671. drome. Thromb. Res. 141 (Suppl 2): S12–S16.
Kunzelmann, K., Nilius, B., Owsianik, G. et al. (2014). Molecular func-
tions of anoctamin 6 (TMEM16F): a chloride channel, cation chan-
Chédiak–higashi syndrome nel, or phospholipid scramblase? Pflugers Arch. 466: 407–414.
Suzuki, J., Umeda, M., Sims, P.J., and Nagata, S. (2010). Calcium-
Kaplan, J., De Domenico, I., and Ward, D.M. (2008). Chediak–Higashi
dependent phospholipid scrambling by TMEM16F. Nature 468: 834–
syndrome. Curr. Opin. Hematol. 15: 22–29.
838.
Nagle, D.L., Karim, M.A., Woolf, E.A. et al. (1996). Identification and
mutation analysis of the complete gene for Chediak–Higashi syn-
drome. Nat. Genet. 14: 307–311. Stormorken syndrome
Westbroek, W., Adams, D., Huizing, M. et al. (2007). Cellular defects in
Chediak–Higashi syndrome correlate with the molecular genotype Misceo, D., Holmgren, A., Louch, W.E. et al. (2014). A dominant STIM1
and clinical phenotype. J. Investig. Dermatol. 127: 2674–2677. mutation causes Stormorken syndrome. Hum. Mutat. 35: 556–564.
Stormorken, H., Holmsen, H., Sund, R. et al. (1995). Studies on the
haemostatic defect in a complicated syndrome. An inverse Scott syn-
Griscelli syndrome drome platelet membrane abnormality? Thromb. Haemost. 74: 1244–
Barral, D.C., Ramalho, J.S., Anders, R. et al. (2002). Functional redun- 1251.
dancy of Rab27 proteins and the pathogenesis of Griscelli syndrome.
J. Clin. Investig. 110: 247–257. Giant platelet syndromes and cytoskeletal
Novak, E.K., Gautam, R., Reddington, M. et al. (2002). The regulation defects
of platelet-dense granules by Rab27a in the ashen mouse, a model of
Hermansky–Pudlak and Griscelli syndromes, is granule-specific and Althaus, K. and Greinacher, A. (2009). MYH9-related platelet disorders.
dependent on genetic background. Blood 100: 128–135. Semin. Thromb. Hemost. 35: 189–203.
Platelet disorders 265

Chen, Z. and Shivdasani, R.A. (2009). Regulation of platelet biogene- occurrence of t(12 : 21) positive pre-B acute lymphoblastic leukemia.
sis: insights from the May–Hegglin anomaly and other MYH9-related Blood Cell Mol. Dis. 39: 107–114.
disorders. J. Thromb. Haemost. 7: 272–276. Freson, K., Devriendt, K., Matthijs, G. et al. (2001). Platelet characteris-
Leon, C., Eckly, A., Hechler, B. et al. (2007). Megakaryocyte-restricted tics in patients with X-linked macrothrombocytopenia because of a
MYH9 inactivation dramatically affects hemostasis while preserving novel GATA1 mutation. Blood 98: 85–92.
platelet aggregation and secretion. Blood 110: 3183–3191. Sun, L., Gorospe, J.R., Hoffman, E.P., and Rao, A.K. (2007). Decreased
Nurden, A.T., Federici, A.B., and Nurden, P. (2009). Altered megakary- platelet expression of myosin regulatory light chain polypep-
ocytopoiesis in von Willebrand type 2B disease. J. Thromb. Haemost. tide (MYL9) and other genes with platelet dysfunction and
7: 277–281. CBFA2/RUNX1 mutation: insights from platelet expression profiling.
Provaznikova, D., Geierova, V., Kumstyrova, T. et al. (2009). Clinical J. Thromb. Haemost. 5: 146–154.
manifestation and molecular genetic characterization of MYH9 dis- White, J.G. (2007). Platelet pathology in carriers of the X-linked GATA-
orders. Platelets 26: 1–8. 1 macrothrombocytopenia. Platelets 18: 620–627.
Saito, H., Matsushita, T., Yamamoto, K. et al. (2005). Giant platelet syn-
drome. Hematology 10: 41–46.
Platelet protein polymorphisms and
Stächele, J., Bakchoul, T., Najm, J. et al. (2015). Congenital macrothrom-
tendency to thrombosis
bocytopenia associated with a combination of functional poly-
morphisms in the TUBB1 gene. Hamostaseologie 35 (Suppl 1): Jones, C.I., Bray, S., Garner, S.F. et al. (2009). A functional genomics
S18–S21. approach reveals novel quantitative trait loci associated with platelet
Toren, A., Rozenfeld-Granot, G., Heath, K.E. et al. (2003). MYH9 spec- signalling pathways. Blood 114: 1405–1416.
trum of autosomal-dominant giant platelet syndromes: unexpected Martini, C.H., Doggen, C.J., Cavallini, C. et al. (2005). No effect of poly-
association with fibulin-1 variant-D inactivation. Am. J. Hematol. 74: morphisms in prothrombotic genes on the risk of myocardial infarc-
254–262. tion in young adults without cardiovascular risk factors. J. Thromb.
White, J.G. (2007). Platelet pathology in carriers of the X-linked GATA- Haemost. 3: 177–179.
1 macrothrombocytopenia. Platelets 18: 620–627. Mega, J.L., Close, S.L., Wiviott, S.D. et al. (2009). Cytochrome P-450
polymorphisms and response to clopidogrel. N. Engl. J. Med. 360:
Transcription factor defects 354–362.
Meisel, C., Lopez, J.A., and Stangl, K. (2004). Role of platelet glycopro-
Breton-Gorius, J., Favier, R., Guichard, J. et al. (1995). A new congeni- tein polymorphisms in cardiovascular diseases. Naunyn Schmiede-
tal dysmegakaryopoietic thrombocytopenia (Paris–Trousseau) asso- bergs Arch Pharmacol. 369: 38–54.
ciated with giant platelet α-granules and chromosome 11 deletion at Ouwehand, W.H. (2007). Platelet genomics and the risk of atherothrom-
11q23. Blood 85: 1805–1814. bosis. J. Thromb. Haemost. 5: 188–195.
Escher, R., Wilson, P., Carmichael, C. et al. (2007). A pedigree with auto- Ouwehand, W.H. (2009). The discovery of genes implicated in myocar-
somal dominant thrombocytopenia, red cell macrocytosis, and an dial infarction. J. Thromb. Haemost. 7: 305–307.
Chapter 20 The molecular basis of blood
cell alloantigens
Cristina Navarrete1,2 , Louise Tilley3 , Winnie Chong4 & Colin J. Brown5,6
1
Histocompatibility and Immunogenetics Service Development Department, NHS Blood and Transplant, London, UK
2
Department of Immunology and Molecular Pathology, University College London, London, UK
3 International Blood Group Reference Laboratory, NHS Blood and Transplant, Bristol, UK
4
Histocompatibility and Immunogenetics, Service Development Laboratory, NHS Blood and Transplant, London, UK
5 Histocompatibility and Immunogenetics Laboratory, NHS Blood and Transplant, London, UK
6
Faculty of Life Sciences & Medicine, King’s College London, London, UK

Introduction, 267 Neonatal alloimmune thrombocytopenia, 277


Identification of HLA gene polymorphism, 268 Neonatal alloimmune neutropenia, 281
HLA antigens, 271 Conclusions, 283
Hemolytic disease of the fetus and newborn, 273 Further reading, 283

ited to one type of blood cell, destruction of cells in the


Introduction newborn by maternal blood cell alloantibodies may lead
to the development of immune-mediated anemia, throm-
The transfusion of blood and fetal–maternal hemorrhage
bocytopenia, or neutropenia of the newborn. In contrast,
during pregnancy have provided unique models for study-
HLA class I alloantibodies do not cause cytopenias in the
ing the immune response against a plethora of polymorphic
newborn, but may compromise the effectiveness of platelet
blood cell surface markers, including the alloantigens of the
transfusions, complicate organ transplantation, cause febrile
human leukocyte antigen (HLA) system. Many blood cell
non-hemolytic transfusion reactions, or, on rare occasions,
membrane determinants show allelic variation, which can
precipitate transfusion-related acute lung injury (TRALI)
elicit the formation of alloantibodies. In nearly all transfusion
and occasionally delay engraftment in hemopoietic stem cell
situations and pregnancies, the recipient’s immune system is
transplantation.
challenged by blood cells mismatched for multiple alloanti-
The formation of alloantibodies after an incompatible
gen systems. As the difference between self and non-self is
challenge in the form of blood transfusion is more the excep-
limited, alloantibodies are only formed by a subset of recipi-
tion than the rule. In contrast to our detailed understand-
ents. Red cell alloantibodies are detected in 1–1.5% of preg-
ing of the molecular basis of blood cell alloantigens, we
nant women and in 2–3% of transfused individuals, and can
remain relatively ignorant about the mechanism of non-
increase significantly in multitransfused patients. The HLA
responsiveness. We have learned from animal experiments
alloantigens are more immunogenic than those of the red
that restriction in the ability to mount an immune response
cells, and 15–25% of multiparous women and 30–40% of
is largely controlled by genes of the major histocompati-
patients on long-term prophylactic platelet transfusions are
bility complex (MHC) or HLA. However, the reason why,
positive for HLA class I antibodies.
for example, some 25% of RhD-negative individuals fail to
Alloantigens were initially defined as polymorphic mem-
mount an anti-D response on repeated challenge with RhD-
brane determinants identified by polyclonal alloantibodies
positive red cells remains elusive. An exception to this is our
in serum samples from alloimmunized patients or pregnant
detailed understanding of the immune response against the
women, but the molecular basis of most alloantigens has
HPA-1a alloantigen on platelets. There is a near-complete
now been resolved and it is possible to use DNA-based tech-
restriction on the ability to form HPA-1a antibodies by the
niques to further characterize their polymorphism. Alloanti-
HLA class II allele DRB3*01:01. However, except for this
gens can be categorized as those shared by all blood cells,
example, our ability to identify the genes controlling the risk
for example HLA class I, and those unique to one blood
of alloimmunization remains limited, and further research
cell type, such as Rh on red cells and human platelet anti-
is needed to identify the genetic basis of this variability in
gen (HPA) on platelets (Table 20.1). When expression is lim-
responsiveness.
This chapter reviews the recent developments in the
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. molecular aspects of blood cell alloantigens and highlights
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. their impact on clinical management. Recognizing the wide

267
268 Molecular Hematology

Table 20.1 Antigen expression on peripheral blood cells

Antigens Erythrocytes Platelets Neutrophils B lymphocytes T lymphocytes Monocytes

A, B, H +++ + +/(+) − − − −
I +++ ++ ++ − − −
Rh* +++ − − − − −
K +++ − − − − −
HLA class I −/(+) +++ +++ +++ +++ +++
HLA class II − − −/+ + +† +++ −/+ + +† +++
GPIIb/IIIa − +++ (+)‡ − − −
GPIa/IIa − +++ − − ++ −
GPIb/IX/V − +++ − − − −
CD109 (+)/+ +† − − (−)/+ +† (+)
FcγRIIIB (CD16b) − − +++ − − −/+ + + §*%
CD177 − − + + +# − − −
CTL2 − ? +++ + +$ + +$ ?
CD11b − − ++ − − + +&
CD11a − − ++ − − ++

+, + +, + + + Level of antigen expression in increasing order. (+) Weak expression. ? Not known. * Non-glycosylated. † On activated cells.
‡ GPIIIa in association with an alternative α chain αv. § When differentiated to macrophages expressing FcγRIIIA. # Expressed on subpopulation
of neutrophils. & Also expressed on natural killer cells. $ B and T lymphocytes not separated. % Expressed on subpopulation of monocytes. GP,
glycoprotein; HLA, human leukocyte antigen

variety of clinical conditions in which the HLA alloantigens Molecular typing techniques
play a role, we have placed the main emphasis on:
Initially, the detection and characterization of the HLA
r antibody-mediated cytopenias in the newborn by maternal molecules and polymorphisms were carried out using sero-
blood cell–specific alloantibodies; logical and cellular techniques. With the development of gene
r the complication of HLA class I alloimmunization in cloning and DNA-based molecular techniques, it is now pos-
patients receiving prophylactic platelet transfusions. The sible to perform a detailed analysis of these molecules at
HLA antigens have also been used to introduce the molec- the nucleotide level. The result of this analysis has shown
ular techniques currently utilized to identify alleles of the existence of shared nucleotide sequence motifs between
genes. alleles of the same and/or different loci. Similarly, it has
been shown that there are certain locus-specific nucleotide
sequences in both the coding regions (exons) and the non-
Identification of HLA gene coding regions (introns) of the various genes.
polymorphism DNA sequencing of many HLA alleles has demonstrated
that most of the polymorphisms are located in exons 2 and 3
The impact of molecular biological techniques on our ability of HLA class I and exon 2 of HLA class II molecules. These
to scan and identify allelic variation of human genes is best exons code for the distal membrane domains of the HLA
exemplified by the HLA system. For decades the enormous molecules, which form the peptide-binding groove (Fig-
diversity of the alloantigens of the HLA system has presented ure 20.1). Based on this information, a number of techniques
both technical and clinical challenges. It is well documented have been developed to identify this polymorphism using the
that the use of molecular techniques, including sequencing- polymerase chain reaction (PCR). These include HLA typ-
based high-resolution typing, is contributing to improved ing using PCR-SSOP (polymerase chain reaction–sequence-
outcome in transplant patients. Improved matching allows specific oligonucleotide probes), PCR-SSP (polymerase chain
graft maintenance at lower levels of immunosuppression, reaction–sequence-specific primers), sequencing-based typ-
which is of great importance with the emerging evidence ing (SBT), and next-generation sequencing (NGS).
that long-term use of potent immunosuppressive drugs is not
without side effects. The following sections review a number
PCR-SSOP
of current molecular techniques used to define alleles of the
HLA genes. The same techniques may be used to define allelic In this technique, the gene of interest is amplified by PCR
variation in genes encoding other blood cell alloantigens. using generic primers complementary to highly conserved
The molecular basis of blood cell alloantigens 269

α1 α2

Peptide-
binding
region S
S
N
β2–m α3
Immunoglobulin- S S
like region
S S

Transmembrane
region
P
Cytoplasmic P
P
region C
3'UT
Class IA

Regulatory Leader Exon Exon Exon Transmembrane and


(a) sequences sequence 2 3 4 cytoplasmic

α1 β1

Peptide-
binding
region NN S

α2 β2
Immunoglobulin- S S
like region
S S
Fig. 20.1 (a) Schematic presentation of human
Papain cleavage Papain cleavage
leukocyte antigen (HLA) class I. The sites sites
non-covalent association between the HLA class I
protein (with three immunoglobulin-like domains, Transmembrane
region
α1 , α2 , and α3 ) and β2 -microglobulin (β2 -m) is
shown. The three α domains are encoded by three Cytoplasmic
exons of the HLA class IA gene on chromosome 6. region C C 3'UT
(b) Schematic presentation of HLA class II. The
α and β chains of the HLA class II protein (each Class IIA
with two distinct immunoglobulin-like domains, α1 Regulatory Leader Exon 2 Exon 3 Transmembrane and
and α2 , and β1 and β2 ) are non-covalently sequences sequence cytoplasmic
associated. Both domains of each chain are
Class IIB
encoded by their respective exons of the α and β
class II genes on chromosome 6. (b) Exon 2 Exon 3

gene segments. The PCR products are then immobilized onto A modification called the reverse SSOP technique is more
support membranes (e.g. nylon membranes) and analyzed widely used, whereby the relevant PCR product is labeled
by probing the membranes with labeled oligonucleotides with biotin and, following the denaturation and neutraliza-
designed to anneal with polymorphic sequences present in tion steps, the product is hybridized with oligonucleotide-
the HLA alleles. By scoring the probes that bind to specific specific probes coupled to fluorescent beads. Following
regions, it is possible to assign an HLA type. the addition of r-phycoerythrin-conjugated streptavidin
270 Molecular Hematology

(SAPE) to the reaction, the fluorescence is then measured method is the presence or absence of an amplicon for which
using a flow cytometer-like instrument, the Luminex. a specific primer was used. PCR-SSP was first developed to
Reverse SSOP is particularly amenable to batch testing define the various HLA-DRB3 alleles (Figure 20.2). Although
large number of samples rather than single sample testing, this is a rapid technique, many PCR reactions have to be
and was a popular technique used by stem cell registries. set up for each sample, for example 24 reactions for low-
resolution DR typing. An obvious advantage of PCR-SSP is
that, as two sequences are detected in cis, ambiguities which
PCR-SSP
may arise from PCR-SSOP typing can be resolved. For PCR-
This technique involves the use of sequence-specific primers SSP typing, however, the target sequence of the alleles must
in the PCR. The amplified DNA product is detected by gel be known, and novel unknown sequences may not always be
electrophoresis and this allows the rapid identification of the detected. PCR-SSP is also the technique of choice for HPA
HLA alleles in individual samples, since the read-out of this typing (see The Molecular Basis of Platelet-Specific Antigens or

Allele-specific PCR (PCR–SSP)

Complete complementarity of PCR primers

Amplification
A
PCR primer mismatch 3'-end

No amplification
B

A B

Control: non-polymorphic PCR


product
Fig. 20.2 (a) Polymerase chain
reaction–sequence-specific primers (PCR-SSP).
With allele-specific PCR, specificity is obtained
Allele-specific PCR product
during the PCR. A single nucleotide mismatch at
the 3′ -end of the allele-specific primer (B primer in
the figure) will prevent the polymerase from
(a) commencing DNA amplification. Therefore, no
amplification of template DNA will occur with the B
HLA–DR/DQ allele primer, whereas with the A allele primer a
PCR–SSP
product is obtained. Ethidium bromide is used to
reveal amplified DNA with ultraviolet light after
DNA gel electrophoresis. Allele-specific DNA is
HLA–DR1, DR13 obtained in the A reaction (lower band) but not in
DRB3 the B reaction. In both reactions a control PCR
product is generated by amplification of a segment
of the human growth hormone gene (upper two
bands). (b) PCR-SSP for DR and DQ alleles.
Results of PCR-SSP with template DNA from a
single donor. Each lane represents the result of gel
HLA–DQ5, DQ6 electrophoresis of a single PCR reaction with one of
the allele-specific DR or DQ primers. From the
pattern of positive results, a DR (upper panel) and
DQ (lower panel) type can be concluded, in this
(b) case DR1, DR13, DRB3, and DQ5, DQ6.
The molecular basis of blood cell alloantigens 271

HPA). Using the PCR-SSP technique, it is possible to deter- and graft-versus-host disease. The genes coding for these
mine whether the detected sequences are in cis or in trans. molecules form part of a complex genetic system called the
MHC, located on the short arm of chromosome 6. This
region spans a distance of approximately 4000 kb and is
Sequencing-based typing
divided into HLA class I, class II, and class III genes. Class
The principle of DNA sequencing is relatively straight- III includes a group of non-MHC genes coding for proteins
forward. It involves the denaturation of the DNA to be with various immunological functions, such as the comple-
analyzed to provide a single-strand template; a sequenc- ment factor 4 and tumor necrosis factor (TNF)-α.
ing primer is then added and the extension is performed The development of recombinant DNA technology has led
by the addition of polymerase in the presence of excess to increased understanding of the genetic complexity, struc-
nucleotides. The sequencing mixture is divided into four ture, and function of the HLA genes and molecules.
tubes, each of which contains a specific dideoxyribonucleo-
side triphosphate (ddATP). When this is incorporated into
HLA class I genes
the DNA strand, elongation is interrupted, leading to chain
termination. In each reaction there is random incorpora- The HLA class I genes have been classified according to their
tion of the chain terminators and therefore products of structure and function as classical and non-classical, or class
all sizes are generated. The products of the four reactions Ib genes. The classical HLA class I genes, HLA-A, HLA-B,
are then analyzed by electrophoresis in parallel lanes of a and HLA-C, code for heterodimers formed by a heavy (α)
polyacrylamide–urea gel, and the sequence is read by com- chain of approximately 43 kDa, non-covalently linked to the
bining the results of each lane using an automated DNA β2 -microglobulin light chain of 12 kDa. The latter is coded
sequencer. for by a gene located outside the HLA region on chromo-
some 15. The extracellular portion of the α chain has three
domains (α1 , α2 , and α3 ) encoded by exons 2, 3, and 4,
Next-generation sequencing
respectively. Each domain is approximately 90 amino acids
NGS involves PCR amplification of the required region, gene, in length. The transmembrane and cytoplasmic domains
or exon, and pooling of the resulting amplicons. Two main are encoded by exons 5, 6, and 7, respectively. The β2 -
approaches have been used in the application of NGS for the microglobulin, which confers stability on the molecule, is
definition of HLA alleles. The first involves the amplifica- non-covalently linked to the α3 domain (see Figure 20.1a).
tion and sequencing of the exons containing the polymor- The α1 and α2 domains are the most polymorphic regions
phic positions of the required genes (exons 2 and 3 for HLA of these molecules and form a groove consisting of two α
class I, A, B, and C; and exon 2 for HLA class II DR, DQ, helices, with an antiparallel-running β-pleated sheet forming
and DP). The second approach involves the amplification and the floor of the groove. This groove, which is approximately
sequencing of the whole gene of interest. The main advantage 2.5 nm long and 1 nm wide, can accommodate a variety of
of this latter approach is that it allows the detection of poly- antigen-derived peptides of about 8–10 amino acids to be
morphisms in all exons and introns of the relevant gene, thus presented to T cells.
removing HLA typing ambiguities obtained with the exon- In addition to the classical HLA class I genes, the
based approach. The amplification step is followed by the non-classical HLA class I genes are also located in this
fragmentation of these amplicons and the ligation of adapters region. They include HLA-E, HLA-F, and HLA-G and their
and unique indexes or identifiers, a step also known as library exon/intron organization is similar to that of the classical
preparation. The fragmented and ligated products are then class I genes, but they have a more restricted polymorphism.
clonally amplified and sequenced. NGS generates hundreds The HLA class I genes are expressed on most tissues and
of thousands of sequences, and sorting and aligning of these blood cells, including T and B lymphocytes and platelets
sequences to the reference genome can be extremely chal- (Table 20.1). The non-classical class I genes HLA-E and HLA-
lenging; to achieve this, complex analysis algorithms have F are expressed on most tissues tested so far, whereas HLA-G
been developed. has to date only been detected on trophoblasts and mono-
cytes.
Two MHC class I chain-related genes (MIC-A and MIC-B),
HLA antigens located centromeric to HLA-B, have been described. Unlike
the classical and non-classical HLA class I, MIC genes do not
These are a group of highly polymorphic cell surface require binding to the β2 -microglobulin or peptide in order
molecules that play a central role in the induction and reg- to be expressed on the cell surface. So far, MIC expression has
ulation of immune responses, and as such they are involved been detected on freshly isolated endothelial cells, fibroblasts,
in self/non-self-recognition, tolerance, rejection of allografts, keratinocytes, and monocytes. These genes have also been
272 Molecular Hematology

found to be expressed on intestinal epithelial cells as a result presented by class I molecules, and the HLA DMA, DMB,
of stress, and on a variety of tumors of epithelial origin. DOA, and DOB genes, which participate in the loading of
HFE is another non-classical class I gene, located 4 Mb peptides in the HLA class II molecules.
telomeric of HLA-A. This gene has been found to be asso- The HLA class II genes (DR, DQ, and DP) are consti-
ciated with the development of hereditary hemochromatosis tutively expressed on B lymphocytes, monocytes, and den-
(HH). A single point mutation, 845A, replacing cysteine dritic cells, and on activated T lymphocytes and granulocytes
with tyrosine at position 282 (C282Y) is found in over (Table 20.1). HLA class II expression can also be induced
90% of HH patients in the UK. The other two mutations, on non-hematopoietic cells such as fibroblasts and endothe-
replacing histidine by aspartate at amino acid position 63 lial cells, as the result of activation or by the effect of cer-
(H63D) and serine by cysteine at amino acid 65 (S65C), tain inflammatory cytokines, such as interferon (IFN)-γ and
appear to be associated with milder forms of HH. This TNF-α.
gene does not have a direct immune function, as it has lost
the ability to bind antigenic peptides due to closure of the
Function
antigen-binding groove. However, since HFE can bind to the
transferrin receptor, and in this way regulate iron uptake and The HLA molecules play a pivotal role in the induction and
availability, it is possible that HFE may indirectly be involved regulation of the immune response. Both the phenomenon of
in the regulation of immune responses. MHC restriction and the development of tolerance, learned
as T cells pass through the thymus, result in the selection of
a T-cell repertoire that will form the basis of an individual’s
HLA class II genes
capacity to respond to antigens. HLA class I molecules are
The HLA class II genes DR, DQ, and DP are all located within primarily but not exclusively involved in the presentation of
the HLA class II region. There is one non-polymorphic DRA endogenous antigens, such as viral peptides, to CD8+ cyto-
and nine highly polymorphic DRB genes, of which DRB2, B6, toxic T cells, whereas HLA class II molecules present primar-
and B9 are pseudogenes. These genes code for heterodimers ily, but not exclusively, exogenous, such as bacterial, antigenic
formed by an α and a β chain, both encoded by genes within peptides to CD4+ -helper T cells. These cells, once activated,
the MHC. The extracellular portion of these molecules has can initiate and regulate a variety of processes leading to the
two domains (α1 and α2 and β1 and β2 ) encoded by exon maturation and differentiation of cellular and humoral effec-
2 and 3 of each gene, respectively. The α1 and β1 domains tor cells, including the secretion of cytokines. The presenta-
form the peptide-binding groove (see Figure 20.1b). tion of antigenic peptides is a highly regulated process and
The number of DRB genes expressed in each haplotype requires fine interaction between the antigenic peptide, the
varies depending on the DRB1 allele expressed; for example, antigen-binding groove of the HLA molecules, and the T-
HLA DRB1*01, DRB1*01:03, DRB1*08, and DRB1*10 hap- cell receptor. Allelic variation of the HLA molecules can pro-
lotypes only express the DRB1 gene. DRB1*15 and DRB1*16 foundly affect the ability to present certain peptides because
haplotypes additionally express the DRB5 gene, which codes of the presence or absence of critical contact residues in the
for the DR51 product. The HLA DRB1*03:01 (DR17), peptide-binding groove.
DRB1*03:02 (DR18), DRB1*11, DRB1*12, DRB1*13, and HLA molecules on donor cells loaded with donor-derived
DRB1*14 haplotypes also express the DRB3 genes, which peptides can also be recognized directly by T cells of the
code for the DR52 specificity, while the HLA DRB1*04, host by a mechanism called allorecognition. Two pathways
DRB1*07, and DRB1*09 alleles also express the DRB4 gene, of allorecognition, direct and indirect, have been identified,
which encodes the DR53 product. There are a few exceptions both of which lead to the strong alloimmunization seen in
to this gene distribution; for example, a DRB5 gene has been patients receiving blood transfusions or a solid organ or bone
found linked to a DRB1*01 haplotype, and null DRB5 and marrow/stem cell transplantation.
DRB4 genes have been identified. More recently, it has been shown that both classical and
In contrast, there are two DQA and three DQB genes; of non-classical HLA class I molecules interact with two func-
these, only A1 and B1 are expressed and both are polymor- tionally distinct types of receptors, inhibitory and activat-
phic. Similarly, there are two DPA and two DPB genes, of ing, present on natural killer (NK) cells. These receptors
which only DPA1 and DPB1 are expressed and are polymor- belong to two families, the immunoglobulin superfamily, also
phic. More than 23 000 HLA alleles had been named by June called killer immunoglobulin receptors (KIRs), and the C-
2019 (http://hla.alleles.org/nomenclature/index.html). type lectin superfamily CD94, which can covalently assemble
Other HLA-related genes located within the MHC class with several members of the NKG2 family. The KIRs inter-
II region include LMP2, LMP7, TAP1, and TAP2, which act with products of the HLA-A, -B, -C, and -G loci, whereas
are involved in the transport and processing of peptides CD94–NKG2 recognize the non-classical HLA-E molecule
The molecular basis of blood cell alloantigens 273

presenting peptides derived from several HLA class I, A, triplet model, but also include amino acids from discontinu-
B, or C alleles and from HLA-G. MIC-A and MIC-B gene ous regions of the sequence brought together by the folding
products are recognized by receptors present on both NK and of the molecule. This is therefore considered to be a more
γδ T cells. accurate representation of the epitope than that derived from
Thus, HLA molecules have become increasingly relevant the triplet HLAMatchmaker model developed by Duques-
in a variety of clinical situations, such as susceptibility to cer- noy http://www.epitopes.net/. Consequently, it is possible to
tain autoimmune and infectious diseases and in solid organ determine the number of epitopes which are either shared
and stem cell transplantation and blood cell alloimmuniza- or differ between the donor and the recipient. The algorithm
tion. With regard to the latter, two examples will be discussed: also performs intra- and interlocus comparisons of poly-
refractoriness for prophylactic platelet transfusion by HLA morphic eplets to determine the spectrum of non-shared
alloimmunization, and HLA class II restriction of the forma- eplets between HLA antigens of the donor and the patient.
tion of anti-HPA-1a antibodies (see later).

Prophylactic platelet transfusions Hemolytic disease of the fetus and


Prophylactic platelet transfusions are essential for preventing newborn
bleeding during intensive chemotherapy or other myeloab-
lative therapies. Increments in the platelet count after the Rh system
infusion of an adult dose of donor platelets (>250 × 109 l−1 )
are frequently disappointing. Non-immune factors, such The Rh system is the most complex red cell blood group
as splenomegaly, bleeding, sepsis, fever, and certain drugs system, and the most clinically important after ABO. While
(e.g. amphotericin), can compromise the beneficial effect ABO antigens are carbohydrate in nature, with a wide tissue
of donor platelet infusions. In 10–20% of patients the distribution, the Rh antigens are protein based and specific
problem of poor increments is further compounded by to red cells. Rh antigens are carried on two homologous
antibody-mediated destruction of donor platelets. Despite non-glycosylated proteins: RhD, carrying the D antigen, and
this, the clinical definition of refractoriness remains much RhCE, carrying the C, c, E, and e antigens, encoded by RHD
disputed; clinically, the picture of an increased frequency of and RHCE genes, respectively.
platelet transfusions to maintain satisfactory platelet counts
and effective hemostasis requires further laboratory investi-
RH genes
gations for HLA class I and for HPA alloantibodies, platelet
autoantibodies, or high-titer anti-A or anti-B antibodies. The RHD and RHCE genes are located on chromosome 1, and
The ability to type donors and patients for HLA class I A are tightly linked but have opposite orientations. Each gene
and B and HPA by molecular techniques using genomic DNA comprises 10 exons and each encodes a 417 amino acid non-
(see Molecular Typing Techniques; The Molecular Basis of glycosylated protein (although the N-terminal methionine is
Platelet-Specific Antigens or HPA) has resulted in more accu- cleaved from the mature protein). The RHD and RHCE genes
rate matching of donors with patients, with improved platelet are highly homologous (93.8% over all introns and coding
recovery. The algorithm currently used for the management regions) and the encoded proteins differ only by between
of the alloimmunized patient with poor increments is shown 32 and 35 amino acids (Table 20.2). The RhD and RhCE
in Figure 20.3. Traditionally, HLA matching for the provi- proteins are both highly hydrophobic and are predicted to
sion of compatible platelets for patients who have become span the red cell membrane 12 times, with 6 extracellular
immunologically refractory to random platelet transfusion loops (Figure 20.4). The function of the Rh proteins remains
has been based on the serological definition of these antigens unknown.
at the specificity level. More recently, a new approach based In D-negative individuals (around 15% of the Caucasian
on the identification of conformational epitopes present in population) the entire RhD protein is missing from the red
each HLA allele has been described. According to this strat- cell membrane, usually arising from homozygosity for a com-
egy, each HLA antigen is converted into a string of potentially plete deletion of the RHD gene. The pairs of antithetical anti-
immunogenic epitopes, which are represented by amino acid gens C/c and E/e are encoded by polymorphisms within the
triplets (called eplets) on exposed parts of the HLA chains RHCE gene. RhC and Rhc proteins differ by four amino acids,
accessible to alloantibodies. These eplet epitopes are amino of which Ser103Pro, located on the second extracellular loop,
acid residues located within small clusters (with diameters appears to be the most critical. RhE/e antigens are defined
of about 0.3–0.35 nm) around a non-self-residue and are by a single amino acid substitution, Pro226Ala, in the fourth
formed by amino acids in linear sequences as described in the extracellular loop of RhCE (Table 20.2 and Figure 20.4).
274 Molecular Hematology

Patients likely to receive multiple platelet transfusion

Assess transfusion response

Poor response to random donor platelets on 2 or more occasions

HLA type
and test for
HLA-specific
antibodies

HLA antibody
test result

Ab positive
Provide HLA- Ab negative
selected platelets

Good response to Poor response to


Factors associated with non-
HLA-selected HLA-selected
immune platelet destruction
platelets platelets

Provide ABO
Continue Absent Present
compatible, “A”
transfusing HLA-
grade matches if
selected platelets
possible
Consider trial of Treat cause
HLA-selected Consider further platelet
Test for HLA platelets transfusion based on clinical
Test for HPA-
antibodies at status of the patient, e.g.
specific antibodies
regular intervals increase dose of platelets or
discontinue prophylactic
Poor response Good response transfusions

HPA antibody
test result Continue
transfusing HLA-
selected platelets
Positive Negative

Provide HLA- and Consider


HPA-selected 1. Non-immune
platelets consumption
2. ABO antibodies
Fig. 20.3 Platelet transfusions in alloimmunized patients. An algorithm outlining the decision process for the management of alloimmunized
patients refractory for random donor platelets. After confirmation of refractoriness for random donor platelets, patients are screened for human
leukocyte antigen (HLA) class I alloantibodies and, if positive, HLA class I-matched platelets are transfused. In 20–30% of patients, increments with
HLA class I-matched platelets are poor, and screening for human platelet antigen (HPA) antibodies should follow. Also, the possible presence of
potent anti-A or anti-B should be excluded, since platelets do carry ABO blood group antigens. If there are no detectable HLA class I antibodies, a
trial of HLA-matched platelets and screening for HPA antibodies should be considered (right arm of algorithm).
The molecular basis of blood cell alloantigens 275

Table 20.2 Amino acid sequence of the RhD and RhCE proteins

RhCE (C) MSSKYPRSVR RCLPLCALTL EAALILLFYF FTHYDASLED QKGLVASYQV GQDLTVMAAI


RhCE (c) ---------- -----W---- ---------- ---------- ---------- ---------L
RhD ---------- -----W---- ---------- ---------- ---------- ---------I
RhCE (C) GLGFLTSSFR RHSWSSVAFN LFMLALGVQW AILLDGFLSQ FPSGKVVITL FSIRLATMSA
RhCE (c) -------N-- ---------- ---------- ---------- --P------- ----------
RhD -------S-- ---------- ---------- ---------- --S------- ----------
RhCE (C) MSVLISAGAV LGKVNLAQLV VMVLVEVTAL GTLRMVISNI FNTDYHMNLR HFYVFAAYFG
RhD L-----VD-- ---------- ---------- -N-------- --------MM -I--------
RhCE (E) LTVAWCLPKP LPKGTEDNDQ RATIPSLSAM LGALFLWMFW PSVNSPLLRS PIQRKNAMFN
RhCE (e) ---------- ---------- ---------- ---------- -----A---- ----------
RhD -S-------- --E----K-- T--------- ---------- --F--A---- --E----V--
RhCE TYYALAVSVV TAISGSSLAH PQRKISMTYV HSAVLAGGVA VGTSCHLIPS PWLAMVLGLV
RhD ----V----- ---------- --G---K--- ---------- ---------- ----------
RhCE AGLISIGGAK CLPVCCNRVL GIHHISVMHS IFSLLGLLGE ITYIVLLVLH TVWNGNGMIG
RhD -----V---- Y--G------ --P-S-I-GY N--------- -I-------D --GA------
RhCE FQVLLSIGEL SLAIVIALTS GLLTGLLLNL KIWKAPHVAK YFDDQVFWKF PHLAVGF
RhD ---------- ---------- ---------- -------E-- ---------- -------

Amino acid sequences (single letter code) showing differences between RhD and RhCE proteins, in addition to C/c (boxed) and E/e (shaded)
polymorphisms. Sequence identity is indicated by dashes.

S103P (C/c) P226A (E/e)

NH2
49 112
162 358

Rh CcEe 211
protein 384
267
313

408
49 112
162 358

Rh D 211
protein 384
267
313 408 COOH
417
NH2

Rh D
mRNA 1 2 3 4 5 6 7 8 9 10
4.5 1 2 3 4 5 6
Thr32 Ser99 Phe161 Ser230 Ile288 Ala354

–4.5
Fig. 20.4 RhD and RhCE proteins. Schematic representation of the topography of the RhCE (upper figure) and RhD (lower figure) proteins. Both
30 kDa non-glycosylated proteins traverse the red cell membrane 12 times. The difference between RhD and RhCE is defined by 36 of the 417
amino acids (colored circles indicate RhD-specific residues; shaded circles indicate C/c and E/e polymorphisms). The rectangular boxes (1–10)
indicate the positions of exon–exon boundaries in the RhD mRNA, and a hydropathy plot of the RhD protein is shown at the bottom.
276 Molecular Hematology

Immunogenicity of RhD and prevention of 21.5 Kb, encoding a 732 amino acid protein homologous to
immunization a family of zinc-binding endopeptidases, including CD10. K
and k antigens differ by a single amino acid in the Kell gly-
The complete absence of the RhD protein in D-negative
coprotein, Met193Thr, encoded by a single nucleotide sub-
individuals explains its high immunogenicity, and anti-D
stitution in exon 6 of KEL. It is of interest to note that despite
can cause severe hemolytic transfusion reactions (HTRs) and
this apparently minor difference between the two forms, K is
hemolytic disease of the fetus and newborn (HDFN). Pro-
one of the most immunogenic blood group antigens. This is
duction of alloanti-D, stimulated by a D-positive fetus carried
possibly explained by the presence of an N-glycosylation site
by a D-negative mother, may result in severe HDFN, associ-
at Asn191 in k, which is absent in the K protein.
ated with hydrops, kernicterus, and possible fetal death. It has
been shown that this maternal anti-D production can be pre-
Immunogenicity of K and the mechanism
vented by the administration of IgG anti-D. The introduction
of fetal anemia
of anti-D prophylaxis for D-negative women after delivery,
or following sensitizing events during pregnancy, resulted in Anti-K can cause both severe HTR and HDFN, and for
a steep decline in HDFN-related mortality. Before 1969, 46 this reason it is recommended that girls and women of
deaths per 100 000 births were attributed to RhD alloimmu- childbearing age be transfused only with K-negative red
nization, falling to 1.6/100 000 births in 1990. More recently, cells. K antigen is present on red cells of approximately 9% of
the introduction of routine antenatal anti-D prophylaxis white people and 2% of black people. In HDFN caused by K
(RAADP) for all D-negative pregnant women in the UK alloimmunization, fetal anemia appears to result mainly from
has further reduced alloimmunization rates. antibody-mediated inhibition of erythropoiesis. This sug-
Knowledge of the molecular basis of blood group anti- gests a possible role for the Kell glycoprotein in the regulation
gens, together with the discovery of cell-free fetal DNA in and growth of erythroid progenitor cells. It is more difficult
the maternal circulation, has allowed the development of to predict the severity of anti-K HDFN than that caused by
non-invasive fetal blood group genotyping from maternal anti-D, as there is little correlation between anti-K levels
blood. Determination of fetal RHD genotype in pregnancies and disease severity. Fetal genotyping to predict Kell antigen
at risk of HDFN allows appropriate monitoring and clinical status in at-risk pregnancies allows close monitoring, and
intervention to be directed at pregnancies where the fetus is treatment where necessary, of those where the fetus is shown
predicted to be D-positive. In the UK, approximately 38% of to be K-positive.
D-negative pregnant women will carry a D-negative fetus and
are therefore not at risk of RhD alloimmunization. Recently, Other red cell antigens and molecular
large-scale non-invasive fetal RHD screening has been intro- typing
duced into the UK for all non-sensitized D-negative pregnant
The molecular bases of the majority of blood group alloanti-
women, to determine the individual requirement for anti-D
gens have now been elucidated, and the genes encoding them
prophylaxis. Targeting of RAADP to those women confirmed
cloned and sequenced. This allows the use of molecular typ-
to be carrying a D-positive fetus prevents unnecessary expo-
ing for the prediction of blood group phenotypes, and such
sure to anti-D in the remainder, reducing the use of anti-D
genotyping is routinely used for the most clinically relevant
and resulting in significant cost savings.
blood group systems (Rh, Kell, Kidd, Duffy, and MNS).
In addition to anti-D, antibodies to other Rh antigens are
Molecular typing is of particular use when serological typing
also clinically significant, and associated with both HTR and
is not possible, such as for multitransfused patients (e.g.
HDFN. Anti-c is the most clinically important after anti-D
those with hemoglobinopathies) and in prediction of fetal
and has been associated with severe HDFN, whereas anti-C,
blood group phenotype by non-invasive methods. Accurate
anti-E, and anti-e have rarely been implicated in severe
interpretation of blood group genotype into predicted phe-
HDFN. Non-invasive fetal genotyping to predict RhC/c and
notype requires in-depth knowledge of the genes involved
E/e antigen status is useful in the management of at-risk preg-
and their many allelic variants, which may be of particular
nancies, although no prophylaxis is available.
relevance in certain ethnic groups. Homology between RHD
and RHCE genes in the Rh system (also relevant to the
homologous genes of the MNS system) and a high degree of
Kell
polymorphism in these genes lead to challenges in designing
The K antigen of the Kell blood group system is the most clin- and correctly interpreting genotyping tests. However, geno-
ically important red cell antigen outside of the ABO and Rh typing offers a highly accurate method for the prediction of
systems. The antithetical antigens K and k are carried on Kell blood group phenotype, and is invaluable in situations where
glycoprotein (CD238), a 93 kDa type II transmembrane gly- serological-based typing is not possible. Fetal genotyping for
coprotein. The KEL gene consists of 19 exons, spanning about clinically significant blood group antigens allows improved
The molecular basis of blood cell alloantigens 277

clinical care of alloimmunized women and pregnancies at can be facilitated by the use of non-invasive fetal geno-
risk of HDFN. typing to identify antigen-positive fetuses being carried by
antigen-negative mothers. Previous blood transfusion is the
most important risk factor for non-D immunization, partic-
Pathology of HDFN
ularly in the case of K immunization. For this reason, girls
Maternal alloantibodies can be formed against a blood group and women of childbearing age are transfused only with
alloantigen present on fetal red cells but absent from mater- K-negative, RhD-matched blood. Further matching of Rh
nal cells. The main antigens implicated in HDFN are RhD, antigens (in particular c and E) in this patient group may have
K, and Rhc. Red cell alloantibodies of the IgG class (mostly the potential to reduce alloimmunization, and consequently
IgG1 and IgG3) can cross the placenta, bind to fetal red cells, the risk of subsequent HDFN.
and shorten their survival, giving rise to hyperbilirubinemia
and anemia. In K immunization, hyperbilirubinemia is a less
reliable parameter for the prediction of the severity of fetal Neonatal alloimmune
anemia, and these pregnancies can be complicated by early thrombocytopenia
fetal loss. In England and Wales, approximately 500 fetuses
develop HDFN annually, with 25–30 perinatal deaths, and
HPA alloantigen systems
an estimated further 20 spontaneous abortions prior to
28 weeks, attributed to HDFN. The HPA alloantigens are expressed on the surface of
platelets, but are not truly platelet-specific since some are also
found on the surface of other blood cell types (see Table 20.1).
Monitoring and treatment of HDFN
Alloimmunization against HPA is associated with three clin-
Screening for clinically significant red cell alloantibodies in ical syndromes:
pregnant women is the standard of care in most European 1 neonatal/fetal alloimmune thrombocytopenia (NAITP);
countries. Of all pregnant women, 1–2% screen positive 2 post-transfusion purpura;
for red cell alloantibodies, although in the vast majority 3 refractoriness for platelet transfusions.
of cases the antibodies are not considered to be of clinical NAITP was first described by van Loghem in 1959 and was
significance. Determination of antibody specificity, and mea- initially thought to be a rare disorder. Prospective screening
surement of antibody levels, allows assessment of the risk of studies in pregnant (Caucasian) women have shown that 1 in
fetal anemia in order to identify cases of possible severe dis- 1100 neonates have severe thrombocytopenia (<50 × 109 l−1 )
ease early in pregnancy, reducing the associated morbidity or due to maternal anti-HPA-1a, confirming the notion that
mortality. Once a fetus is assessed to be at moderate to severe the most frequent cause of severe thrombocytopenia in the
risk of developing fetal anemia, regular ultrasound exami- term newborn is maternal alloantibodies against a fetal HPA
nation and serial Doppler measurement of middle cerebral alloantigen.
artery peak systolic velocity (MCA PSV) are required. This serious clinical condition is caused by the destruction
Fetal anemia can be treated antenatally by intrauterine of fetal/neonatal platelets by maternal HPA alloantibodies of
transfusion (IUT), significantly reducing perinatal mortality, the IgG class. Cerebral bleeds in the perinatal period are the
with reported fetal survival rates of greater than 90% in IUT- most concerning complication, which either occur in utero
treated cases. In neonates, phototherapy has been shown or during delivery. In cases of severe thrombocytopenia
to effectively reduce the need for exchange transfusion in (<20 × 109 l−1 ), there remains a small but definite risk of
the treatment of hyperbilirubinemia, in order to prevent this serious complication in the first days of life, warranting
kernicterus. However, severe neonatal hemolytic anemia and treatment. For proper clinical management, the cause of
hyperbilirubinemia may be treated by exchange transfusion severe thrombocytopenia in an otherwise healthy term
with compatible donor red cells, correcting anemia and neonate should be determined with urgency, and prompt
removing both bilirubin and maternal alloantibodies from correction of a count less than 20 × 109 l−1 by platelet
the circulation. transfusion is of the utmost importance. This should precede
the outcome of platelet antibody investigation, which can be
a time-consuming process.
Prevention of HDFN
Before the introduction of anti-D prophylaxis, most HDFN
The molecular basis of platelet-specific
cases were caused by RhD immunization. The routine RhD
antigens or HPA
testing of all pregnant women, combined with RAADP for
D-negative pregnant women, has been extremely successful To date, 35 HPAs have been defined by immune sera, which
in lowering HDFN-associated morbidity and mortality. This are grouped into 29 platelet-specific alloantigen systems. The
278 Molecular Hematology

Table 20.3 Platelet-specific alloantigen systems

System Antigen Alternative names Glycoprotein Nucleotide change Amino acid change

HPA-1 HPA-1a Zwa , PlA1 GPIIIa 176T Leu33


HPA-1b Zwb , PlA2 176C Pro33
HPA-2 HPA-2a Kob GPIbα 482C Thr145
HPA-2b Koa , Siba 482C Met145
HPA-3 HPA-3a Baka , Leka GPIIb 2621T Ile843
HPA-3b Bakb 2621G Ser843
HPA-4 HPA-4a Yukb , Pena GPIIIa 506G Arg143
HPA-4b Yuka , Penb 506A Gln143
HPA-5 HPA-5a Brb , Zavb GPIa 1600G Glu505
HPA-5b Bra , Zava 1600A Lys505
HPA-6bw Caa , Tua GPIIIa 1544G>A Arg>Gln489
HPA-7bw Moa GPIIIa 1297C>G Pro>Ala407
HPA-8bw Sra GPIIIa 1984T>C Arg>Cys636
HPA-9bw Maxa GPIIb 2602G>A Val>Met837
HPA-10bw Laa GPIIIa 263G>A Arg>Glu62
HPA-11bw Groa GPIIIa 1976G>A Arg>His633
HPA-12bw Iya GPIbβ 119G>A Gly>Glu15
HPA-13bw Sita GPIa 2483C>T Thr>Met799
HPA-14bw Oea GPIIIa 1109-1911delAAG Lys611del
HPA-15 HPA-15a Govb CD109 2108C Ser682
HPA-15b Gova 2108A Tyr682
HPA-16bw Duva GPIIIa 497C>T Thr>Ile140
HPA-17bw Vaa GPIIb/IIIa 662C>T Thr>Met195
HPA-18bw Caba GPIa 2235G>T Gln>His716
HPA-19bw Sta GPIIIa 487A>C Lys>Gln137
HPA-20bw Kno GPIIb 1947C>T Thr>Met619
HPA-21bw Nos GPIIIa 1960G>A Glu>Lys628
HPA-22bw Sey GPIIb 584A>C Lys>Thr164
HPA-23bw Hug GPIIIa 1942C>T Arg>Trp622
HPA-24bw Cab2a+ GPIIb 1508G>A Ser>Asn472
HPA-25bw Swia GPIa 3347C>T Thr>Met1087
HPA-26bw Seca GPIIIa 1818G>T Lys>Asn580
HPA-27bw Cab3a+ GPIIb 2614C>A Leu>Met841
HPA-28bw War GPIIb 2311G>T Val>Leu740
HPA-29bw Khab GPIIIa 98C>T Thr>Met7

HPA, human platelet antigen.

molecular basis of these have been determined and are all and patients, even when the latter are thrombocytopenic.
mapped to certain platelet membrane glycoproteins (GPs). High-throughput donor HPA typing can also be performed
Twelve of the most clinically relevant HPAs are grouped into using the 5′ -nuclease (TaqMan®) assay.
six biallelic systems. Of the 29 HPA systems, 22 are on the
integrin heterodimer αIIbβ3 or glycoprotein (GP)IIb/IIIa
(Table 20.3 and Figure 20.5); of the remaining seven, two
Immunogenicity and immune
are on GPIb–IX–V, four on the integrin α2β1 or GPIa/IIa,
response restriction
and one on CD109. The difference between the two alle- As the difference between self and non-self is defined by a
les is a single-nucleotide substitution in the gene encod- single amino acid substitution, the immunogenicity of the
ing the relevant glycoprotein, with the exception of HPA- HPA alloantigens is relatively low when compared with that
14bw (Table 20.3). Amplification of genomic DNA by PCR- of some of the other blood cell alloantigens (e.g. RhD and
SSP (Figure 20.6) and SBT can be used to type donors HLA class I). The two most clinically relevant HPA antigens
The molecular basis of blood cell alloantigens 279

GPIIa GPIIa GPVI GPIIb GPIIa CD109 GPIbα GPV


α2 β2 αIIB β3 HPA-19w
K137Q HPA-2
T145M
HPA-5 HPA-16w
E505K T140l
HPA-17w
HPA-18w T195M
TQ716H HPA-4
HPA-13w R143Q
HPA-10w HPA-7w
T799M R62Q HPA-15
P407A S703Y
HPA-20w HPA-1
1 T619M 1 GPIX
L33P
HPA-9w HPA-6w
2 V837M 2
R489Q
3 HPA-3 3 HPA-8w
18435 s R636C GPIbβ
4 4 s
s HPA-11w HPA-12w s
βTD βTD R633H G15Q
HPA-14w
Del K611
HPA-21w
E628K

Talin
Fig. 20.5 Schematic presentation of the platelet glycoproteins. Schematic presentation of the platelet glycoprotein from which the human
platelet antigens (HPAs) can be found. The positions of the amino acid substitutions arising from allelic variation of the various glycoprotein genes
are marked by black arrows and the name of the HPA system is noted. Amino acids are described with one letter abbreviations.
Source: Adapted from Lucas G. (2009). Practical Transfusion Medicine, 3rd edn. Chichester: Wiley-Blackwell. Reproduced by permission of John
Wiley & Sons.

are HPA-1a and HPA-5b on the β3 and α2 integrins, namely magnitudes higher than that of its antithetical antigen HPA-
those on platelet glycoproteins GPIIIa and GPIa, respectively. 1b (Pro33) was initially not well understood. In the early
Alloantibodies against other HPA alloantigens are observed 1980s, it was discovered that the formation of anti-HPA-1a in
infrequently in pregnancy, but do occur, albeit at a low fre- pregnancy was positively associated with the HLA haplotype
quency, in hemato-oncological patients and other patients on A*01-B*08-DRB1*03. Further studies revealed that nearly
long-term prophylactic platelet transfusion. all antibody formers were positive for the *01:01 allele of the
The HPA-1a and -1b alloantigens are based on a C → G DRB3 gene (DRB3*01:01 or DR52a; see HLA Class II Genes;
mutation in the GPIIIa gene, resulting in a Leu33Pro Figure 20.7). A prospective study in 25 000 pregnant women
mutation. Why the immunogenicity of HPA-1a (Leu33) is showed that this class II marker has an odds ratio of 140,
which makes it one of the most reliable HLA associations
reported to date, with negative predictive power equal to
that of HLA-B27 in ankylosing spondylitis. A difference in
1a 1b 2a 2b 3a 3b 4a 4b 5a 5b
the efficiency of presentation of the GPIIIa-Leu33 (HPA-
1a)-derived oligopeptide between DRB3*01:01-positive and
-negative antigen-presenting cells to CD4+ T cells is the most
likely explanation of this restriction in alloimmunization.
The frequency of the HLA DRB3*01:01 allele in Caucasians is
33%, and this marker therefore has a high negative predictive
Fig. 20.6 Polymerase chain reaction–sequence-specific primers
value, but a low positive one for anti-HPA-1a formation.
(PCR-SSP) for human platelet antigen (HPA) alleles. Results of
agarose gel electrophoresis of PCR products obtained by amplification
of segments of the GPIIIa, GPIb𝛼, GPIIb, and GPIa genes using Allele frequencies
allele-specific primers for the “a” and “b” alleles of the HPA-1, HPA-2,
HPA-3, HPA-4, and HPA-5 systems. The results for this donor are In Caucasians, the allele frequency for most HPA systems
HPA-1b1b, 2a2a, 3a3b, 4a4a, and 5a5a. Products of the allele-specific is skewed toward the “a” allele. The allele frequencies vary
amplification are the lower bands. In all reactions, a set of control between populations; for example, GPIIIa-Pro33 (HPA-1b)
primers has been included to produce an amplicon (upper band) is extremely rare or absent in East Asia, while the opposite is
derived from the human growth hormone gene. the case for the GPIIIa-Gln143 form (HPA-4b).
280 Molecular Hematology

Fig. 20.7 Structure of human leukocyte


antigen (HLA) DRB3*01:01 molecule. A view of
CHO
the peptide-binding site of the HLA class II
DRB3*01:01 molecule, as seen by the T-cell
receptor of a T lymphocyte. Two α helices (see
Figure 20.1b) are resting on a base of
antiparallel-running β-pleated sheets. The
antigen-derived peptide is not shown, but in our
example of immune response restriction, an
oligopeptide derived from fetal GPIIIa-Leu33 will
be juxtaposed between the two helices. The
presence of this peptide defines the difference
N between self and non-self and triggers the
proliferation of GPIIIa-Leu33 (HPA-1a)
alloantigen-specific helper T cells.

Incidence of NAITP alloantibodies against a paternally inherited platelet alloanti-


gen present on the fetal platelets but absent from the mater-
Alloantibodies against the HPA-1a (PlA1 , Zwa ) alloanti-
nal ones. In cases where the partner is homozygous for the
gen occur in 1 in 365 pregnancies and cause severe
relevant alloantigen, there is a 100% chance that his future
thrombocytopenia, with a neonatal platelet count of less than
offspring will be at risk, whereas in the case of heterozygosity
50 × 109 l−1 in 1 in 1100 term neonates.
50% will be affected.

Pathology Treatment
Maternal IgG alloantibodies against a fetal HPA alloantigen A neonatal platelet count of less than 20 × 109 l−1 should be
can cross the placenta and bind to the fetal platelets, thus corrected immediately, preferably with HPA-1a- and HPA-
reducing platelet survival. The HPA-1 and HPA-5 systems are 5b-negative donor platelets, as these will be compatible with
the two most clinically relevant, with the majority of cases the maternal HPA alloantibody in over 95% of cases. In cases
caused by antibodies against HPA-1a (80%) and HPA-5b of absence of HPA-compatible platelets, initial transfusion of
(10–15%). random ABO/RhD-compatible platelets should be consid-
ered, followed by the transfusion of compatible ones if the
Characteristics platelet count decreases further. In a typical case, the platelet
count should recover to normal within a week, although a
Neonatal alloimmune thrombocytopenia presents in the oth- more protracted recovery can occur.
erwise healthy newborn as petechiae or ecchymoses, or is
found coincidentally by a whole blood count. Severe cases Counseling
can present neurological symptoms due to cerebral bleeds or
with hydrops fetalis or cerebral cysts. NAITP can affect the Counseling of couples with an index case about the risks
first pregnancy and has a 10% risk of severe intracranial hem- of severe fetal/neonatal thrombocytopenia in a subsequent
orrhage. Diagnosis is based on the detection of HPA alloanti- pregnancy needs to be based on the severity of disease in the
bodies in the maternal serum combined with a parental HPA index case and the outcome of immunological investigations.
incompatibility, as determined by molecular typing meth- The following should be considered:
r thrombocytopenia in subsequent cases is as severe or
ods such as PCR-SSP, TaqMan® assay, or sequencing methods
such as SBT or NGS. generally more severe.
r antibody specificity and titer have some correlation with

severity; for example, HPA-5b antibodies generally cause


Inheritance
mild thrombocytopenia, which rarely results in a cerebral
All systems described to date are biallelic with codominant bleed. The latter is generally associated with HPA-1a anti-
expression of both alleles. Homozygous women can produce bodies.
The molecular basis of blood cell alloantigens 281

Table 20.4 Human neutrophil antigens (HNA)

Antigen system Alleles Antigens Former name Gene CD

HNA-1 FCGR3B*01 HNA-1a NA1 FCGR3B CD16


FCGR3B*02 HNA-1b, HNA-1d NA2 (HNA-1b)
FCGR3B*03 HNA-1c, HNA-1b SH (HNA-1c)
FCGR3B*04 HNA-1a
FCGR3B*05 HNA-1b var

HNA-2* n/a HNA-2 NB1 CD177 CD177


HNA-3 SLC44A2*01 HNA-3a 5b SLC44A2 Not known
SLC44A2*02 HNA-3b
SLC44A2*03 HNA-3a var
HNA-4 ITGAM*01 HNA-4a Mart ITGAM CD11b
ITGAM*02 HNA-4b
HNA-5 ITGAL*01 HNA-5a Ond ITGAL CD11a
ITGAL*02 HNA-5b

*HNA-2 is defined by isoantibodies.

r a high-titer HPA antibody is more likely to be associated FCGR3B*01, FCGR3B*02, FCGR3B*03, FCGR3B*04 and
with severe thrombocytopenia, but cerebral bleeds also FCGR3B*05. Five amino acid substitutions at positions 36,
occur with low titers. 65, 78, 82 and 106 determine the four HNA-1 antigens
(Table 20.4). A single amino acid substitution at position
78 of FCGR3B causes the expression of HNA-1c instead of
Neonatal alloimmune neutropenia HNA-1d next to HNA-1b. Thus, individuals homozygous for
the gene encoding HNA-1a lack expression of HNA-1b, 1c,
Maternal alloimmunization against neutrophil-specific and 1d. Due to duplication of the FCGR3B gene, as found
alloantigens on fetal/neonatal neutrophils is rare, although in HNA-1c positive Caucasians, the FCGR3B*01 (HNA-1a)
there are no precise prevalence figures. Clinical presenta- allele and the FCGR3B*03 (HNA-1b and HNA-1c) allele
tion is one of mainly bacterial infection with a selective are expressed from the same chromosome. Individuals that
neutropenia on a whole blood cell count. possess this haplotype together with the FCGR3B*02 allele
(HNA1-b and HNA-1d expression) on the other chromo-
HNA system some will express all four antigens of the HNA-1 system.
The single nucleotide polymorphisms (SNPs) responsible
Neutrophils, like all other cells present in blood, express
for the expression of the different HNA-1 antigens are
polymorphic molecules that can induce strong antibodies
shown in Table 20.5. Individuals genotyped positive for the
when transfused or transplanted or as result of pregnancy.
FCGR3B*01 and FCGR3B*04 alleles will express the HNA-1a
These antigens are called human neutrophil antigens
epitope, while individuals positive for the FCGR3B*05 will
(HNAs). HNA-specific antibodies have been implicated
express the HNA-1bvar epitope for HNA-1b antibodies.
in the development of neonatal alloimmune neutropenia,
Some individuals who do not express the FCGR3B gene and
TRALI, and other alloimmune or autoimmune neutrope-
carry a null phenotype can develop isoantibody of known
nias. So far five HNA antigenic systems have been described
clinical significance in cases of NAITP. The FcγRIIIB null
(HNA-1 to HNA-5), which are the result of polymorphisms,
phenotype is rare and is based on a double deletion of
except for HNA-2.
the FCGR3B gene, and is in some cases associated with a
deletion of the FcγRIIC (FCGR2C) gene. Deficiency for the
HNA-1
most abundant Fc receptor on neutrophils does not seem to
HNA-1 is located on a glycosylphosphatidylinositol (GPI)- be associated with an obvious clinical phenotype. This is in
anchored glycoprotein that forms the low-affinity Fcγ recep- contrast with a mutation in the FcγRIIIA (FCGR3A) gene,
tor. HNA-1 is encoded by the FcγRIIIB (FCGR3B) gene which encodes a Leu48His substitution in the first extra-
located on chromosome 1 and mediates IgG-induced phago- cellular domain of the NA cell FcγRIIIA which, although
cytosis. Five alleles of the HNA-1 system have been described, only described in one infant, was associated with recurrent
282 Molecular Hematology

Table 20.5 HNA-1 antigens and associated alleles, SNP and amino acid positions

Nucleotide at SNP position Amino acid positions


Antigen Allele 108 194 233 244 316 36 65 78 82 106

HNA-1a FCGR3B*01 G A C G G Arg Asn Ala Asp Val


HNA-1b, 1d FCGR3B*02 C G C A A Ser Ser Ala Asn Ile
HNA-1b, 1c FCGR3B*03 C G A A A Ser Ser Asp Asn Ile
HNA-1a FCGR3B*04 G A C G A Arg Asn Ala Asp Ile
HNA-1bvar FCGR3B*05 C G C G A Ser Ser Ala Asp Ile

HNA, human neutrophil antigens; SNP, single nucleotide polymorphism.

and serious respiratory tract viral infection from birth. The weaker affinity. HNA-3a allo-antibodies are implicated in
FcγRIIIB null phenotype can cause immune neutropenia in febrile transfusion reactions, neonatal immune neutropenia,
the newborn due to maternal anti-FcγRIIIB isoantibodies. and severe, often fatal cases of TRALI.
PCR techniques can be used to determine the HNA-1
and FcγRIIIB null genotypes, and panels of HNA-typed HNA-4
granulocytes are useful probes for alloantibody detection.
HNA-1 alloantibodies have also been implicated in cases HNA-4 (previously known as Marta ) is located on the αM
of autoimmune and alloimmune neutropenia, including chain (CD11b) of the β2 integrin MAC-1. HNA-4 is encoded
neonatal alloimmune neutropenia and TRALI. by the ITGAM gene located at chromosome 16p11.2. This
system has two antigens, HNA-4a (ITGAM*01) and HNA-4b
HNA-2
(ITGAM*02), the result of a single amino acid substitution
at position 61. aM/b2-integrin is a transmembrane protein
HNA-2 (previously known as NB1) is expressed on a expressed on the surface of many leukocytes and involved
58–64 kDa glycoprotein (CD177) found on the plasma mem- in adhesion, transmigration, phagocytosis, and cell-mediated
brane and secondary granules of neutrophils. This glycopro- cytotoxity. The difference between the antigens is caused by a
tein is linked to the plasma membrane by a GPI anchor and unique SNP at position 230 of the gene, changing an arginine
is coded for by a gene located on chromosome 19. HNA-2 is into a histidine at protein level.
expressed in approximately 50% of the total neutrophils by
95% of individuals. Some individuals do not express HNA-2 HNA-5
due to a transcription defect in the CD177 gene. The HNA-2
alloantigen typing is based on the use of human immune anti- HNA-5 (previously known as Onda ) is located on the αL inte-
sera and immunofluorescence. HNA-2 antibodies have been grin unit of the leucocyte function-associated antigen (LFA)-
found in patients with alloimmune and autoimmune neu- 1, also known as CD11b. It is encoded by the ITGAL gene
tropenia. which is located at chromosome 16p11.2. aL/b2-integrin is a
leukocyte-specific adhesion molecule involved in leukocyte
HNA-3 interactions and trafficking. The HNA-5a antigen is deter-
mined by a single amino acid substitution at position 766 and
HNA-3 (HNA-3a antigen, previously known as 5b) is car- HNA-5a alloantibodies have been involved in NAIN.
ried on the choline-transporter-like protein 2, which is a
transmembrane protein encoded by the SLC44A2 gene on
Detection of HNA antigens and antibodies
chromosome 19p13.1. There have been two HNAs iden-
tified which are encoded by three alleles (SLC44A02*01, Some HNA antigens are uniquely expressed on neutrophils
SLC44A02*02, SLC44A02*03). These are the result of two (HNA-1 and HNA-2), whereas others are also present on
SNPs at positions 451 and 455, giving rise to amino acid other cells or tissues (HNA-3, HNA-4, and HNA5). With
changes at positions 151 and 152, respectively. A SNP the exception of HNA-2, the polymorphisms that determine
(455G > A) changing an arginine to glutamine at position the HNA genotype of an individual can be detected using
152 of the protein is responsible for the difference between molecular technique including PCR-SSP, PCR-SSOP, and
HNA-3a and HNA-3b. SLC44A02*03 encodes the HNA-3avar DNA sequencing. The detection of HNA-specific antibodies
epitope, which is recognized by HNA-3a antibodies but with however is still difficult, unreliable, and time-consuming and
The molecular basis of blood cell alloantigens 283

can involve the use of a number of tests such as the granu- Creary, L.E., Guerra, S.G., Chong, W. et al. (2019). Next-generation
locyte agglutination test, granulocyte immunofluorescence HLA typing of 382 International Histocompatibility Working Group
test, chemiluminescence test, and the monoclonal antibody reference B-lymphoblastoid cell lines: report from the 17th Interna-
immobilization of granulocyte antigens (MAIGA) test. tional HLA and Immunogenetics workshop. Hum. Immunol. 80: 449–
Indeed, many of these techniques (except for MAIGA) are 460.
Curtis, B.R. and McFarland, J.G. (2014). Human platelet antigens – 2013.
not able to distinguish specific HNA from HLA antibodies.
Vox Sang. 106: 93–102. Review.
There is a need to develop a new generation of techniques Dendrou, C.A., Petersen, J., Rossjohn, J., and Fuggar, L. (2018). HLA
for the detection and characterization of HNA-specific variation and disease. Nat. Rev. Immunol. 18: 325–339.
antibodies. Duquesnoy, R.J., Kamoun, M., Baxter-Lowe, L.A. et al. (2015). A per-
In addition to neonatal neutropenia, neutrophil-specific sonal viewpoint: should HLA mismatch acceptability for sensitized
antibodies are implicated in non-hemolytic febrile transfu- transplant candidates be determined at the high-resolution rather
sion reactions, TRALI, and autoimmune neutropenia. Severe than the antigen level? Am. J. Transplant. 15: 923–930.
but reversible neutropenia in the newborn may require treat- Flesch, B.K. for the International Society of Blood Transfusion (ISBT)
ment with antibiotics to control bacterial infection. So far, HNA Nomenclature Subcommittee. (2015). Human neutrophil anti-
there is minimal or no evidence that the mutations in the gens: a nomenclature update based on new alleles and new antigens.
FcγRIIIB protein have any functional consequences, exem- ISBT Sci. Ser. 10: 243–249. Review.
Flesch, B.K. and Reil, A. (2018). Molecular genetics of the human neu-
plified by the FcγRIIIB null phenotype, which is not linked
trophil antigens. Transfus. Med. Hemother. 45: 300–309. Review.
with an obvious pathological phenotype. Gajjar, K. and Spencer, C. (2009). Diagnosis and management of non-
anti-D red cell antibodies in pregnancy. Obstet. Gynaecol. 11: 89–95.
Guerra, S.G., Chong, W., Brown, C.J., and Navarrete, C.V. (2018). Evalu-
ation of Ion Torrent sequencing technology for rapid clinical human
Conclusions leucocyte antigen typing. Int. J. Immunogenet. 45: 230–235.
de Haas, M., Thurik, F.F., Koelewijn, J.M., and van der Schoot, C.E.
The molecular basis of most blood cell alloantigens, includ- (2015). Haemolytic disease of the fetus and newborn. Vox Sang. 109:
ing those of the HLA system, has been discovered with the 99–113.
use of molecular techniques. This has enabled their reliable Horton, R., Wilming, L., Rand, V. et al. (2004). Gene map of the
definition at high resolution, replacing previous techniques extended human MHC. Nat. Rev. Genet. 5: 889–899.
that were based on the use of polyclonal antibody reagents. Janeway, C. Jr., Travers, P., Walport, M. et al. (2001). Immunology: The
New molecular techniques such as NGS allow allelic resolu- Immune System in Health and Disease, 5e. New York: Garland Science.
tion and the discovery of new alleles. This body of knowledge Le van Kim, C., Mouro, I., Cherif Zahar, B. et al. (1992). Molecu-
lar cloning and primary structure of the human blood group RhD
has already made a significant contribution to current clin-
polypeptide. Proc. Natl. Acad. Sci. U.S.A. 89: 10925–10929.
ical management. On the one hand, first-trimester fetal
Lieberman, L., Greinacher, A., Murphy, M.F. et al. (2019). Fetal
typing for blood cell-specific alloantigens can now be used and neonatal alloimmune thrombocytopenia: recommendations for
in cases of severe maternal alloimmunization to prevent fetal evidence-based practice, an international approach. Br. J. Haematol.
and neonatal morbidity, and improved selection of HLA- 185: 549–562.International Collaboration for Transfusion Medicine
and HPA-matched donor platelets aids the management Guidelines (ICTMG).
of hemato-oncological patients. On the other hand, it is van Loghem, J.J., Dorfmeyer, H., van de Hart, M. et al. (1959). Serolog-
envisaged that bone marrow and solid organ transplants ical and genetical studies on the platelet antigen (zw). Vox Sang. 4:
across apparent mismatches will be able to progress because 161–169.
of the identification of so-called permissive mismatches, Muschter, S., Berthold, T., and Greinacher, A. (2011). Developments in
and better HLA matching will allow accommodation of the the definition and clinical impact of human neutrophil antigens. Curr.
Opin. Hematol. 18: 452–460. Review.
graft at lower levels of immune suppression. Finally, more
Peterson, J.A., McFarland, J.G., Curtis, B.R., and Aster, R.H. (2013).
complete understanding of the molecular rules defining a
Neonatal alloimmune thrombocytopenia: pathogenesis, diagnosis
subject’s immune response status should lead to the identi- and management. Br. J. Haematol. 161: 3–14. Review.
fication of high responders, in order to target these for more Porcelijn, L. and de Haas, M. (2018). Neonatal alloimmune neutropenia.
specific manipulation of their immune system, with the aim Transfus. Med. Hemother. 45: 311–316. Review.
of establishing alloantigen-specific non-responsiveness. Porto, G., Brissot, P., Swinkels, D.W. et al. (2016). EMQN best prac-
tice guidelines for the molecular genetic diagnostic of hereditary
hemochromatosis (HH). Eur. J. Hum. Genet. 4: 479–495.
Qureshi, H., Massey, E., Kirwan, D. et al. (2014). BCSH guideline for
Further reading the use of anti-D immunoglobulin for the prevention of haemolytic
disease of the fetus and newborn. Transfus. Med. 24: 8–20.
Brown, C.J. and Navarrete, C.V. (2011). Clinical relevance of the HLA Reil, A. and Bux, J. (2015). Geno- and phenotyping of human neutrophil
system in blood transfusion. Vox Sang. 101: 93–105. antigens. Methods Mol. Biol. 1310: 193–203.
284 Molecular Hematology

Reil, A., Sachs, U.J., Siahanidou, T. et al. (2013). HNA-1d: a new human Wagner, F.F. and Flegel, W.A. (2004). Review: the molecular basis of the
neutrophil antigen located on Fcγ receptor IIIb associated with Rh blood group phenotypes. Immunohematology 20: 23–36.
neonatal immune neutropenia. Transfusion 53: 2145–2151. Wienzek-Lischka, S., König, I.R., Papenkort, E.M. et al. (2017). HLA-
Rozman, P. (2002). Platelet antigens. The role of human platelet DRB3*01:01 is a predictor of immunization against human platelet
alloantigens (HPA) in blood transfusion and transplantation. Transpl. antigen-1a but not of the severity of fetal and neonatal alloimmune
Immunol. 10: 165–181. Review. thrombocytopenia. Transfusion 57: 533–540.
Soothill, P.W., Finning, K., Latham, T. et al. (2015). Use of cffDNA to Williamson, L.M., Hackett, G., and Rennie, J. (1998). The natural his-
avoid administration of anti-D to pregnant women when the fetus is tory of fetomaternal alloimmunization to the platelet-specific antigen
RhD-negative: implementation in the NHS. BJOG 122: 1682–1686. HPA-1a (PlA1, Zwa) as determined by antenatal screening. Blood 92:
Suzuki, S., Ranade, S., Osaki, K. et al. (2018). Reference grade character- 2280–2287.
ization of polymorphisms in full-length HLA class I and II genes with Winkelhorst, D. and Oepkes, D. (2019). Foetal and neonatal alloim-
short-read sequencing on the ION PGM system and long-reads gen- mune thrombocytopenia. Best Pract. Res. Clin. Obstet. Gynaecol.
erated by single molecule, real-time sequencing on the PacBio plat- Epub ahead of print. Review.
form. Front. Immunol. 9, article 2294. Zdravic, D., Yougbare, I., Vadasz, B. et al. (2016). Fetal and neonatal
Veldhuisen, B., Porcelijn, L., Ellen van der Schoot, C., and Haas, M.D. alloimmune thrombocytopenia. Semin. Fetal Neonatal. Med. 21: 19–
(2014). Molecular typing of human platelet and neutrophil antigens 27. Review.
(HPA and HNA). Transfus. Apher. Sci. 50: 189–199. Review.
Chapter 21 Functions of blood group antigens
Jonathan S. Stamler1 & Marilyn J. Telen2
1 Harrington Discovery Institute and Institute of Transformative Molecular Medicine, University Hospitals Cleveland Medical Center

and Case Western Reserve University, Cleveland OH, USA


2 Department of Medicine, Division of Hematology, Duke University Medical Center, Durham NC, USA

Introduction, 285 Blood group antigen proteins that function as adhesion molecules, 291
Anion-exchanger protein 1 (band 3 protein), 285 Blood group antigen proteins with other functions, 293
Rh protein, 289 Summary, 293
Glycophorins C and D, 290 Further reading, 293

(∼1 million copies/cell), constituting 20–30% of the protein


Introduction mass. Previously referred to as band 3 protein, on the basis
of its original electrophoretic migration profile, AE1 is a
Erythrocyte blood group antigens are, by definition, poly-
95–102 kDa variably glycosylated protein constructed of
morphic epitopes of red cell surface structures that are rec-
911 amino acids, yielding three distinct structural and
ognizable by antibodies. These epitopes generally include rel-
functional domains: the N-terminal 45-kDa cytoplasmic
atively small changes, such as the substitution of one amino
domain, the 55-kDa C-terminal hydrophobic transmem-
acid, in molecules that are largely the same from one indi-
brane domain, and a small 28-amino-acid acidic cytoplasmic
vidual to another. After the initial discovery of ABO blood
C-terminus (Figure 21.1). Extracellular domains of AE1
groups at the beginning of the twentieth century, red cell
express the Diego blood group system, composed of two
serologists went on to discover hundreds of such polymor-
sets of antithetical antigens, Diego (Dia /Dib ) and Wright
phic epitopes and gradually succeeded in organizing them
(Wra /Wrb ). AE1 also expresses ABH and Ii antigens on a
into blood groups, antigens that arise from polymorphisms of
single multibranched N-linked oligosaccharide attached to
a single parent molecule. During the late twentieth century,
the fourth extracellular domain.
most of these blood groups were localized to carrier struc-
tures (proteins or polysaccharides). Discovery of the bio-
chemical and genetic basis for blood group antigens has now Transmembrane domain
led to exploration of the function of these proteins beyond
The transmembrane segment of AE1 (amino acids 404–882)
their importance in transfusion medicine.
is responsible for the electroneutral cotransport of HCO− 3 for
This chapter reviews current knowledge about erythro-
Cl− , and is also likely involved in the export of nitric oxide
cyte membrane proteins that bear blood group antigens and
(NO) equivalents. The membrane-spanning domain accom-
whose functional importance has been characterized. Pri-
modates the tide of HCO− 3 generated by the activity of ery-
mary attention is paid to two of the most interesting of
throcytic carbonic anhydrase on carbon dioxide (CO2 ) enter-
these proteins: the anion exchanger protein, which bears anti-
ing the RBC, thus increasing the capacity of the blood to carry
gens of the Diego blood group system, and the Rh proteins.
CO2 from the tissues to the lungs. In the peripheral tissues,
Functional information about other proteins is summarized
CO2 taken up by RBCs is converted to H+ and HCO− 3 by the
briefly.
action of carbonic anhydrase. The transmembrane domain
of AE1 exports HCO− −
3 in exchange for Cl to maintain phys-
+
Anion-exchanger protein 1 iological pH in the plasma, while H binds to hemoglobin
(band 3 protein) (Hb), right-shifting its oxygen (O2 ) affinity to facilitate O2
unloading. The reverse occurs in the lungs. The anion

Anion-exchanger protein 1 (AE1) is the most prevalent exchange domain is not specific for HCO− 3 ∕Cl and can
2− −
integral protein of the red blood cell (RBC) membrane also transport other anions (SO4 , H2 PO4 ), anionic phos-
pholipids, and even divalent cations. Of particular interest
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. is that AE1 also transports negatively charged NO metabo-
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. lites (NO− − −
2 , NO3 and OONO , see later), and more generally

285
286 Molecular Hematology

Transmembrane domain N-terminal cytoplasmic domain


The primary function of the N-terminal cytoplasmic domain
(amino acids 1–403) of AE1 (CDAE1) is presumably to
–SH –SH maintain erythrocyte integrity and stability by bracing the
cytoskeleton to the plasma membrane, and consequently it
C-terminus
may influence RBC rheology. This large hydrophilic domain
projects into the RBC cytoplasm with a spatial orientation
Carbonic anhydrase II
– – – that is strongly pH dependent. Under alkaline conditions (pH

– – Polyanionic N-terminus
10) the appendage is fully extended, whereas a slightly acidic
HS SH - Hemoglobin milieu (pH 6.5) induces flexion, such that the N-terminus is
HS SH
- Glyceraldehyde-3-phosphate brought close to the cell membrane. Flexibility in this part
dehydrogenase of AE1 is conferred by a proline-rich hinge region between
Cytoplasmic - Phosphofructokinase
domain - Aldolase
amino acids 175 and 190. The actual orientation of CDAE1
is likely to be a semi-flexed state, because the internal pH
of RBCs is closer to 7.0–7.2. However, given that carbonic
Proline-rich
hinge region anhydrase II binds to the C-terminus of AE1 in a juxtamem-
Fig. 21.1 Schematic representation of an anion-exchanger 1
brane location and that each AE1 channel can export 2000–
protein dimer. The cytoplasmic domains (CDAE1) are enclosed by the 3000 molecules of HCO− 3 per second, the micromilieu imme-
tinted circle. CDAE1 cysteine thiols are indicated by –SH, and ⊝ diately adjacent to CDAE1 may experience much lower pH
indicates the negative charges of acidic amino acids in the N-terminus. values under certain conditions, inducing greater degrees of
Proteins known to bind specifically to the N-terminus are also listed. CDAE1 flexion. These regions were once thought to be dis-
crete operational units, but it is now clear that remote alter-
ations in membrane domain structure can influence cyto-
plays a central role in the export of NO vasodilatory activ- plasmic domain activity, suggesting that these regions are
ity from RBCs that likely involves a Cys-NO relay, includ- functionally linked.
ing Cys843 within the membrane-spanning region, Cys317 CDAE1 contains high-affinity binding sites for three
within the N-terminus, and hemoglobin (Cysβ93), which peripheral membrane proteins that link AE1 to the under-
associates with the N-terminus of AE1. Importantly, trans- lying cytoskeletal meshwork. Ankyrin is a 206 kDa pyrami-
membrane C843 communicates with both the intracellu- dal protein that interacts with several sites throughout the
lar N-terminus and the extracellular plasma to mediate NO length of CDAE1, hinting that ankyrin binds to a flexed
transport across the membrane. An N-terminal truncated conformation of CDAE1 in vivo. Ankyrin also possesses
form of AE1 is expressed by the basolateral membrane of the three structural–functional domains: an N-terminal AE1 and
α-intercalated cells in the renal collecting ducts and mediates tubulin-binding domain, a middle spectrin-binding domain,
acid secretion. and a C-terminal domain containing sequences that reg-
ulate ankyrin binding to AE1 and spectrin. CDAE1 also
binds protein 4.2, or pallidin, a 77 kDa protein that binds
C-terminal cytoplasmic domain to ankyrin as well, acting to reinforce the AE1–ankyrin
The C-terminus of the transmembrane domain of AE1 con- interaction. Pallidin shows striking homology to transglu-
cludes in a 33-residue cytoplasmic protrusion consisting pre- taminases, but lacks enzymatic activity owing to loss of a nec-
dominantly of acidic amino acids and has been shown to essary conserved active site residue. Finally, CDAE1 inter-
possess a binding site for the carbonic anhydrase II iso- acts (at least in vitro) with protein 4.1, a 66 kDa moiety that
form. Using glutathione S-transferase (GST) fusion proteins firmly binds spectrin near the actin-binding site, thus inten-
with normal and mutated constructs, Vince and colleagues sifying the spectrin–actin interaction. The spectrin-binding
demonstrated that the critical binding site for carbonic anhy- activity of protein 4.1 is concurrently regulated by phos-
drase II is the acidic residue 887–890 region of AE1. A GST phorylation/dephosphorylation, Ca2+ –calmodulin associa-
fusion protein containing the C-terminal 33 amino acids of tion, and phosphatidylinositol pathways. In addition, spec-
AE2 could also bind carbonic anhydrase II, suggesting that trin, ankyrin, and AEI are known to be S-nitrosylated and it is
this tethering of carbonic anhydrase II to anion exchang- thought that S-nitrosylation leads to improvements in mem-
ers is a general mechanism promoting bicarbonate transport brane deformability.
across membranes. AE2 is the most widely expressed form of The N-terminus of CDAE1 also includes specific
AE, located in the basolateral membrane of nearly all epithe- high-affinity binding sites for the glycolytic enzymes
lial cells. glyceraldehyde 3-phosphate dehydrogenase (GAPDH),
Functions of blood group antigens 287

phosphofructokinase (PFK), and aldolase. Binding to FeNO SH FeO2 SNO FeNO SH


O2 SNO
CDAE1 results in a decrease in enzymatic activity (90% for
aldolase and 100% for GAPDH), while at the same time T R T
enzyme substrates, products, and cofactors displace the NO/SNO O2
bound enzymes, suggesting that specific CDAE1–enzyme Vein Lungs Arterioles/
interactions take place at the catalytic site or a linked Capillaries
allosteric site. Aldolase and PFK mutually inhibit each Fig. 21.2 Simplified allosteric model of NO–hemoglobin
other from binding CDAE1, but neither enzyme can dis- interactions. Nitric oxide (NO) (whether sourced from NO itself,
place bound GAPDH, indicative of a unique binding site for nitrite, or SNOs) binds to the heme groups of deoxygenated or T-state
GAPDH. S-nitrosylation of GAPDH, which may entail mod- hemoglobin (Hb). On oxygenation, Hb assumes the R conformation
ification of both active-site and allosteric Cys, regulates both and NO is transferred to the cysteine thiol in position 93 of the
β-chain (Cysβ93) to form S-nitrosohemoglobin (SNO-Hb). During
GAPDH activity and its binding to the RBC membrane. In
transit through the physiological oxygen gradient, partial pressure of
addition, phosphorylation of tyrosine 8 of AE1 regulates gly-
oxygen (PO2 ) falls, SNO-Hb converts to the T state, and NO is released
colytic enzyme binding, and thus activity. Binding affinities from Cysβ93 to evoke vasodilation (thus regulating blood flow).
of the glycolytic enzymes for CDAE1 are strongly dependent
on H+ and salt concentration, decreasing dramatically as
the pH and ionic strength are raised into the physiological Hb), SNO-Hb can release NO bioactivity to effect vasodila-
range, calling into question the extent of binding in vivo. tion (and potentially inhibit platelet activation; Figure 21.2).
However, several studies support the notion of significant However, effective NO export from the RBC requires that
enzyme–CDAE1 binding in the intact erythrocyte. NO itself be shielded from scavenging by the hemes in Hb.
This is accomplished by complexing NO with cysteine thiols
to form S-nitrosothiols (SNOs). Bioactive NO is thus present
Nitric oxide export
principally in the form of SNOs (rather than NO per se)
The role of NO as a critical third gas in the human res- and, as discussed earlier, these SNOs participate in a relay
piratory cycle (in addition to O2 and CO2 ) has been gain- whereby NO is transferred between Cys in Hb and AE1 and
ing acceptance and has been recently confirmed by stringent thereby exported from RBCs. Alternatively stated, RBCs
genetic criteria. Only three amino acids are strictly conserved sense ambient O2 levels by regulating the R/T transition
in all mammalian hemoglobins; two of them are required in Hb and adjust NO bioavailability accordingly. RBCs can
for O2 binding and one (Cysβ93) for NO bioactivity. Mice thereby cause microvessels to dilate in proportion to the
deficient in Cysβ93 (replaced by alanine) possess RBCs that degree of hypoxia. To the extent that tissue Po2 is determined
are essentially normal by all traditional measures (O2 bind- principally by blood flow, impairment in RBC-SNO function
ing, O2 affinity, O2 content, hematocrit, etc.), but cannot oxy- would be expected to lower tissue Po2 . The implications for
genate tissues adequately. The classic physiological response disorders of RBCs such as sickle cell disease are considered
of hypoxic vasodilation through which blood flow is cou- later.
pled to O2 delivery is profoundly impaired in these animals Compartmentalization of SNO in the erythrocyte mem-
whose hearts and limbs are starved of O2 . RBCs from these brane requires interaction between SNO-Hb and the inner
animals cannot export NO bioactivity and thus cannot dilate membrane surface. Hemoglobin binds to the cytoplasmic
blood vessels or stimulate brain centers to counteract hypoxia face of the RBC membrane via both non-specific binding and
(increase respiratory rate). Most recent data indicate that the specific protein–protein interactions. The predominant site
animals are very susceptible to heart attacks and heart fail- of specific interaction between Hb and the RBC membrane
ure, suggesting that hemoglobin S-nitrosylation is essential is through high-affinity binding to the N-terminal cytoplas-
for both normal cardiovascular function and cardioprotec- mic domain of AE1. More specifically, the site of Hb bind-
tion against heart disease. ing within CDAE1 has been localized to the polyanionic
Vascular-derived NO is generated chiefly by endothelial extreme N-terminus, which contains 18 acidic amino acids,
NO synthase and is a principal vasodilator and inhibitor of thus forming an anionic milieu at physiological pH. Analy-
platelet activation. Vascular NO regulates blood pressure, sis of co-crystals of Hb and a CDAE1 N-terminal peptide has
but is not involved directly in tissue oxygenation. A por- shown that this stretch of acidic residues inserts deeply into
tion of NO – whether in the form of NO itself, nitrite, or the 2,3-diphosphoglycerate-binding pocket formed between
S-nitrosothiols – ultimately penetrates circulating RBCs, β-globin subunits of tetrameric Hb. CDAE1 thus binds with
where it interacts with hemoglobin (Hb) at the heme pros- higher affinity to deoxyHb than to oxyHb (in which the β-
thetic groups or with β-chain cysteine thiols (Cysβ93) to cleft is occluded) and, consistent with the requirements of
form biologically active S-nitrosohemoglobin (SNO-Hb). thermodynamic linkage, the O2 affinity of Hb is reduced in
Under low O2 tension (conditions that promote the T state in the presence of isolated CDAE1.
288 Molecular Hematology

Interestingly, the AE1 of kidney intercalated cells is a enzymatic digestion of the IOVs with chymotrypsin. In
truncated isoform, lacking the N-terminal 65 residues of cross-linking studies, Hb forms a disulfide bond (–S–S–)
the CDAE1, and does not bind Hb, suggesting that these between its Cysβ93 thiols and the cytoplasmic domain
residues perform an AE1-related function specific to the cysteines of AE1, suggesting close spatial interplay in this
RBC. Binding of Hb to CDAE1 is non-cooperative but region. Studies on selenium transport across RBCs point
causes a rightward shift in the O2 dissociation curve. Simply to communication between Hb Cysβ93 and Cys317 in
stated, binding of Hb to CDAE1 stabilizes the “T” or deoxy CDAE1 and between Cys 317 and Cys843 in the membrane-
conformation of Hb. CDAE1 also binds oxidized Fe3+ or spanning region of AE1. In addition, Cys843 communicates
metHb more avidly than oxyHb. Because AE1 is present in with extracellular thiols in albumin. Thus coupled equilibria
insufficient quantity to bind a significant fraction of the total between Cys-NO in Hb, AE1, and extracellular proteins
Hb in an RBC, the influence of AE1 on overall Hb O2 delivery provides a potential route to relay NO from Hb into the
is probably negligible. This raises the possibility that AE1- plasma and vessel wall.
bound Hb serves an alternate function sufficiently executed Hemichromes, which are native Hb molecules with altered
by a small fraction of T-state Hb molecules. Indeed, SNO-Hb conformation near the heme, have a very high affinity for
binding to AE1 would promote the T structure and enhance CDAE1. Moreover, hemichromes formed by the covalent
NO group release. Thus, the CDAE1/SNO-Hb interaction modification of the Cysβ93 thiol (an SNO-Hb mimetic)
would appear to be designed to accelerate SNO release from bind preferentially to AE1. Therefore, transnitrosylation of
SNO-Hb (Figure 21.3). In support of this assertion, seques- a vicinal thiol (presumably Cys 317) within the cytoplasmic
tration of SNO in RBC membranes following treatment domain of AE1 is a strong candidate mechanism for trans-
with NO is markedly reduced by prior treatment with AE1 fer of the NO group from Cysβ93 of SNO-hemichromes to
inhibitors, including DIDS (4,4′ -diisothiocyanostilbene- the RBC membrane (Figure 21.3). In fact, like Hb, SNO-Hb
2,2′ -disulfonic acid), and phenyglyoxal and niflumic acid. forms disulfide links with AE1 under oxidative conditions
Further, SNO-Hb can transnitrosylate inside-out vesicles that predispose to hemichrome formation.
(IOVs) of RBC membranes, an effect blocked in IOVs In sum, a role for the anion exchange function of AE1
purified from RBCs treated with AE1 inhibitors. Finally, in transmembrane movement of NO bioactivity is likely.
S-nitrosylated AE1 can be immunoprecipitated from AE1 can import anionic NO metabolites such as nitrite,
the membranes of RBCs and from IOVs treated with which may participate in SNO-Hb formation. AE1-affiliated
SNO-Hb. SNO-Hb constitutes a major source of NO-based vasodila-
CDAE1 contains two cysteine residues (Cys201 and tory activity. NO derived from SNO-Hb is relayed to Cys
Cys317) with reactive thiols that are clustered at the subunit within the N-terminus of AE1. AE1-liganded (S)NO is trans-
interface of dimeric AE1. These thiols, which are surrounded ferred to Cys in plasma proteins (or the vascular wall), likely
by amino acids that fit an S-nitrosylation motif, are removed through the intermediacy of SNO within the membrane-
by chymotrypsin (which cleaves CDAE1 from the trans- spanning region of AE1.
membrane domain). Accordingly, transnitrosylation of RBC SNO-Hb in RBCs is distributed equally between the
IOVs by SNO-Hb is blocked by thiol-alkylating agents or cytosol and plasma membrane. However, only the plasma
membrane fraction is bioactive. Deficiency of plasma mem-
brane SNO can result from defects in either SNO-Hb syn-
thesis or its transfer of NO groups into the membrane. In
either case, RBC vasoactivity is compromised. Vasodilation
O2 by RBCs is impaired in several diseases, including sickle cell
Fe(II)O2 Fe(II) disease, which is characterized by a triple defect in SNO pro-
S NO S NO [NO]
cessing: SNO-Hb synthesis is reduced (an intrinsic property
R T of Hb S); the interaction between Hb S and AE1 is disrupted;
S
and the transfer of NO into the membrane is impaired. Most
AE1 notably, Cys residues of AE1 that would otherwise accept the
Cytosol Membrane
NO groups are oxidized in sickle RBCs. Membrane SNO is
therefore greatly diminished. Impaired vasodilation in sickle
Fig. 21.3 Interaction between S-nitrosohemoglobin (SNO-Hb)
and the cytoplasmic domain of erythrocyte anion-exchanger 1
patients corresponds with clinical symptoms (painful crises
(CDAE1) to effect vasodilation. Binding to CDAE1 stabilizes the T and organ damage) and may therefore represent a molec-
state of SNO-Hb and reduces affinity for nitric oxide (NO). SNO-Hb ular corollary of vaso-occlusive disorders. A deficiency in
transfers NO to vicinal thiols in CDAE1, which can then effect plasma membrane SNO-AE1 has also been found in banked
vasodilation and platelet inhibition. In this model, vasodilation entails blood, which likewise exhibits impaired vasodilation. Deficits
export of SNO through sequential transnitrosylation reactions, [NO]. in SNO-Hb and/or in RBC-mediated vasodilation have also
Functions of blood group antigens 289

been found in subsets of patients with heart failure, pul- a pathway involving protein disulfide isomerase. The only
monary hypertension, and diabetes. reported human case of total absence of AE1 occurred in a
Portuguese female delivered by cesarean section at 36 weeks
of gestation. The neonate was hydropic and severely anemic
Deficiencies and mutations of AE1
and had massive hepatosplenomegaly. Polymerase chain
Clinical conditions resulting from mutations or deficiencies reaction (PCR) demonstrated homozygosity for the Coimbra
in AE1 range from asymptomatic to severe, with manifesta- mutation (V488M), complete absence of AE1 and protein
tions predictably reflecting perturbation of one or both of 4.2, and reduction of spectrin and ankyrin by 43% and
the two primary functions of AE1, exchange/transport and 57%, respectively. Spherocytes and erythroblasts dominated
cytoskeletal membrane stability. A significant segment of the the peripheral blood morphology, while hyperchloremic
population (5–20%) are heterozygous for an AE1 polymor- acidosis was identified by age three months. The patient is
phism called “band 3 Memphis,” involving a lysine to glu- being treated with an intensive transfusion regimen, chelator
tamic acid substitution in codon 56, resulting in a function- therapy, and oral base replacement to counteract the acidosis.
ally normal AE1. Other than slightly delayed psychomotor development and
Southeast Asian ovalocytosis (SAO), characterized geno- stable nephrocalcinosis, her condition remains stable.
typically as a deletion of amino acids 400–408 in AE1, results In addition, hereditary renal tubular acidosis can result
in oval RBCs with increased rigidity, decreased osmotic from mutations in the genes encoding AE1, carbonic anhy-
fragility, and reduced expression of certain red cell antigens. drase II, or different subunits of the H+ -ATPase. Mutations
Natural selection dramatically increases the prevalence of of AE1 may account for up to 70% of cases in some areas of
SAO in areas with endemic malarial infection, given that the world. These mutations, which affect the renal isoform of
this defect confers reduced parasitemia and disease sever- AE1, cause defects in urinary acidification and also lead to
ity. However, homozygosity for the SAO mutation appears to hypokalemia.
be incompatible with life, as homozygotes have never been
observed (even in areas with 40% prevalence), and the inci-
dence of miscarriage in heterozygous parents is twice that for Rh protein
normals (20% vs. 9.8%).
Over 40 mutations of the AE1 protein have been found The rhesus (Rh) blood group antigens constitute the most
to be associated with a subset of individuals with domi- polymorphic family of human erythrocyte antigens and
nant hereditary spherocytosis, a condition characterized by are also the most frequent inducers of transfusion- and
loss of erythrocyte surface area and a mild to severe hemol- parturition-related alloantibodies. Three separate proteins
ysis, almost invariably improved by splenectomy. Despite make up the Rh family of proteins, including RhD and RhCE,
being scattered through the entire protein, many mutations which are unglycosylated proteins of 417 amino acids with
in the membrane-spanning domain involve substitutions of 97% homology, and rhesus-associated glycoprotein (RhAG),
highly conserved arginine residues, thought to be important which shows about 40% homology to RhD/CE. While scores
in maintaining proper membrane orientation. Other muta- of individual Rh antigens are recognized, only five epitopes
tions causing hereditary spherocytosis, including those in the (D, C, c, E, and e) carried on the RhD or CE proteins
cytoplasmic domain, lead to decreased binding affinities for are routinely identified; RhAG is thought to carry only the
the cytoskeletal bridging proteins ankyrin and pallidin. high-frequency Duclos antigen. RhCE expresses the antithet-
Complete AE1 deletion in a murine model results in a ical Cc/Ee antigens, while no antithetical antigen exists for
deadly hypercoagulable state in which only 5–10% of AE1 D. However, the presence (Rh-positive) and absence (Rh-
null mice survive the neonatal period. Almost uniformly, ani- negative) of the D antigen have sometimes been designated
mals were found at necropsy to have large thrombotic lesions as D and d, respectively. Thus, the eight most common haplo-
of the heart, subcapsular liver necrosis suggesting arteriolar types associated with the Rh system are DCe, DcE, Dce, dce,
ischemia, and large vein thrombi. This phenotype is also DCE, dcE, dCe, and dCE. The D antigen is a collection of
consistent with a defect in SNO export. Inaba and colleagues conformation-dependent epitopes along the entire RhD pro-
similarly described a total deficiency of AE1 in Japanese black tein. Deletion of the RHD gene characterizes most D-negative
cattle homozygous for a nonsense mutation (CGA → TGA) Caucasians, while in other populations (mainly Japanese and
in codon 646, causing an early stop signal. These animals black Africans) the D-negative phenotype appears to arise
exhibit moderate uncompensated anemias, spherocytosis, from a variety of missense and nonsense mutations in oth-
and mild acidosis at rest that is exacerbated by exercise. erwise intact RHD genes. In addition, there are multiple
It should be noted in this regard that RBCs may export “weak D,” “D category,” and other variant D phenotypes.
vasodilatory NO bioactivity through AE1-independent These are catalogued by several websites, including Rhesus-
mechanisms, including S-nitrosoglutathione transport and Base (http://www.rhesusbase.info). RhAG is a glycosylated
290 Molecular Hematology

409-amino-acid 50 kDa protein that appears to serve as a expression affects both NH3 and CO2 excretion. Thus, to
chaperone for RhD and RhCE to the RBC membrane; Rh date, the roles of RhAG, BG, and CG proteins in NH3 ,
antigens are only expressed if RhAG is also present. NH4 + transport, and whether transport is electrogenic or
Rh proteins appear to serve both as structural proteins electroneutral (with −H+ exchange) remain unclarified. At
necessary for appropriate membrane conformation and cell this point, further studies in mammalian systems will be
shape and also as transport proteins. Using known amino needed to confirm the function(s) of Rh and RhAG proteins.
acid sequences, studies using both topology prediction and Rhnull disease is characterized by the complete absence of
immunochemical analysis suggest that Rh proteins span the Rh proteins in the RBCs of affected subjects. The resulting
erythrocyte membrane 12 times, with both termini in the erythrocytes exhibit stomatocytic and spherocytic changes,
cytoplasm. Many multispanning membrane proteins have increased osmotic fragility, and deviations in ionic fluxes and
a transporter function. Rh proteins, including D, CE, and cell volume. Two variations of Rhnull disease have been iden-
RhAG, belong to the ammonium transport (Escherichia coli tified, historically termed “amorph” and “regulator,” based
AmtB)/methylammonium permease superfamily. RhAG, as on the underlying molecular defect from which they arise.
well as the related proteins RhBG and RhCG found in non- The amorph variety involves a genetic change in the RhCE
erythroid tissues, shares a molecular architecture much more gene coupled with deletion or inactivation of RHD, while the
similar to the E. coli AmtB channel than do RhD and RhCE. regulator phenotype results from two mutant RHAG genes
Current evidence is that RhD and RhCE proteins do not (homozygote or compound heterozygote). These null pheno-
function as transporters. However, Marini and colleagues types also display marked reductions in Rh-associated mem-
showed that RhAG expressed in a yeast mutant with deletions brane proteins, including the LW glycoprotein (ICAM-4),
in the three endogenous ammonium transporters (mep1Δ integrin-associated protein (IAP), and glycophorin B (a type
mep2Δ mep3Δ) could restore growth defects elicited by low- I membrane glycoprotein that bears the Ss antigens and may
ammonium medium. However, this study has been criticized serve to couple Rh with larger complexes containing AE1 and
because the expressed RhAG was not glycosylated and the GPA).
authors failed to show actual NH+ 4 uptake. Westhoff and
colleagues showed that Xenopus oocytes expressing solely
human RhAG protein were able to specifically transport NH+ 4 Glycophorins C and D
in a saturable, non-electrogenic, and pH-dependent man-
ner, suggesting that the RhAG transport mechanism involves Glycophorins C and D (GPC/GPD) are 32 and 23 kDa mem-
exchange of NH+ +
4 for H , resulting in the net transport of brane glycoproteins arising from the same four-exon gene
ammonia (NH3 ) into the cell. More recent work has sug- (GE, also referred to as GYPC, 2q14–q21) via alternative
gested that erythroid RhAG may serve to sequester NH3 in in-frame mRNA translation initiation sites, giving rise to
red cells, since it has been noted that blood plasma NH3 and the Gerbich erythrocyte antigens. The Gerbich blood group
NH+ 4 levels are three times lower than the levels found in ery- system currently includes six high-prevalence and five low-
throcytes. Furthermore, in non-erythroid tissues, RhBG and prevalence antigens, of which Ge:2 and Ge:3 are the most
RhCG are located at specific cellular surfaces where there is commonly identified and are used to define the Gerbich-
a disparate concentration of NH3 , such as at the luminal sur- null Leach phenotype (Ge:-2,-3). GPC and GPD are identi-
face in the kidney. cal, except that GPD has been truncated by 21 amino acids
Others have argued that Amt proteins act as gas channels at the N-terminus. There are two or three copies per cell of
handling NH3 , rather than as transporters of NH+ 4 cations, GPC for each copy of GPD. While being infrequent targets
and that both erythrocytic and non-mammalian Rh proteins of alloantibodies, GPC/GPD appear to be vital membrane-
represent CO2 transporters. Soupene and colleagues found stabilizing components serving as high-affinity binding sites
that Rh protein expression in the green alga Chlamydomonas for the peripheral membrane protein 4.1, which acts to affix
reinhardtii was significantly higher under elevated CO2 actin and spectrin elements. In addition to AE1–ankyrin–
conditions (air +3% CO2 for three hours) than in air alone spectrin, the GPC/GPD–protein 4.1–spectrin/actin bridge
(0.035% CO2 ). Further, Rh mRNA and protein expression constitutes the other major connection site between the ery-
remained low under conditions in which methylammonium throcyte membrane and its spectrin skeleton. Evidence for
uptake was high (nitrogen-limiting, with arginine as the sole the importance of GPC/GPD in membrane/cytoskeletal sta-
nitrogen source instead of ammonium chloride, NH4 Cl). bility originally arose from observations in cells contain-
Studies of CO2 transport in erythrocytes have variably sug- ing natural defects in one or more members of this bridge.
gested that Rh and/or RhAG may be functionally coupled For instance, complete absence of erythrocyte GPC/GPD
with either AE1 or aquaporin to regulate CO2 permeability. results in a rare form of hereditary elliptocytosis character-
In oocytes, RhCG expression also appears to facilitate ized by impaired red cell mechanical properties. However,
CO2 permeation. In zebrafish, knockdown of Rh protein these same GPC/GPD-deficient elliptocytic cells were also
Functions of blood group antigens 291

found to contain reduced amounts of protein 4.1. In addi- date. However, subjects with other RBC polymorphisms that
tion, GPC interacts with p55, a member of the membrane- are found in high frequencies in endemic malaria regions,
associated guanylate kinase family. The C-terminal region of notably sickle cell hemoglobin (Hb S), also demonstrate
GPC and GPD interacts with the PDZ domain in p55 as part infectivity rates similar to those of subjects lacking the sickle
of the ternary complex. trait phenotype. Nevertheless, individuals with Hb S trait
GPC/GPD deficiency is relatively common in Southeast show reduced parasite density and experience fewer cases of
Asia. One particular variant, the Melanesian Gerbich- cerebral or severe malaria. Thus, it may be that the GPCΔex3
negative phenotype, has been shown to reach a high phenotype is selected for by reducing erythrocyte invasion
frequency (46.5%) in coastal areas of Papua New Guinea, rates by malaria parasites, thus lessening disease severity.
where Plasmodium falciparum malaria is hyperendemic, Studies to determine whether there has been natural selection
suggesting that it may confer protection against erythrocyte of the GPCΔex3 allele as a safeguard against severe malaria
invasion by the parasite. This polymorphism is character- remain to be conducted.
ized genotypically by deletion of exon 3 (thus GPCΔex3)
and phenotypically by ovalocytic RBCs. The P. falciparum
erythrocyte-binding antigen EBA-140 appears to bind with Blood group antigen proteins that
high affinity to GPC, and this interaction mediates a principal function as adhesion molecules
invasion pathway into human erythrocytes. EBA-140 does
not bind to GPC in Ge-negative erythrocytes from Papua A number of erythrocyte membrane proteins have been iden-
residents homozygous for GPCΔex3, nor can P. falciparum tified as adhesion molecules. As shown in Table 21.1, these
infect such cells using this invasion pathway. Using recombi- include the proteins that bear the Lutheran, LW, and Indian
nant P. falciparum EBA-140 expressed in Baculovirus, inves- blood group antigens.
tigators have demonstrated that EBA-140 binding is depen- CD44 bears the Indian antigens and was first described
dent on both GPC amino acid residues 36–63 as well as the on erythrocytes as In(Lu)-related p80, because expression
N-glycan attached to the N-terminal of GPC (but not GPD). on erythrocytes is downregulated by a dominant In(Lu)
Interestingly, while the invasion of GPCΔex3 erythrocytes allele, which was eventually identified by Singleton and
by P. falciparum parasites is less efficient in vitro, no dif- colleagues as any of numerous single-copy mutations in
ferences in infection rates for either P. falciparum or Plas- EKLF1. CD44 was the first erythrocyte membrane protein
modium vivax in Ge-negative subjects have been observed to to be characterized as an adhesion molecule and bears

Table 21.1 Adhesion molecules of erythrocytes

Blood group antigen Alternative name(s) Ligand/adhesive function

Ina /Inb CD44, In(Lu)-related p80 Hyaluronan, possibly also fibronectin/also involved in immune
stimulation and signaling between cells.
JMH CD108, semaphorin K1 (SEMA7A) α1β1 integrin, plexin C1/possible role in adhesion of activated
lymphocytes
Lutheran (LU) B-CAM/LU, CD239 Laminin containing the α5 chain, α4β1 integrin
LW CD242, ICAM-4, LW Leukocyte, endothelial, and platelet integrins, including αVβ3,
LFA-1 (αLβ2), Mac-1 (αMβ2), αVβ1, αVβ5, αIIbβ3
Naka (platelets) CD36 (reticulocytes only), platelet GPIV, Thrombospondin (platelets), LDL
Naka (platelets)
Oka CD147, neurothelin, EMMPRIN Type IV collagen, fibronectin/laminin in other tissues/required for
expression of monocarboxylate (lactate) transporters
None known VLA-4 (reticulocytes only), α4β1 integrin Thrombospondin, VCAM-1, possibly fibronectin
(CD49d/CD29)
None known CD47, integrin-associated protein (IAP) Thrombospondin, SIRP-1α
None known CD58, lymphocyte-associated antigen-3 CD2
(LFA-3)
RAPH1 CD151, MIC2 gene product Integrins and laminin/proposed role in development and
structure of kidney and skin basement membranes
Scianna (SC) ERMAP (erythroid membrane-associated Possible adhesion molecule that localizes to points of cell–cell
protein) contact
292 Molecular Hematology

homology to the cartilage link and proteoglycan core pro- B-CAM/LU, thereby activating its adhesive function. Genetic
teins, which are known to interact with hyaluronan. This studies of red cell adhesion in patients with sickle cell disease
similarity led to the demonstration that CD44 also bound have shown that higher adhesion to laminin is associated
to this component of extracellular matrix and basement with specific polymorphisms in the genes encoding the
membranes. On leukocytes, CD44 appears to be involved in β2 -adrenergic receptor and adenylyl cyclase 6, one of the
the interaction of leukocytes with endothelial cells, including more ubiquitous isoforms of adenylyl cyclase. B-CAM/LU
the homing of lymphocytes to peripheral lymphoid organs has also been implicated in the abnormal behavior of red
and sites of inflammation. Ligand binding to CD44 can cells from patients with polycythemia vera. In this disease,
also induce cytokine release and T-cell activation. However, red cells are also abnormally adherent to laminin and bear
on erythrocytes the functional significance of CD44 is less constitutively phosphorylated B-CAM/LU, most likely due
clear. One report has indicated that CD44 contributes to the to downstream effects of activated JAK2. Finally, B-CAM/LU
ability of early erythroid precursors to bind to bone marrow has also been shown to mediate adhesion to the α4β1
fibronectin, and CD44 also contributes to red cell–leukocyte integrin, also known as very late antigen 4 (VLA-4), which is
interactions in sickle cell disease. Further, CD44 plays an expressed by reticulocytes and leukocytes, among other cells.
important role in the metastatic behavior of tumor cells. However, the contribution of B-CAM/LU-mediated red cell
The molecule that bears Lutheran blood group antigens adhesion to the pathophysiology of vaso-occlusion or to the
has been identified as a laminin receptor. Lutheran antigens frequent thrombotic complications of polycythemia remains
are expressed by two proteins that arise from alternate splic- unproven in vivo, although B-CAM/LU adhesive activity has
ing of a single gene. These two proteins (sometimes called been proposed as a biomarker for JAK2 mutation.
B-CAM and LU) are identical except for their cytoplasmic The LW protein also appears to be an adhesion receptor,
domains, and they both appear to function equally well although, unlike CD44 and B-CAM/LU, LW interacts
as laminin receptors. Interestingly, B-CAM/LU expression with cell surface proteins rather than extracellular matrix
and function are increased on red cells from patients components. LW has also been called intercellular adhesion
with sickle cell disease, and this function can be activated molecule (ICAM)-4 because it is highly homologous to
further by exposure of sickle red cells to epinephrine and other members of the ICAM family of adhesion molecules.
other reagents that lead to increased intracellular cyclic Like other ICAMs, LW can bind to the leukocyte integrins
adenosine monophosphate (AMP). Increase in cyclic AMP CD11a/CD18 (LFA-1, αLβ2), CD11b/CD18 (MAC-1,
leads to activation of protein kinase A, which phosphorylates αMβ2), and CD11c/CD18. Recently, it has been shown that

Table 21.2 Diverse functions of blood group antigen proteins

Blood group Alternative name(s) Function

Cartwright (Yt) ACHE Acetylcholinesterase


Chido/Rodgers (CH/RG) C4B/C4A C4B and C4A complement components adsorbed from plasma
Colton (Co) Aquaporin-1 (AQP-1) Water channel
Cromer (Cr) Decay accelerating factor, CD55 Promotes the degradation of C3 and C5 convertases
Dombrock (Do) DO (ADP-ribosyltransferase-4 (ART4)) ADP-ribosyltransferase ectoenzyme
Duffy (Fy) DARC Promiscuous chemokine receptor, clears proinflammatory cytokines,
and also affects circulating neutrophil count
Gil AQP3 Water channel that also transports other small molecules, including
glycerol
Jr CD338, JR, ABCG2 Adenosine triphosphate (ATP)-dependent transport protein with broad
substrate specificity
Kell (K) KEL Zinc-binding neutral endopeptidase; endothelin-3 converting enzyme
that cleaves big endothelin-3 to bioactive endothelin-3
Kidd (Jk) UT1 Urea transporter important in renal urea concentrating ability
Knops/McCoy (Kn/McC) C3b/C4b receptor (CD35), Binds C3b and C4b and facilitates immune clearance
complement receptor type 1
Kx XK Possibly a neurotransmitter transporter; deficiency causes
neuroacanthocytosis or McLeod syndrome
Lan ABCB6 Binds heme and porphyrins and has a role in ATP-dependent uptake
into mitochondria
Functions of blood group antigens 293

LW can bind to αV integrins, as well as to the αIIbβ3 integrin Bruce, L.J., Kay, M.M.B., Lawrence, C. et al. (1993). Band 3 HT, a human
of platelets. LW contributes to the adhesion of sickle red cells red-cell variant associated with acanthocytosis and increased anion
to the endothelium by binding to αVβ3. This interaction transport, carries the mutation pro 868 leu in the membrane domain
appears to be the dominant high-affinity interaction between of band 3. Biochem. J. 293: 317–320.
sickle red cells both in vitro and in vivo. Interestingly, LW Celedon, G., Gonzalez, G., Pino, J., and Lissi, E.A. (2007). Peroxynitrite
oxidizes erythrocyte membrane band 3 protein and diminishes its
adhesive function is also activated by exposure of red cells
anion transport capacity. Free Radical Res. 41: 316–323.
to epinephrine, which leads to cyclic AMP- and protein Chu, H., Breite, A., Ciraolo, P. et al. (2008). Characterization of the
kinase A-dependent LW phosphorylation. In addition, some deoxyhemoglobin binding site on human erythrocyte band 3: impli-
investigators speculate that LW may be important in facili- cations for O2 regulation of erythrocyte properties. Blood 111: 932–
tating the adhesion of erythroid precursors to bone marrow 938.
macrophages in erythroid islands during erythropoiesis. Chu, H. and Low, P.S. (2006). Mapping of glycolytic enzyme-binding
sites on human erythrocyte band 3. Biochem. J. 400: 143–151.
Cohen, C.M., Dotimas, E., and Korsgren, C. (1993). Human erythro-
Blood group antigen proteins cyte membrane protein 4.2 (pallidin). Semin. Hematol. 30: 119–
with other functions 137.
Davis, L., Lux, S.E., and Bennett, V. (1989). Mapping the ankyrin-
binding site of the human erythrocyte anion exchanger. J. Biol. Chem.
Table 21.2 lists blood group antigens associated with func-
264: 9665–9672.
tions other than those already discussed. As indicated, pro- Funder, J. and Wieth, J.O. (1966). Chloride and hydrogen ion distribu-
teins bearing blood group antigens have a broad diversity tion between human red cells and plasma. Acta Physiol. Scand. 68:
of functions. Some, such as the proteins that bear the Kidd 234–245.
and Colton blood group antigens, are transporters. Others, Galli, F., Rovidati, S., Ghibelli, L., and Canestrari, F. (1998).
such as those that bear the Cartwright and Kell antigens, S-nitrosylation of glyceraldehyde-3-phosphate dehydrogenase
are ectoenzymes. In addition, erythrocytes bear receptors decreases the enzyme affinity to the erythrocyte membrane. Nitric
for complement components and chemokines. The degree to Oxide 2: 17–27.
which polymorphisms and deficiency of these proteins con- Gaston, B., May, W.J., Sullivan, S. et al. (2014). Essential role of
tribute to human disease continues to be further explored. hemoglobin beta-93-cysteine in posthypoxia facilitation of breathing
in conscious mice. J. Appl. Phys. 116 (10): 1290–1299.
Genton, B., Al-Yaman, F., Mgone, C.S. et al. (1995). Ovalocytosis and
cerebral malaria. Nature 378: 564–565.
Summary Grau, M., Pauly, S., Ali, J. et al. (2013). RBC-NOS-dependent
S-nitrosylation of cytoskeletal proteins improves RBC deformability.
Proteins that bear blood group antigens have diverse func- PLoS One 8: e56759.
tions, and some proteins, such as AE1, encompass several Guizouarn, H., Martial, S., Gabillat, N., and Borgese, F. (2007). Point
functions within a single protein molecule. Abnormalities of mutations involved in red cell stomatocytosis convert the electroneu-
these proteins, in the form of either deficiencies or mutations, tral anion exchanger 1 to a nonselective cation conductance. Blood
can lead to red cell disorders, such as hemolytic anemia, or 110: 2158–2165.
have more far-reaching effects, as in the association of Kx Gutierrez, E. and Sung, L.A. (2007). Interactions of recombinant
mouse erythrocyte transglutaminase with membrane skeletal pro-
deficiency with neuroacanthocytosis and mutations of AE1
teins. J. Membr. Biol. 219: 93–104.
with renal tubular acidosis. Finally, these proteins undoubt-
Hassoun, H., Wang, Y., Vassiliadis, J. et al. (1998). Targeted inactivation
edly contribute both to normal physiology and to the patho- of murine band 3 (AE1) gene produces a hypercoagulable state caus-
physiology of human diseases, including sickle cell anemia, ing widespread thrombosis in vivo. Blood 92: 1785–1792.
malaria, and perhaps others. Hess, D.T., Matsumoto, A., Kim, S.-O. et al. (2005). Protein S-
nitrosylation: purview and parameters. Nat. Rev. Mol. Cell Biol. 6:
150–166.
Further reading Hongo, M., Haratake, M., Fuchigami, T., and Nakayam, M. (2012). A
thiol-mediated active transport of selenium by the erythroid anion
exchange 1 protein. Dalton Trans. 41: 7340–7349.
AE1
Inaba, M., Yawata, A., Koshino, I. et al. (1996). Defective anion trans-
Anderson, R.A. and Marchesi, V.T. (1985). Regulation of the association port and marked spherocytosis with membrane instability caused by
of membrane skeletal protein 4.1 with glycophorin by polyphospho- hereditary total deficiency of red cell band 3 in cattle due to a non-
inositide. Nature 318: 295–298. sense mutation. J. Clin. Invest. 97: 1804–1817.
Bennett-Guerrero, E., Veldman, T.H., Doctor, A. et al. (2007). Evolution Jarolim, P., Palek, J., Amato, D. et al. (1991). Deletion in erythrocyte
of adverse changes in stored RBCs. Proc. Natl. Acad. Sci. U.S.A. 104: band 3 gene in malaria-resistant Southeast Asian ovalocytosis. Proc.
17063–17068. Natl. Acad. Sci. U.S.A. 88: 11022–11026.
294 Molecular Hematology

Jarolim, P., Rubin, H.L., Brabec, V. et al. (1995). Mutations of conserved Rauenbuehler, P.B., Cordes, K.A., and Salhany, J.M. (1982). Identifica-
arginines in the membrane domain of erythroid band 3 protein lead tion of the hemoglobin binding sites on the inner surface of the ery-
to a decrease in membrane-associated band 3 and to the phenotype throcyte membrane. Biochim. Biophys. Acta 692: 361–370.
of hereditary spherocytosis. Blood 85: 634–640. Reinhart, A.F.R., Casey, J.R., Kalli, A.C. et al. (2016). Band 3, the human
Jia, L., Bonaventura, C., Bonaventura, J. et al. (1996). red cell chloride/bicarbonate anion exchanger (AE1, SLC4A1), in a
S-Nitrosohaemoglobin: a dynamic activity of blood involved in structural context. Biochim. Biophys. Acta 1858: 1507–1532.
vascular control. Nature 380: 221–226. Reynolds, J.D., Ahearn, G.S., Angelo, M. et al. (2007).
Khositseth, S., Sirikanerat, A., Wongbenjarat, K. et al. (2007). Distal S-Nitrosohemoglobin deficiency: a mechanism for loss of physio-
renal tubular acidosis associated with anion exchanger 1 mutations logical activity in banked blood. Proc. Natl. Acad. Sci. U.S.A. 104:
in children in Thailand. Am. J. Kidney Dis. 49: 841–850. 17058–17062.
Kindt, J.T., Woods, A., Martin, B.M. et al. (1992). Covalent alteration of Ribeiro, M.L., Allosio, N., Almeida, H. et al. (2000). Severe hereditary
the prosthetic heme of human hemoglobin by BrCCl3 cross-linking spherocytosis and distal renal tubular acidosis associated with the
of heme to cysteine residue 93. J. Biol. Chem. 267: 8739–8743. total absence of band 3. Blood 96: 1602–1604.
Kittanakom, S., Cordat, E., and Reithmeier, R.A. (2008). Dominant- Shingles, R., Roh, M.H., and McCarty, R.E. (1997). Direct measure-
negative effect of Southeast Asian ovalocytosis anion exchanger 1 in ment of nitrite transport across erythrocyte membrane vesicles using
compound heterozygous distal renal tubular acidosis. Biochem. J. 410: the fluorescent probe, 6-methoxy-n-(3-sulfopropyl) quinolinium.
271–281. J. Bioenerg. Biomembr. 29: 611–616.
Kollert-Jons, A., Wagner, S., Hubner, S. et al. (1993). Anion exchanger 1 Singel, D.J. and Stamler, J.S. (2005). Chemical physiology of blood
in human kidney and oncocytoma differs from erythroid AE1 in its flow regulation by red blood cells: the role of nitric oxide and
NH2 terminus. Am. J. Phys. 265: F813–F821. S-nitrosohemoglobin. Annu. Rev. Physiol. 67: 99–145.
Ling, E., Danilov, Y.N., and Cohen, C.M. (1988). Modulation of red cell Stamler, J.S., Jia, L., Eu, J.P. et al. (1997). Blood flow regulation by
band 4.1 function by cAMP-dependent kinase and protein kinase C S-nitrosohemoglobin in the physiological oxygen gradient. Science
phosphorylation. J. Biol. Chem. 263: 2209–2216. 276: 2034–2037.
Liu, S.C., Zhai, S., Palek, J. et al. (1990). Molecular defect of the band 3 Stamler, J.S., Toone, E.J., Lipton, S.A. et al. (1997). (S)NO signals:
protein in Southeast Asian ovalocytosis. N. Engl. J. Med. 323: 1558– translocation, regulation, and a consensus motif. Neuron 18: 691–696.
1560. Stehberger, P.A., Shmukler, B.E., Stuart-Tilley, A.K. et al. (2007). Distal
Low, P.S., Allen, D.P., Zioncheck, T.F. et al. (1987). Tyrosine phospho- renal tubular acidosis in mice lacking the AE1 (band3) Cl−/HCO3−
rylation of band 3 inhibits peripheral protein binding. J. Biol. Chem. exchanger (slc4a1). J. Am. Soc. Nephrol. 18: 1408–1418.
262: 4592–4596. Thevenin, B.J.M., Bicknese, S.E., Park, J. et al. (1996). Distance between
McMahon, T.J., Moon, R.E., Luschinger, B.P. et al. (2002). Nitric oxide cys-201 in erythrocyte band 3 and the bilayer measured by single-
in the human respiratory cycle. Nat. Med. 8: 711–717. photon radioluminescence. Biophys. J. 71: 2645–2655.
McMahon, T.J., Stone, A.E., Bonaventura, J. et al. (2000). Functional Tsuneshige, A., Imai, K., and Tyuma, I. (1987). The binding of
coupling of oxygen binding and vasoactivity in S-nitrosohemoglobin. hemoglobin to red cell membrane lowers its oxygen affinity. J.
J. Biol. Chem. 275: 16738–16745. Biochem. 101: 695–704.
Miki, Y., Tazawa, T., Hirano, K. et al. (2007). Clearance of oxidized ery- Vince, J.W. and Reithmeier, R.A.F. (2000). Identification of the carbonic
throcytes by macrophages: involvement of caspases in the genera- anhydrase II binding site in the Cl−HCO3−anion exchanger AE1.
tion of clearance signal at band 3 glycoprotein. Biochem. Biophys. Res. Biochemistry 39: 5527–5533.
Commun. 363: 57–62. von Kalckreuth, V., Evans, J.A., Timmann, C. et al. (2006). Promoter
Nagarajan, S., Raj, R.K., Saravanakumar, V. et al. (2016). Mechanical per- polymorphism of the anion-exchange protein 1 associated with
turbations trigger endothelial nitric oxide synthase activity in human severe malarial anemia and fatality. J. Infect. Dis. 194: 949–957.
red blood cells. Sci. Rep. 6: 26935. Weaver, D.C., Pasternack, G.R., and Marchesi, V.T. (1984). The struc-
Ohanian, V., Wolfe, L.C., John, K.M. et al. (1984). Analysis of the ternary tural basis of ankyrin function. II. Identification of two functional
interaction of the red cell membrane skeletal proteins spectrin, actin, domains. J. Biol. Chem. 259: 6170–6175.
and 4.1. Biochemistry 23: 4416–4420. Zhang, R.L., Hess, D.T., Qian, Z. et al. (2015). Hemoglobin βCys93
Pawloski, J.R., Hess, D.T., and Stamler, J.S. (2001). Export by red blood is essential for cardiovascular function and integrated response to
cells of nitric oxide bioactivity. Nature 409: 622–626. hypoxia. Proc. Natl. Acad. Sci. U.S.A. 112: 6425–6430.
Pawloski, J.R., Hess, D.T., and Stamler, J.S. (2005). Impaired vasodilation Zhang RL Hess DT, Reynolds JD, Stamler JS. (2016) Hemoglobin S-
by red blood cells in sickle cell disease. Proc. Natl. Acad. Sci. U.S.A. nitrosylation plays an essential role in cardioprotection. J. Clin. Invest.
102: 2531–2536. 126(12): 4654–4658.
Pawloski, J.R. and Stamler, J.S. (2002). Nitric oxide in RBCs. Transfusion Zhou, J. and Low, P.S. (2001). Characterization of the reversible confor-
42: 1603–1609. mational equilibrium in the cytoplasmic domain of human erythro-
Pawloski, J.R., Swaminathan, R.V., and Stamler, J.S. (1998). Cell-free and cyte membrane band 3. J. Biol. Chem. 276: 38147–38151.
erythrocyte S-nitrosohemoglobin inhibits human platelet aggrega-
tion. Circulation 97: 263–267. Rh
Perrotta, S., Borriello, A., Scaloni, A. et al. (2005). The N-terminal 11
amino acids of human erythrocyte band 3 are critical for aldolase Avent, N.D., Madgett, T.E., Lee, Z.E. et al. (2006). Molecular biology of
binding and protein phosphorylation: implications for band 3 func- Rh proteins and relevance to molecular medicine. Expert Rev. Mol.
tion. Blood 106: 4359–4366. Med. 8: 1–20.
Functions of blood group antigens 295

Callebaut, I., Dulin, F., Bertrand, O. et al. (2006). Hydrophobic cluster homologue proteins forming the protein 4.1/FERM superfamily.
analysis and modeling of the human Rh protein three-dimensional Folia Histochem. Cytobiol. 44: 231–248.
structures. Transfus. Clin. Biol. 13: 70–84. Gascard, P. and Cohen, C.M. (1994). Absence of high-affinity band 4.1
Conroy, M.J., Bullough, P.A., Merrick, M., and Avent, N.D. (2005). Mod- binding sites from membranes of glycophorin C- and D-deficient
elling the human rhesus proteins: implications for structure and func- (Leach phenotype) erythrocytes. Blood 83: 1102–1108.
tion. Br. J. Haematol. 131: 543–551. Hemming, N.J., Anstee, D.J., Mawby, W.J. et al. (1994). Localization of
Endeward, V., Cartron, J.P., Ripoche, P., and Gros, G. (2008). RhAG pro- the protein 4.1-binding site on human erythrocyte glycophorins C
tein of the rhesus complex is a CO2 channel in the human red cell and D. Biochem. J. 299: 191–196.
membrane. FASEB J. 22: 64–73. Kusunoki, H. and Kohno, T. (2007). Structural insight into the inter-
Khademi, S. and Stroud, R.M. (2006). The Amt/MEP/Rh family: struc- action between the p55 PDZ domain and glycophorin C. Biochem.
ture of AmtB and the mechanism of ammonia gas conduction. Phys- Biophys. Res. Commun. 359: 972–978.
iology (Bethesda) 21: 419–429. Lobo, C.A., Rodriguez, M., Reid, M., and Lustigman, S. (2003). Gly-
Le Van Kim, C., Colin, Y., and Cartron, J.-P. (2006). Rh proteins: key cophorin C is the receptor for the Plasmodium falciparum erythro-
structural and functional components of the red cell membrane. cyte binding ligand PfEBP-2 (BAEBL). Blood 101: 4628–4631.
Blood Rev. 20: 93–110. Maier, A.G., Duraisingh, M.T., Reeder, J.C. et al. (2003). Plasmodium
Marini, A.M., Urrestarazu, A., Beauwens, R. et al. (1997). The Rh (rhe- falciparum erythrocyte invasion through glycophorin C and selection
sus) blood group polypeptides are related to NH4+ transporters. for Gerbich negativity in human populations. Nat. Med. 9: 87–92.
Trends Biochem. Sci. 22: 460–461. Mayer, D.C., Jiang, L., Achur, R.N. et al. (2006). The glycophorin C N-
Nakhoul, N.L. and Lee Hamm, L. (2013). Characteristics of mammalian linked glycan is a critical component of the ligand for the Plasmodium
Rh glycoproteins (SLC42 transporters) and their role in acid-base falciparum erythrocyte receptor BAEBL. Proc. Natl. Acad. Sci. U.S.A.
transport. Mol. Aspects Med. 34: 629–637. 103: 2358–2362.
Nicolas, V., Mouro-Chanteloup, I., Lopez, C. et al. (2006). Functional Patel, S.S., Mehlotra, R.K., Kastens, W. et al. (2001). The association of
interaction between Rh proteins and the spectrin-based skeleton in the glycophorin C exon 3 deletion with ovalocytosis and malaria sus-
erythroid and epithelial cells. Transfus. Clin. Biol. 13: 23–28. ceptibility in the Wosera, Papua New Guinea. Blood 98: 3489–3491.
Perry, S.F., Braun, M.H., Noland, M. et al. (2010). Do zebrafish Rh pro- Pinder, J.C., Chung, A., Reid, M.E. et al. (1993). Membrane attachment
teins act as dual ammonia-CO2 channels? J. Exp. Zool. Ecol. Genet. sites for the membrane cytoskeletal protein 4.1 of the red blood cell.
Physiol. 313: 618–621. Blood 82: 3482–3488.
Planelles, G. (2007). Ammonium homeostasis and human rhesus glyco- Rydzak, J., Kaczmarek, R., Czerwinski, M. et al. (2015). The baculovirus-
proteins. Nephron Physiol. 105: 11–17. expressed binding region of Plasmodium falciparum EBA-140 ligand
Soupene, E., Inwood, W., and Kustu, S. (2004). Lack of the rhesus protein and its glycophorin C binding specificity. PLoS One 10: e0115437.
Rh1 impairs growth of the green alga Chlamydomonas reinhardtii at Takakuwa, Y., Tchernia, G., Rossi, M. et al. (1986). Restoration of nor-
high CO2 . Proc. Natl. Acad. Sci. U.S.A. 101: 7787–7792. mal membrane stability to unstable protein 4.1-deficient erythrocyte
Soupene, E., King, N., Feild, E. et al. (2002). Rhesus expression in a green membranes by incorporation of purified protein 4.1. J. Clin. Invest.
alga is regulated by CO2 . Proc. Natl. Acad. Sci. U.S.A. 99: 7769–7773. 78: 80–85.
Szekely, D., Chapman, B.E., Bubb, W.A., and Kuchel, P.W. (2006). Rapid Telen, M.J., Le Van Kim, C., Chung, A. et al. (1991). Molecular basis for
exchange of fluoroethylamine via the rhesus complex in human ery- elliptocytosis associated with glycophorin C and D deficiency in the
throcytes: 19F NMR magnetization transfer analysis showing compe- Leach phenotype. Blood 78: 1603–1606.
tition by ammonia and ammonia analogues. Biochemistry 45: 9354–
9361.
Westhoff, C.M. (2007). The structure and function of the Rh antigen Adhesion proteins
complex. Semin. Hematol. 44: 42–50.
Westhoff, C.M., Ferreri-Jacobia, M., Mak, D.O., and Foskett, J.K. (2002). Bailly, P., Tontti, E., Hermand, P. et al. (1995). The red cell LW blood
Identification of the erythrocyte Rh blood group glycoprotein as group protein is an intercellular adhesion molecule which binds to
a mammalian ammonium transporter. J. Biol. Chem. 277: 12499– CD11/CD18 leukocyte integrins. Eur. J. Immunol. 25: 3316–3320.
12502. Brittain, J.E., Han, J., Ataga, K.I. et al. (2004). Mechanism of CD47-
induced alpha4beta1 integrin activation and adhesion in sickle retic-
ulocytes. J. Biol. Chem. 279: 42393–42402.
Glycophorins C and D De Grandis, M., Cambot, M., Wautier, M.P. et al. (2013). JAK2V617F
activates Lu/BCAM-mediated red cell adhesion in polycythemia vera
Chang, S.H. and Low, P.S. (2001). Regulation of the glycophorin C– through an EpoR-independent Rap1/Akt pathway. Blood 121: 658–
protein 4.1 membrane-to-skeleton bridge and evaluation of its contri- 665.
bution to erythrocyte membrane stability. J. Biol. Chem. 276: 22223– De Grandis, M., Cassinat, B., Kiladjian, J.J. et al. (2015). Lu/BCAM-
22230. mediated cell adhesion as biological marker of JAK2V617F activity
Colin, Y., Le Van Kim, C., Tsapis, A. et al. (1989). Human erythrocyte in erythrocytes of polycythemia vera patients. Am. J. Hematol. 90:
glycophorin C. Gene structure and rearrangement in genetic variants. E137–E138.
J. Biol. Chem. 264: 3773–3780. El Nemer, W., Rahuel, C., Colin, Y. et al. (1997). Organization of the
Diakowski, W., Grzybek, M., and Sikorski, A.F. (2006). Protein 4.1, a human LU gene and molecular basis of the Lu(a)/Lu(b) blood group
component of the erythrocyte membrane skeleton and its related polymorphism. Blood 89: 4608–4616.
296 Molecular Hematology

El Nemer, W., Wautier, M.P., Rahuel, C. et al. (2007). Endothe- vera is mediated by laminin alpha5 chain and Lu/BCAM. Blood 110:
lial Lu/BCAM glycoproteins are novel ligands for red blood cell 894–901.
alpha4beta1 integrin: role in adhesion of sickle red blood cells to Zen, Q., Batchvarova, M., Twyman, C.A. et al. (2004). B-CAM/LU
endothelial cells. Blood 109: 3544–3551. expression and the role of B-CAM/LU activation in binding of low-
Eyler, C.E., Jackson, T., Elliott, L.E. et al. (2008). Beta2-adrenergic recep- and high-density red cells to laminin in sickle cell disease. Am. J.
tor and adenylate cyclase gene polymorphisms affect sickle red cell Hematol. 75: 63–72.
adhesion. Br. J. Haematol. 141: 105–108. Zen, Q., Cottman, M., Truskey, G. et al. (1999). Critical factors in basal
Hermand, P., Gane, P., Huet, M. et al. (2003). Red cell ICAM-4 is a novel cell adhesion molecule/Lutheran-mediated adhesion to laminin.
ligand for platelet-activated alpha IIbbeta 3 integrin. J. Biol. Chem. J. Biol. Chem. 274: 728–734.
278: 4892–4898. Zennadi, R., Hines, P.C., De Castro, L.M. et al. (2004). Epinephrine
Hines, P.C., Zen, Q., Burney, S.N. et al. (2003). Novel epinephrine and acts via erythroid signaling pathways to activate sickle cell adhe-
cyclic AMP mediated activation of B-CAM/LU-dependent sickle (SS) sion to endothelium via LW–αvβ3 interactions. Blood 104: 3774–
RBC adhesion. Blood 101: 3281–3287. 3781.
Lee, G., Lo, A., Short, S.A. et al. Targeted gene deletion demonstrates Zennadi, R., Moeller, B.J., Whalen, E.J. et al. (2007). Epinephrine-
that the cell adhesion molecule ICAM-4 is critical for erythroblastic induced activation of LW-mediated sickle cell adhesion and vaso-
island formation. Blood 108: 2064–2071. occlusion in vivo. Blood 110: 2708–2717.
Mankelow, T.J., Burton, N., Stefansdottir, F.O. et al. (2007). The Laminin
511/521-binding site on the Lutheran blood group glycoprotein is
Other blood group antigen proteins
located at the flexible junction of Ig domains 2 and 3. Blood 110: 3398–
3406. Afenyi-Annan, A., Kail, M., Combs, M.R. et al. (2008). Lack of Duffy
Murphy, M.M., Evans, A., Zayed, M.A. et al. (2005). Role of Rap1 in antigen expression is associated with organ damage in patients with
promoting sickle red blood cell adhesion to laminin via BCAM/Lu. sickle cell disease. Transfusion 48: 917–924.
Blood 105: 3322–3329. Andolfo, I., Russo, R., Gambale, A., and Iolascon, A. (2016). New
Singleton, B.K., Burton, N.M., Green, C. et al. (2008). Mutations in insights on hereditary erythrocyte membrane defects. Haematolog-
EKLF/KLF1 form the molecular basis of the rare blood group In(Lu) ica 101: 1284–1294.
phenotype. Blood 112: 2081–2088. Hara-Chikuma, M. and Verkman, A.S. (2005). Aquaporin-3 func-
Telen, M.J., Eisenbarth, G.S., and Haynes, B.F. (1983). Human erythro- tions as a glycerol transporter in mammalian skin. Biol. Cell 97:
cyte antigens. Regulation of expression of a novel erythrocyte sur- 479–486.
face antigen by the inhibitor Lutheran In(Lu) gene. J. Clin. Invest. 71: Karamatic Crew, V., Burton, N., Kagan, A. et al. (2004). CD151, the first
1878–1886. member of the tetraspanin (TM4) superfamily detected on erythro-
Telen, M.J., Udani, M., Washington, M.K. et al. (1996). A blood group- cytes, is essential for the correct assembly of human basement mem-
related polymorphism of CD44 abolishes a hyaluronan-binding con- branes in kidney and skin. Blood 104: 2217–2223.
sensus sequence without preventing hyaluronan binding. J. Biol. Ogasawara, K., Osabe, T., Suzuki, Y. et al. (2015). A new ABCG2 null
Chem. 271: 7147–7153. allele with a 27-kb deletion including the promoter region causing
Udani, M., Zen, Q., Cottman, M. et al. (1998). Basal cell adhesion the Jr(a-) phenotype. Transfusion 55: 1467–1471.
molecule/Lutheran protein: the receptor critical for sickle cell adhe- Peng, J., Redman, C.M., Wu, X. et al. (2007). Insights into extensive dele-
sion to laminin. J. Clin. Invest. 101: 2550–2558. tions around the XK locus associated with McLeod phenotype and
Verfaillie, C.M., Benis, A., Iida, J. et al. (1994). Adhesion of commit- characterization of two novel cases. Gene 392: 142–150.
ted human hematopoietic progenitors to synthetic peptides from Tournamille, C., Filipe, A., Badaut, C. et al. (2005). Fine mapping of the
the C-terminal heparin-binding domain of fibronectin: cooperation Duffy antigen binding site for the Plasmodium vivax Duffy-binding
between the integrin alpha 4 beta 1 and the CD44 adhesion receptor. protein. Mol. Biochem. Parasitol. 144: 100–103.
Blood 84: 1802–1811. Zhang, W., Zitron, E., Homme, M. et al. (2007). Aquaporin-1 channel
Wautier, M.P., El Nemer, W., Gane, P. et al. (2007). Increased adhesion function is positively regulated by protein kinase C. J. Biol. Chem. 282:
to endothelial cells of erythrocytes from patients with polycythemia 20933–20940.
Chapter 22 Autoimmune hematological
disorders
Drew Provan1 & John W. Semple2
1
Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
2
Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden

Introduction, 297 Immune thrombocytopenia as a model of autoimmune blood disease, 308


The immune system, 297 Targeted versus untargeted therapies for autoimmune disease, 311
The spectrum of autoimmune diseases, 304 Novel therapies for the treatment of other autoimmune diseases, 314
Role of genetic factors, 304 Conclusions, 315
Mouse models of autoimmune disease, 305 Further reading, 315
Human studies, 306

autoantigen-containing tissues, which may result in disease.


Introduction Factors that play a role in this process include immune dys-
regulation, genetic factors, and environmental factors as trig-
Autoimmune diseases are disorders where antibodies or cells
gering events. We discuss all of these after briefly reviewing
react against self antigens to cause disease, at which point an
the structure and function of the immune system.
adaptive immune response is mounted against the self anti-
gen or antigens. This results in clearance of the antigen from
the body. The normal adaptive response results in complete
removal of the non-self antigen, whereas in autoimmune The immune system
disease there is incomplete clearance of the antigen, which
leads to perpetuation of the immune response. Autoimmune The immune system comprises cells and molecules whose
disorders occur in about 5% of the population, although main role is defense against invading pathogens. The two
many individuals have no symptoms. In all, there are more principal components are the innate immune system, com-
than 70 different disorders, most of which are uncommon, prising skin, mucous membranes, neutrophils, macrophages/
apart from rheumatoid disease and autoimmune thyroiditis. dendritic cells that serve as antigen-presenting cells (APCs),
Autoimmune diseases are clinical syndromes mediated by and other scavenging cells, in addition to the complement
activation of T and B lymphocytes, in the absence of infection system and natural killer (NK) cells; and the adaptive
or other discernible cause. Until recently, although we could immune system, which involves exclusively B and T lympho-
describe the pathological features of autoimmune disease, we cytes (Figure 22.1). The B cells are responsible for differenti-
had little idea as to their actual cause. Through the develop- ating into plasma cells and secreting antibody, aided by T cells
ment of animal models and the identification of target genes, that can also act as effector cells. Key features of the adaptive
we have gained considerable insight into the pathogenetic system include antigen receptor diversity, antigen specificity,
basis of these complex diseases. Autoreactive cells may affect and immunological memory. This is in sharp contrast to the
virtually any body tissue, including blood, and blood disor- innate system, which lacks these features.
ders in which autoantibodies are found include cytopenias
such as autoimmune hemolytic anemia, immune thrombo-
The innate immune system
cytopenia (ITP), and autoimmune neutropenia, in addition
to coagulation disorders such as acquired hemophilia. Despite varied challenges by many antigens, the innate sys-
Although autoimmune diseases are clinically and patho- tem lacks the ability to develop immunological memory;
logically diverse, the common end result is damage to however, this system’s responses remain constitutive and the
same throughout life. In evolutionary terms, the innate sys-
tem probably developed before the adaptive system. The
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. innate immune system is composed of physical, chemical,
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. and cellular components that act together to mediate the

297
298 Molecular Hematology

Microbes
Innate immunity Adaptive immunity

Epithelial B lymphocytes Antibodies


barriers

Phagocytes Effector T cells


T lymphocytes

NK cells

0 6 12 1 3 5
Hours Days
Time after infection
Fig. 22.1 Innate and adaptive immune systems. The innate system comprises physical barriers (e.g. skin) along with scavenger cells, while the
adaptive system comprises B and T lymphocytes. Temporarily, the innate system is the immediate first line of defense, but lacks specificity. The
adaptive system comes into play later and possesses immunological specificity and memory. NK, natural killer.
Source: Abbas A.K., Lichtman A.H., Pober J.S. (2000). Cellular and Molecular Immunology, 4th edn. © 2000. Reproduced with permission of
Elsevier.

first line of defense against invading microorganisms. These (IFN)-α, and other stimuli. Dendritic cells are professional
components are intimately linked with inflammatory pro- APCs that are able to migrate to lymph nodes, process
cesses and ultimately lead to the removal of most of the infec- antigens, and present them to T cells in conjunction with
tious organisms encountered by a host. The innate immune major histocompatibility complex (MHC) molecules, of
response activates quickly (within seconds) after exposure which there are two classes, class I and class II.
of foreign infectious agents and is antigen non-specific, in
that there is no specific memory associated with the immu-
nity. These characteristics distinguish the innate immune Complement
system from the adaptive immune response, which is medi- Because of limitations of space, the role of complement is
ated exclusively by B and T cells, is slower to activate, and is only very briefly discussed here. Following infection, cells of
exquisitely antigen specific, generating memory with subse- the innate immune system migrate toward the affected site.
quent exposure to the stimulating antigen. Complement component C3b coats the pathogen surface,
APCs are key players of innate immunity and physically whereas the molecules C3a, C4a, and C5a act as neutrophil
link innate and adaptive immune responses by presenting chemoattractants, which can then trigger mast cells to release
antigens to T cells. APCs additionally possess surface histamine. This enhances vasodilation and permeability and
receptors for antibody (immunoglobulin) and complement. thus increased blood flow and vessel permeability, allowing
Microorganisms opsonized by antibody and/or complement neutrophils to pass through the blood vessel walls more eas-
molecules are recognized by specific receptors (e.g. Fc ily, an essential requirement for an effective innate response.
receptors) on phagocytes (APC) and are phagocytosed and The complement system is a part of the innate immune
broken down within the interior of the APCs. Within cells system and has a major function in enhancing the ability of
such as neutrophils, killing and digestion of the pathogen antibodies and phagocytes to clear infectious agent and dam-
involve the generation of superoxide and hydroxyl radicals, aged cells. In addition, complement promotes inflammation
nitric oxide, and proteolytic enzymes. In addition to the and initiates the development of the membrane attack com-
removal of pathogens, the innate system also plays a role in plex on the cell membrane, which leads to pore formation and
the removal of dead cells and remodeling of tissue during osmotic lysis of the cell. The complement system consists of
healing. Cells undergoing programmed cell death (apopto- a number of liver-derived small proteins which are inactive
sis) express molecules such as phosphatidylserine on their zymogens (inactive proteases) that circulate in the plasma.
surface, targeting their removal. Dendritic cells also play a Over 30 proteins and protein fragments make up the com-
key role in innate immunity, and activation of dendritic cells plement system and they account for approximately 10% of
occurs following exposure to heat-shock proteins, interferon the globulin fraction of blood (Figure 22.2).
COMPLEMENT SYSTEM
In order to avoid excessive spontaneous amplification of the complement system, Decay
the body has some regulatory mechanisms in place (gray diamonds) Accelerating
Factor (DAF)
Spontaneous Acts as an
Spontaneous cleavage of anaphylatoxin
Factor B Factor D decay thioester bond
(free in plasma) (free in plasma) C3a Action of C3 (purple)
(smaller)
Increases
vascular
permeability
Alternative pathway (red) Factor B binds C3b
[preferentially to C3b bound to Activation of C3 found
Factor D ‘Complement fixation’ ‘Opsonisation’
cellular membrane of pathogen C3bBb freely in blood plasma
cleaves Factor B Binds firmly by covalent Tags pathogen for
(during infection) or C3b convertase by proteolytic
bound to C3b bonding to reactive phagocytosis as
bound to collagen or basement cleavage
membrane (during injury)] molecules on pathogen phagocytes have C3b
(usually carbohydrates) receptors
C3b
(larger)

C3bBbC3b
(C5 convertase)
Pathogen Lectin pathway (green)
surface presents
mannose Acts as an
Binding of MBL to C5a
anaphylatoxin
mannose on
surface of pathogen C4bC2a Cleaves
Mannose C-5
convertase
Binding Loctin
Acute Phase (MBL) Common pathway for classical and
Response C5b
leading lectin systems (blue)
to increase in
production of
acute phase C4b Opsonisation
proteins by Initiates Hydrophobic
up to x 1000 Membrane regions of
CRP binds
C-Reactive phosphocholine CRP then binds Attack Complex C6,7 and 8
protein component of C1 present in (MAC) become
C2a
(CRP) lipopolysaccharides in plasma formation exposed and
Proteolytic by binding bind to cell
cell wall of pathogen cleavage of with C6, membrane
C4 and C2 C7 and C8 of target cell
Classical pathway (orange) C2b

Immunoglobins C1 activated C9 is
from the adaptive C4a Acts as an recruited and Leakage of
immune response anaphylatoxin polymerises cytoplasm
(at a later stage) on the C5b678 leading
complex to to death
form a large of target
pore in the cell
Membrane Attack membrane
Antibody binding to Complex
target antigen on
pathogen surface
CD59 found in
human cell
membrane
Alternative pathway acts first. Classical and Lectin pathways rely on the activation of Acute Phase Resonse and hence take longer.

Fig. 22.2 The complement system.


Source: https://commons.wikimedia.org/wiki/File:Complement system.jpg#filelinks
Autoimmune hematological disorders 299
300 Molecular Hematology

The adaptive immune system T cells, whereas class I molecules present peptide antigens to
CD8+ T cells.
Two requirements of an effective immune system are (i) the
NK cells and others such as neutrophils have recep-
ability to recognize millions of potential antigens; and (ii)
tors for the immunoglobulin Fc region and are responsible
the prevention of self-reacting lymphocytes from causing
for antibody-dependent cellular cytotoxicity following FcγR
tissue damage. The former is achieved through irreversible
linkage of NK cells and antibody-opsonized targets. In addi-
somatic recombination of immunoglobulin and T-cell recep-
tion, NK cells can effect killing using killer-activating recep-
tor (TCR) genes, generating many millions of different anti-
tors, which recognize specific molecules on nucleated cells.
body and TCR molecules.
An inhibitory molecule (killer inhibitory) recognizes MHC
class I on nucleated cells, preventing killing, but if MHC
B and T cells possess antigen receptors class I is lost (e.g. during infection of the cell by virus or after
on their surface malignant transformation), the nucleated cell is recognized
as being abnormal and is therefore targeted for destruction.
The antigen receptor of the B cell is an immunoglobulin and
that of the T cell is the TCR. These molecules are expressed
on their respective cell surfaces and interact with antigen, Soluble molecules: cytokines orchestrate
either as native (immunoglobulin) or processed (TCR) the immune response
antigens. The TCR is a transmembrane protein and consists The innate system relies on a complex network of soluble
of a heterodimer of either αβ or γδ subunits. TCRs, like molecules, such as cytokines and complement components,
immunoglobulins, contain hypervariable regions, and in which coordinate the entire immune response. We discuss
evolutionary terms the two receptors are probably related. these briefly here, since they are implicated in the pathogen-
Unlike TCRs, antibody molecules are both expressed on the esis of autoimmune disease.
B-cell surface and secreted into body fluids. One feature that Cytokines are mediators secreted by cells that influence the
both receptors share is the ability to generate enormous diver- behavior of other cells. Most cytokines are soluble, apart from
sity through recombination of germline variable (V), diver- interleukin (IL)-1 and tumor necrosis factor (TNF)-α, which
sity (D), and joining (J) region segments, in addition to ran- also have membrane-bound forms. Cytokines are small pro-
dom mutations within the rearranged genes, although only tein molecules of around 15–25 kDa whose actions include
immunoglobulins undergo somatic mutation. Immunoglob- promotion of cell growth, cellular communication, inflam-
ulin molecules possess two key regions: the hypervariable mation, immunity, and repair of tissues. These molecules
region (antigen binding) and the Fc portion at the C-terminal are responsible for the regulation and orchestration of the
end which, as already outlined, is recognized by the Fc recep- entire immune response. Their effects are short-lived and
tor (e.g. FcγR) on macrophages. Antibodies may be of the their actions are largely local. In order to exert their effect,
immunoglobulin (Ig)G, IgA, IgM, IgD, or IgE class, with cytokines interact with specific receptors and promote signal
subclasses within some of the groups (e.g. there are four IgG transduction. Their main routes of action are via the Janus
subclasses and two IgA subclasses). Having such enormous kinase (JAK)/STAT and Ras/MAP pathways. The cytokine
diversity ensures that there is an antibody for almost every profile may be pro-inflammatory or anti-inflammatory and
potential antigen encountered, but the downside of this the cytokine balance will dictate whether a helper T-cell
extreme diversity is that antibodies are generated that can clone engages and differentiates into a Th1 (e.g. interferon-
recognize self antigens (autoantibodies), and it is likely that in mediated) or a Th2 (e.g. IL-4- mediated) response. In general,
normal healthy subjects autoantibodies are generated against Th1 responses are effective against intracellular pathogens
a wide variety of antigenic targets. Since autoimmune disease and Th2 responses aid B cells and type I hypersensitivity
is not common, there must exist a mechanism for removing reactions.
self-reacting antibodies. In effect, an immunological lack of
responsiveness or tolerance must exist, whereby self-reactive
cells are prevented from causing damage. Recent research has Cytokine profiles of Th1 and Th2 responses
shown this to be the case. APCs, and in particular dendritic cells, are responsible for
T-cell differentiation toward the Th1 or Th2 phenotype; the
cytokines interferon and IL-12 play key roles in the Th1
The major histocompatibility complex
response, whereas IL-4 mediates Th2 responses (Table 22.1).
Class I MHC molecules comprise human leukocyte antigen Since cytokines play such a key role in orchestrating an
(HLA)-A, -B, and -C, and class II molecules consist of HLA- effective immune response, it is likely that dysregulation
DP, -DQ, and -DR. Class II molecules are responsible for the of cytokine levels may induce an autoimmune response in
presentation of peptide antigens to the TCR on CD4+ helper some disorders. This has been shown to be the case in
Autoimmune hematological disorders 301

experimental models and also in human disease. For exam-


Table 22.1 Cytokine profiles of Th1 and Th2 responses
ple, transfection of the gene for IFN-γ on the insulin pro-
Th1 response Th2 response moter has been shown to induce inflammation within the
(activates macrophages) (deactivates macrophages) pancreas, with aberrant expression of MHC class II and
the development of diabetes. In addition, proinflammatory
IL-2 – cytokines, such as IL-12, TNF, and IFN-γ, can induce organ-
IL-3 – specific autoimmunity.
– IL-4
– IL-5
T cells
– IL-6
– IL-10 T cells develop within the thymus (Figure 22.3) and T cells
– IL-13 bearing αβ TCRs recognize processed antigen presented to
TNF-α – them by APCs, including dendritic cells. Within the thymus,
TNF-β TNF-β T cells are selected in order to prevent autoreactivity; that is
IFN-γ –
to say, mechanisms exist whereby T cells are prevented from
GM-CSF GM-CSF
reacting with self antigens. Positive selection occurs when T
These are the principal cytokines involved in generation of Th1 and
cells express TCRs that are able to interact with MHC com-
Th2 responses. Imbalance in Th1 or Th2 cytokines may play a role plexes on thymic cortex epithelial cells. The effect of positive
in the development of autoimmune disease, allergy, and other dis- selection is to prevent apoptosis. T cells expressing TCRs with
orders. high or low affinity for self molecules are negatively selected
and undergo apoptosis. The end result is that the thymus
releases only those T cells whose TCRs have a medium reac-
tivity against peptide antigens, and are thus are quiescent
until they encounter their specific peptide.

Cortex Medulla
MHC Dendritic cell
CD8 or macrophage
Cortical
epithelial cell
T cell
T-cell
receptor T-cell MHC
receptor
CD4
Medulla Positive CD8 or CD4
T cell selection
Cortex T cell
Positive
CD8 and CD4 Apoptosis selection
Negative
selection CD8 or CD4
T cell
T cell

Surviving
Apoptosis cells leave the
thymus

Thymus gland
Fig. 22.3 T-cell selection within the thymus. T cells undergo positive and negative selection. CD4+ CD8+ T cells interact with major
histocompatibility complex (MHC)–peptide complexes. Depending on the strength of the interaction, the T cells either undergo apoptosis (the
majority) or survive and leave the thymus (the minority).
Source: Delves P.J., Roitt I.M. (2000). The immune system. First of two parts. New England Journal of Medicine 343: 37–49. Reproduced with
permission of New England Journal of Medicine.
302 Molecular Hematology

Immunological tolerance prevents damage processed antigen. In general, antigens in blood are taken to
to self antigens the spleen and tissue antigen is taken to the lymph nodes.
Antigen is then processed and presented on MHC molecules
Tolerance defines the body’s ability to recognize, but not react
by two major routes: (i) the antigen may be produced within
with, self antigens while retaining the ability to respond to
the cell itself, for example viral antigens may be expressed
non-self antigens. This process involves the negative selec-
within cells and complexed with MHC class I molecules; or
tion of T cells, as already outlined. In addition, the process
(ii) APCs may take up antigen exogenously and then phago-
involves the control of autoreactive B cells. The process of
cytose, process, and express the antigen on MHC class II
selecting T cells and B cells in the thymus and bone marrow
molecules.
respectively is known as central tolerance. For autoreactive
lymphocytes that escape into the periphery, there are addi-
tional peripheral tolerance mechanisms to provide a safety net Recognition of antigen by T cells
for unwanted autoreactivity.
Antigen recognition differs between CD4+ and CD8+ T cells.
For example, CD4+ T cells can only recognize peptide anti-
T-cell tolerance: central mechanisms gens on class II molecules, whereas CD8+ T cells recognize
antigen in association with MHC class I molecules. In this
Immature T cells from the bone marrow migrate to the thy- way the MHC dictates the type of response generated.
mus, where they complete their development. The T cells
within the thymus interact with MHC molecules in low- or
high-affinity interactions. If the TCRs have low affinity for TCR signaling
the peptide (e.g. self peptide), they receive apoptotic signals TCR molecules are found on the T-cell surface complexed
and die within the thymus. T cells participating in high- with CD3 molecules. CD3 transmits signals intracellularly,
affinity interactions have a similar fate, and it is only when the with tyrosine phosphorylation of residues in the CD3 cyto-
interaction is of intermediate affinity that the T cells survive plasmic tail. This transmits signals to the nucleus, resulting
and migrate to the periphery, a process termed positive selec- in T-cell proliferation. Co-receptor molecules on T cells are
tion. In general, positive selection occurs when CD4+ CD8+ important, since they play a major role in T-cell activation
T cells interact with TCR–MHC–peptide complexes. For following engagement of the TCR. The generation of a
most T cells the interaction is of low avidity and the T cells specific immune response to a pathogen requires the recog-
die before leaving the thymus. A minority of CD4+ CD8+ nition of the foreign agent by specialized cells in the body,
T cells have intermediate avidity reactions and hence these presentation of the foreign antigen to T cells, and orchestra-
cells survive, after which they mature into either CD4+ CD8− tion of the subsequent immune response. T cells must receive
or CD4− CD8+ cells. CD4+ T cells are the main effectors of two signals from the APCs for a complete immune response
autoimmune disease. to occur. The first signal involves the presentation of antigens
in the context of the MHC on the surface of the APC to
T-cell tolerance: peripheral mechanisms the TCR complex on the T cells (antigen-specific signal 1).
This interaction provides the specificity of the immune
From studies of animal models, it is apparent that self- response. This signal alone is not sufficient to trigger an
reactive lymphocytes are present peripherally, having optimal immune response and a second signal is required.
escaped central tolerance checkpoints. Furthermore, some The second, or costimulatory, signal involves the interaction
form of immunological ignorance is invoked whereby such of B7 on the APC with CD28 receptors on the T cells. The
circulating autoreactive T cells fail to respond to the specific interaction of B7 with CD28 is required for the T cells to
self antigen. The mechanisms for such ignorance are not proliferate, produce cytokines, and provide help for the
fully characterized, but may involve low levels of circulating induction of antibody- and cell-mediated immune responses
antigen (i.e. below a critical threshold), or the antigen may directed at the original antigen. The B7-induced CD28 signal
be in a separate compartment from the autoreactive cells is delivered to the T-cell nucleus via several intermediate
(e.g. the blood–brain barrier), or there may be an absence of steps, and results in the direct induction of the IL-2 gene and
the costimulatory signals required for T-cell costimulation the receptor for this cytokine. T cells stimulated via their
and activation. TCR (antigen-specific signal) and the costimulatory signal
proliferate, produce many cytokines, and direct the specific
immune response against the presented antigen.
T-cell activation: role of costimulatory molecules
The principal co-receptors on APCs are CD80 (B7-1),
Antigen reaches the lymphoid tissue via the lymphatic system CD86 (B7-2), and CD40, binding respectively to CD28, cyto-
or, in some cases, within dendritic cells that have ingested and toxic T-lymphocyte antigen (CTLA)-4, and CD40 ligand on
Autoimmune hematological disorders 303

Antigen specific
CD4+ T cell recognition APC (DC or B cell)

CD4

1 TCR MHC II
Fig. 22.4 Interaction between antigen-presenting Protection from
cells (APCs; e.g. dendritic cells or B cells) and CD4+ apoptosis
T cells. The first signal to the T cell is via major Proliferation
Antibody
histocompatibility complex (MHC) and T-cell receptor IL-2 production CD40L CD40
production
(TCR). This signal is not sufficient for proliferation and
IL-2 production and a second signal, such as interaction 2 APC function
CD80
of B7-1 or B7-2 with CD28, is required. If no second CD28
CD86
signal is received, the T cells undergo apoptosis.
Courtesy of Dr Masataka Kuwana, Institute for Costimulation
Advanced Medical Research, Keio University School of
CD152 (CTLA-4) CD80 T cell anergy
Medicine, Tokyo.

T cells. Dendritic cells express large quantities of both B7 might otherwise cause. We now know that low-level autore-
and CD40, are professional APCs, and are the most effective activity is found in all healthy individuals and that autoanti-
stimulators of naı̈ve T cells. Following activation, the T cells gens play a role in generating our normal immune reper-
undergo clonal expansion, generating effector cells that move toire. Self and non-self antigens have few differences and
toward the site of inflammation after leaving the lymphoid
tissue (Figure 22.4).
Generation of immune repertoires

Mechanisms for controlling autoreactive T cells Bone marrow


Thymus
Harmful autoreactive T cells may be eliminated by several
mechanisms, including apoptosis, anergy, inhibition, clonal Central tolerance
deletion, and suppression. Anergy refers to T cells that do not Anti-self lymphocytes deleted by apoptosis
produce IL-2 after their encounter with antigen. Such cells (negative selection)
are not activated and may produce IL-10, which further sup-
presses T-cell activation. Inhibition of T cells is mediated by
Leakage of anti-self lymphocytes to periphery
CTLA-4, also known as CD152. This molecule binds to the
B7 family of receptors (B7-1 and B7-2) on APCs, including
Controlled by
B cells and dendritic cells, with higher affinity than to CD28,
a counter-receptor for B7. CTLA-4 mediates suppression of
the T cell. Peripheral tolerance

Loss of immunological tolerance leads to autoimmunity Wrong environment Tolerance fails Wrong genes
In order for autoimmune disease to occur, there must be loss
of tolerance to self antigens. This occurs despite central and Autoimmune disease
peripheral T-cell tolerance controls. Possible mechanisms
leading to loss of tolerance include variability in intrathymic
Global (present) Therapies Selective (new)
T-cell deletion and activation of harmful autoreactive T cells
in the periphery, for example by infection. The latter has been Fig. 22.5 Autoimmunity is multifactorial. Central tolerance is
achieved within the bone marrow or thymus, but some self-reacting
postulated to cause type 1 diabetes (insulin-dependent dia-
cells leak out into the periphery, where peripheral tolerance
betes mellitus) in some individuals (Figure 22.5).
checkpoints exist. Self-reactive lymphocytes escaping central and
peripheral tolerance may cause autoimmune disease in predisposed
Beneficial effects of autoimmunity individuals (the “wrong” genes) when they encounter a specific
trigger (the “wrong” environment).
The concept of clonal deletion has been central to immunol- Source: Mackay I.R. (2000). Science, medicine, and the future:
ogy for many years. This process deletes self-reactive lym- tolerance and autoimmunity. British Medical Journal 321: 93–96.
phocytes in order to prevent the harmful effects that these Reproduced with permission of British Medical Journal.
304 Molecular Hematology

lymphocytes have probably not evolved in order to distin- monozygotic than dizygotic twins: in monozygotic twins the
guish between the two; their role is rather to respond to anti- chance of both twins having autoimmune disease is higher
gen under specific circumstances, for example as directed by than in dizygotic twins. Examples of diseases with high con-
the cytokine network. In addition, autoreactive cells are also cordance rates between monozygotic twins include type 1
responsible for the remodeling of tissues, wound healing, and diabetes, rheumatoid arthritis, and SLE; the concordance
other physiological processes. Autoimmunity therefore dif- rate for monozygotic twins is around 20% and for dizygotic
fers from autoimmune disease. In fact, transient autoimmune twins it is less than 5%. Some disorders are due to mutations
attack of self antigens has been shown to occur when there is in a single gene, for example autoimmune polyglandular
tissue damage, but this type of autoimmune response is gen- endocrinopathy with candidiasis and ectodermal dyspla-
erally not sustained. sia (APECED) and the autoimmune lymphoproliferative
disorder. In APECED there is a mutation in the gene for
an autoimmune regulator protein that is active within the
The spectrum of autoimmune diseases thymus. The mutation leads to autoimmunity and immune
deficiency. In the autoimmune lymphoproliferative disorder,
For clinical convenience, autoimmune diseases are subdi- the autoreactivity is due to an inability to induce apoptosis
vided arbitrarily into those that are organ specific, such as of activated lymphocytes following encounter with antigen.
Hashimoto thyroiditis, and those that are non-organ specific The disorder is autosomal dominant and involves Fas or its
(or systemic), such as systemic lupus erythematosus (SLE). receptor, both of which are involved in the downregulation of
Many diseases lie between these two extremes. Conditions activated cells following antigen exposure.
in which there are circulating immune complexes tend to be However, most autoimmune diseases are not caused by
systemic, while conditions associated with autoantibodies or single gene mutations but rather are multigenic, several
autoreactive T-cell responses are organ specific. In general, genes acting together to cause the disease phenotype. Type 1
however, this classification tells us nothing about the causes diabetes is a prime example of a disease in which multiple
of disease, and it may be better to classify diseases into those genes are implicated. For example, 95% of Caucasians with
in which there is disturbed selection, regulation, or death of type 1 diabetes possess HLA-DR3, HLA-DR4, or both (only
T and B cells (e.g. mediated by Fas or Fas receptor abnor- found in 40% of normal subjects), and 40–50% of patients
malities), and secondly those in which there is abnormal are heterozygous for HLA-DR3/DR4 (found in 5% of normal
expression of an antigen, for example the demyelination syn- subjects). Other non-HLA genes are also implicated in type 1
drome that follows gut infection with the bacterium Campy- diabetes, including IL-2 polymorphism and another region
lobacter jejuni. This method of classification may help guide that maps close to CTLA-4. Surprisingly, some of these
treatment. genetic polymorphisms occur in normal individuals who
Thus, the factors that may play a role in the development have normal immune function with no evidence of autoim-
of autoimmune disease include: mune disease. So it would appear that there is a need for
r failure of tolerance to self antigens; specific genetic polymorphisms in conjunction with other
r infection; susceptibility polymorphisms. The MHC is one candidate for
r tissue injury; this role.
r abnormalities of APCs; Several autoimmune diseases described to date have link-
r imbalance between proinflammatory and anti- age with specific MHC class I or II polymorphisms, but again
inflammatory cytokines; these do not produce the disease phenotype in isolation;
r genetic factors; rather, they require the presence of polymorphisms within
r variable effector mechanisms, for example production other genes, such as those for cytokines (e.g. TNF-α). For
of immune complexes, autoantibodies, or autoreactive diseases such as ankylosing spondylitis, type 1 diabetes, and
T cells. rheumatoid disease, certain HLA subtypes induce disease
susceptibility. The link between MHC genotype and disease
susceptibility is logical given the fact that autoimmune
Role of genetic factors diseases involve autoreactive T cells. The ability of a T cell to
respond to a particular antigen depends largely on the MHC,
It has long been recognized that genetic factors play a key role since the MHC determines how the antigen is presented to
in autoimmune disease, the strongest correlation being with the autoreactive T cells. In addition, the MHC has a powerful
MHC genes, especially MHC class II. Support for a genetic influence on determining the body’s T-cell repertoire. Other
basis comes from a variety of observations. For example, HLA alleles appear to be able to offer protection from
autoimmune diseases often cluster within families, and twin autoimmune disease. For example, when the disease-causing
studies show that there is much higher concordance between allele HLA-DQB1*0301 or 0302 is present together with
Autoimmune hematological disorders 305

HLA-DQB1*0602, the latter appears to offer protection from other antibodies, possess two distinct functional compo-
disease. The mechanism underlying such protection is not nents: a Fab end, which binds to the targeted antigen, and an
known. Some HLA alleles cause disease only within specific Fc end, which is recognized by scavenger cells (e.g. macro-
populations. For example, HLA-DRB1*0401 and *0402 are phages) in the spleen, liver, and bone marrow. The recep-
associated with rheumatoid disease in Europeans, but not in tors on the macrophages that sense the presence of antibodies
black or Hispanic individuals. Again, the mechanism here are termed Fc receptors. For example, how well the Fc recep-
is not clearly understood. tors recognize and bind antibody attached to platelets deter-
mines, in part, how aggressively the platelets are destroyed
in ITP. Polymorphisms of the FcγRIIA (FCGR2A) gene have
Some autoimmune diseases have strong
also been implicated in heparin-induced thrombocytopenia,
association with cytokine gene
SLE, and childhood recurrent bacterial infections.
polymorphisms
Intravenous immunoglobulin (IVIg) is effective in ITP,
Since cytokines, through interaction with their ligands, although its mechanisms of action are not fully understood.
orchestrate the immune response, it is possible that dysreg- However, one of the actions of IVIg is to attach to Fc recep-
ulation of cytokines may play a role in autoimmune disease. tors on the macrophages, thereby blocking the receptor.
Indeed, many single nucleotide polymorphisms (SNPs) have Once the receptor is blocked it cannot bind antibody bound
been described within cytokine or cytokine receptor genes, to platelets, and the antibody-coated platelets are spared
and such SNPs may alter cytokine structure or their expres- destruction by the immune system.
sion. This may result in overexpression or underexpression. Recent reports have highlighted genetic variations in FcγR
In the blood disorder ITP and other autoimmune diseases, genes which alter their ability to recognize and bind antibody.
abnormal cytokine profiles have been described. In chronic The genetics of FcγR genes is complex, but essentially there
ITP, IL-2, IFN-γ, and IL-10 levels are increased, suggesting a are three major types: FcγRIA, FcγRIIA, and FcγRIIIA. A
Th1 activation profile (Table 22.2). recent study of FcγRIIA suggests that a polymorphism in the
Not only do cytokine gene SNPs predispose to disease, but FCGR2A gene may be responsible for causing refractory ITP
data from animal models such as the rheumatoid rat indicate (ITP that responds poorly to treatment). The polymorphism
that specific polymorphisms may determine disease chronic- influences the efficiency of the receptor to bind with antibody
ity and severity. In addition, there are data to suggest that molecules. A similar polymorphism in the FCGR2A gene has
specific genetic polymorphisms may correlate with responses been shown to alter the function of the receptor and may pre-
to specific treatments, but it is probably too early to draw firm dispose individuals to autoimmune disease such as ITP, and
conclusions from such studies. in those individuals developing ITP the disease is more likely
to be chronic. Human FCGR2A-transgenic mice have been
shown to have a more severe form of ITP than normal mice,
Antibody Fc receptor gene polymorphisms which provides further evidence that FcγRIIA contributes to
For autoimmune blood diseases in which autoantibody- platelet destruction.
opsonized cells are sequestered and destroyed within the
reticuloendothelial system, there are data from a number of
studies showing that polymorphisms within the Fcγ recep- Mouse models of autoimmune disease
tors may influence disease. Antiplatelet antibodies, as with
These have provided much insight into autoimmune disease
and to date around 25 genes have been shown to play a role
Table 22.2 Autoimmune diseases in which strong associations with
in the development of disease, either when the genes are
cytokine single nucleotide polymorphisms have been described deleted or when they are overexpressed. The genes encode
cytokines or their receptors, costimulatory molecules, or pro-
Implicated cytokine teins involved in apoptotic pathways. Whether a mutation
Disease polymorphism within one of these critical genes induces disease depends
on the genetic background of the animal, which would tend
Juvenile chronic arthritis IL-6 to imply that other genes can influence the phenotype. One
Myasthenia and multiple sclerosis TGF-β
other point worth noting is that mutations within a gene may
Rheumatoid disease IL-10
be involved in more than one clinical disorder.
Associations have now been reported for many autoimmune
With respect to hematological autoimmune diseases, there
disorders and a full database is provided at www.bris.ac.uk/ have been several animal models of ITP, including the
pathandmicro/services/GAI/cytokine4.htm. secondary autoimmune model, where thrombocytopenia is
secondary to lupus nephritis; the passive transfer model,
306 Molecular Hematology

where continuous injection of platelet-specific antibodies is concordance in monozygotic twins. However, the concor-
required to maintain a steady state of thrombocytopenia; and dance rate for the second twin if the first twin has autoim-
the viral acute ITP model, where thrombocytopenia develops mune disease is less than 50%, which suggests that addi-
following a viral infection. Although these models have been tional, possibly external factors are required for autoimmune
important in studying treatments such as IVIg, they are not disease. Possible external influences might include infec-
ideal for pathophysiological studies, since either the induced tion, and evidence for a role of infection in the develop-
thrombocytopenia is secondary, or there is no endogenous ment of autoimmune disease is provided by disorders such
antibody production. Recently, however, a platelet-specific as Lyme disease and its associated arthritis. In this condition,
immune model of ITP was developed using CD61 (GPIIIa) there is cross-reactivity between the pathogen and host tis-
knockout mice. In this active ITP model, immune spleen sue antigens. The self protein targeted is leukocyte function-
cells from immunized CD61 KO mice are transferred into associated antigen (LFA)-1, also called CD11a and CD18.
severe combined immunodeficient (SCID) mice, which then LFA-1 shares determinants with protein antigens of Borrelia
develop both an antibody- and T-cell-mediated ITP. This burgdorferi. Another example is Epstein–Barr virus (EBV),
model has the advantage in that both antibody- and T-cell- which appears to have a role in the development of SLE, from
mediated ITP is endogenously produced and various thera- case–control studies using stored serum from patients with
pies can be tested on both effector systems. With the advent SLE. Whether significant or not, there is one antigenic deter-
of transgenic mice expressing human Duffy and glycophorin minant on EBV that is shared with SLE. However, this evi-
antigens, there are also now murine models of autoimmune dence is fairly circumstantial at present.
hemolytic anemia.
How might infection cause autoimmunity?

Human studies Pathogens may be able to induce autoimmunity through a


variety of mechanisms:
r production of local inflammation;
A number of candidate genes have been studied in human r change in reticuloendothelial function, e.g. phago-
autoimmune disease, including variants of CTLA-4. This
molecule is a natural downregulator of activated T cells. One cytosis;
r production of neoantigens;
polymorphism within this gene causes a reduction in the r molecular mimicry.
inhibitory signal normally induced by the CTLA-4 molecule,
and this polymorphism has been found to be associated with Local inflammation can expose costimulatory molecules
human type 1 diabetes, thyroid disease, and primary biliary on APCs and lead to the breakdown of T-cell anergy and
cirrhosis. the development of autoimmune disease. Tissue injury may
result in the production of neoantigens for which an autoan-
tibody may have specificity. Lastly, in molecular mimicry,
Critical events in the generation
antigens on microorganisms may resemble those on the host
of autoreactivity
tissues, such that antibodies produced against the pathogen
Since autoimmune diseases are multifactorial, there are will cross-react with the host tissue. Examples may include
obviously triggering events within genetically predisposed multiple sclerosis, type 1 diabetes, and childhood acute ITP,
individuals that lead to the development of autoimmune if the antibody produced in a childhood viral infection by
disease. Animal models show that if animals genetically chance cross-reacts with antigen(s) on the platelet surface.
susceptible to autoimmune disease are injected with self anti- Although elegant, there is little apart from circumstantial
gens from genetically identical animals with an appropriate evidence at present to support the existence of molecular
adjuvant (e.g. bacterial), the animal will mount an immune mimicry in humans.
response against the self antigen. In humans, autoimmune
disease usually arises spontaneously, although it is accepted Non-infectious triggers
that there must be triggering events giving rise to the disease
Infection may play a role in autoimmune disease, but what
phenotype. What factors might be responsible for inducing
non-infectious triggers might there be which could induce
autoimmune disease? Several have been proposed, including
autoimmune disease in susceptible individuals? Hormones
infectious and non-infectious triggers.
may play a role, since we know that most autoimmune dis-
orders are commoner in women than men, and in mice with
Environmental factors in the development
SLE the administration of estrogens worsens the disease;
of autoimmune disease
this effect is believed to be due to an alteration of the B-cell
If the development of autoimmune disease were entirely repertoire. Also in SLE, complement deficiency (e.g. C1 or
genetic, then we would expect complete or near-complete C4 components) may worsen the disorder. Haptens such as
Autoimmune hematological disorders 307

drugs can also induce autoimmune disease. For example, are at their highest. The mechanism whereby the hormonal
penicillin acts as a hapten when it binds to the red cell profile of females influences the development of autoimmune
membrane, inducing an autoimmune response that leads to disease is unknown.
autoimmune hemolytic anemia. Complexes formed between
two proteins may trigger disease. For instance, celiac disease Autoimmune disease is complex
is an autoimmune disorder in which transglutaminase and its and multifactorial
substrate gliadin form a complex that induces autoantibody
generation against both gliadin and transglutaminase. From studies of autoimmune disease, it is clear that ITP and
other autoimmune disorders are multifactorial. It is likely
Epitope spread that loss of tolerance to a self antigen alone is insufficient to
generate the autoimmune disorder. Rather, patients prob-
This phenomenon may play a role in some autoimmune
ably require (i) a specific set of genetic determinants (e.g.
diseases and involves the initial generation of autoreactivity,
polymorphisms within MHC, CTLA-4, or other genes); (ii)
with subsequent development of chronic autoimmune
dysregulation of the immune response (involving dendritic
disease. During the process there is a gradual increase in
cells, T or B cells, or all three); and (iii) an environmental
the number of autoantigens targeted by T cells; hence there
trigger. Autoimmune disease arises only when all these
is spread to epitopes beyond the initial triggering epitope
determinants are present in an individual at one particular
(Figure 22.6).
time. This is reinforced by the observation that self-reactive
lymphocytes are commonly found in healthy individuals. For
Hormonal influences
example, siblings of patients who have autoimmune disorders
Many autoimmune diseases are more common in females, are more likely to have autoantibodies themselves, albeit at
particularly in the childbearing years, when hormone levels lower titers than their affected siblings, but no overt evidence

Antibody coated Fcγ Activated macrophage


platelet receptor

Glycoprotein
IIb/IIIa
Autoantibody
Glycoprotein
Ib/IX

CD4
CD154
Anti- CD154
Glycoprotein CD40
glycoprotein CD40
IIb/IIIa IIb/IIIa CD4
B-cell clone 1 T-cell clone 1

Glycoprotein
Ib/IX
Glycoprotein
Anti-
IIb/IIIa
glycoprotein
IIb/IIIa Glycoprotein
Ib/IX

B-cell clone 2 T-cell clone 2


Fig. 22.6 Epitope spread. In immune thrombocytopenia (ITP), for example, platelet glycoproteins are processed and presented to T cells by
antigen-presenting cells (APCs). A variety of different peptides may be presented by APCs, resulting in multiple T-cell clones, each targeting distinct
epitopes on different glycoprotein molecules.
Source: Cines D.B., Blanchette V.S. (2002). Immune thrombocytopenic purpura. New England Journal of Medicine 346: 995–1008. Reproduced by
permission of New England Journal of Medicine.
308 Molecular Hematology

of autoimmune disease per se, perhaps because they have not approximately 40% of patients with chronic ITP have no
been exposed to the environmental trigger required to tip the detectible antibodies, yet are thrombocytopenic. CD8+ T
balance toward autoimmune disease. cells have been linked to the pathogenesis of many autoim-
mune diseases such as type 1 diabetes, and it was shown
How do autoreactive cells induce tissue that ITP patients have a CD8+ T-cell-mediated cytotoxic-
damage in autoimmune disease? ity that induces platelet destruction. This finding was also
shown in a murine model of active ITP. At present, however, it
This is a large topic which is discussed only briefly here. In is unknown whether cell-mediated platelet destruction con-
autoimmune disease, since the autoantigen is not cleared tributes to the severity of disease or the difficulty of treatment
completely from the affected individual, the process tends to in some patients with ITP.
be prolonged, because there is always a supply of autoantigen The etiology of ITP is unknown and the clinical course is
available to keep the process going. Autoantibodies, espe- variable and unpredictable. ITP has an incidence of around
cially IgG and IgM, may attach to cell membrane antigens 60 new cases per million population per year in the USA.
on cells and cause local tissue damage. Where the antigens Childhood ITP is generally seasonal, typically follows a trivial
are soluble, a more systemic disease profile may result. In viral infection or vaccination, is transient in most cases, and
addition to autoantibody-mediated disease, T cells can them- requires no treatment, with spontaneous recovery in 80% of
selves directly cause disease. When autoantibodies attach cases. One proposed mechanism invoked in childhood ITP is
to blood cells, premature destruction of the cells results molecular mimicry, in which the antibody directed against an
within the spleen. Cells involved include red cells, resulting invading pathogen coincidentally cross-reacts with one of the
in autoimmune hemolytic anemia, white cells, causing platelet glycoprotein (GP) epitopes. As discussed earlier, the
neutropenia (any white cell may be involved but neutropenia normal adaptive immune response ceases once the offend-
is commonest), and platelets, inducing thrombocytopenia. ing pathogenic antigen is destroyed, and this might account
Red cell attack by IgG antibodies results in premature red for the acute nature of ITP in childhood. That is to say, once
cell destruction by the macrophages of the reticuloendothe- the pathogen is eradicated, the source of non-self antigen is
lial system, via Fcγ receptors. When the autoantibody is removed and cross-reacting antibody levels fall.
IgM, complement activation also occurs and there may be In the adult form there is usually no obvious antecedent
intravascular lysis, although more commonly the comple- illness and most patients have chronic thrombocytope-
ment cascade does not reach the lytic stage, but generally nia; spontaneous recovery is uncommon. In most cases of
stops at the C3d stage because of the presence of complement adult ITP the platelet glycoprotein (GP) antigen targets are
regulatory proteins. However, even with incomplete comple- GPIIb/IIIa and GPIb/IX.
ment activation, the presence of molecules such as C5a gener-
ates local inflammation and tissue damage through activation
of cytokines. Autoimmune blood cell disorders are discussed Etiology
shortly. It is believed that ITP is most likely due to an inappropriate
Rather than cause direct cellular damage, some autoanti- immune response to an environmental trigger; the nature
bodies may activate or block receptors. For example, Graves of this trigger has not yet been identified. The disorder may
disease is caused by an autoantibody that stimulates the represent an abnormality of APCs, with an increase in the
thyroid-stimulating receptor, leading to elevated levels of numbers of CD4+ and CD8+ cells. It has been increasingly
thyroid hormones. In myasthenia gravis, the autoantibody recognized that T cells play a significant role in autoantibody
blocks acetylcholine receptors, preventing neuromuscular production and recent data suggest that, in some instances,
transmission. Other mechanisms of autoantibody and T-cell T cells may actually mediate platelet and megakaryocyte
damage also occur, but there is insufficient space to discuss (MK) destruction in ITP. Furthermore, two new and excit-
these here. ing possibilities have emerged in ITP research related to
T-cell abnormalities that may prove be key to unraveling
the pathogenesis of the disorder, and may also reveal new
Immune thrombocytopenia as a model therapeutic options for ITP. It appears that patients with ITP
of autoimmune blood disease have functionally reduced numbers of T-regulatory cells that
may relieve T-cell tolerance mechanisms and allow platelet
ITP is usually an acquired disorder in which platelets autoimmunity to proceed, whereas therapeutic removal
are coated (opsonized) with antiplatelet autoantibodies and of B cells in the disorder by therapies such as rituximab
removed prematurely by the reticuloendothelial system, pre- raises platelet counts by actually normalizing these T-cell
dominantly the spleen, leading to a reduced peripheral blood functional deficiencies. Figure 22.7 highlights the cellular
platelet count. Of interest is that it has been estimated that immune abnormalities found in patients with ITP.
Autoimmune hematological disorders 309

Inflammatory
stimuli CD40L
GPllbllla

Platelet M-CSF CD40


A
autoantigens sIg CD20
Activated
macrophage
Autoreactive
IL-1β, IL-1α B cell
CD68 TGF-β
CD40 D

Treg Autoantigen B7.1/7.2


processing
peptides B
IL-2, IL-4, IL-15
CD40 CD40L
IFN-γ, TNF-α,
CTLA-4 GM-CSF
Class II
F CD28
Class I Autoantibody
CD40L
production
C
TCR
sIL 2R
Microparticles Autoreactive
TCR IL2
Th cell
E
sIL2R-IL2

TCR
CD8+T cell Cell-mediated Platelet
NK cell immunity destruction
Class I
Fig. 22.7 Potential stimulatory (solid arrows) and inhibitory (hatched arrows) pathways that may affect the pathogenesis of chronic
immune thrombocytopenia (ITP). This model assumes that a tolerance event has broken down, allowing autoreactive T and B cells to be
present. (a) Platelets are normally taken up by macrophages during senescence and are presumably destroyed intracellularly within lysosomes.
Inflammatory stimuli can activate macrophages that may alter the way they normally process platelet autoantigens, or induce the expression of
inflammatory cytokines (e.g. IL-1) that may support autoimmune responses. (b) In the activated antigen-presenting cells (APC), platelet
autoantigenic peptides can be loaded onto major histocompatibility complex (MHC) class II molecules for presentation to the T-cell receptor (TCR)
of autoreactive Th cells. (c) Once the TCR has been occupied by the MHC class II molecules and platelet autoantigen, it can initiate a series of
molecular costimulatory interactions within the T cell. First, CD40L upregulated on the T-cell surface interacts with CD40 on the APC that
stimulates B7.1 and B7.2 expression. The TCR–MHC interaction also enhances CD28 expression on the Th cells, which then interact with the B7.1,
culminating in a strong costimulation response and activation of the Th cell. (d) Th activation leads to the secretion of Th0/Th1 cytokines (e.g. IL-2,
IL-10, IFN-γ) that effectively drive autoreactive B cells to divide and differentiate into plasma cells and secrete antiplatelet autoantibodies. Also
shown are a number of potential regulatory events that could either enhance or suppress autoimmunity in ITP. For example, macrophage
colony-stimulating factor (M-CSF) can activate macrophages within the spleen to enhance their phagocytosis of platelets. Additionally, IFN-γ and
TNF-α produced by the autoreactive Th cells can feed back on macrophages, enhancing their expression of MHC class II molecules and potentially
aggravate the response. On the other hand, transforming growth factor (TGF)-β (produced by either Th3 cells or platelets) together with the Th
cell’s soluble IL-2 receptor can control the autoimmune response by inhibiting lymphocyte activation. This may also occur during costimulation via
the expression of CTLA-4 on the Th cell. Also shown are the potential interactions of MHC class II- and CD40L-positive platelets with Th and B cells
activation, respectively. These events may be responsible for initiating and/or aggravating autoimmunity in patients with autoimmune
thrombocytopenia. (e) Recent evidence suggests that CD8+ T cells are active and present in patients with ITP who do not demonstrate
autoantibodies. These cytotoxic cells bind to platelets and mediate cytolysis. (f) It appears that many of the defects shown in the figure may be the
result of defective or lack of CD4+ CD25+ T regulatory (Treg) cells due to a breakdown in self-tolerance.
Source: Adapted from Coopamah M.D., Garvey M.B., Freedman J., Semple J.W. (2003). Cellular immune mechanisms in autoimmune
thrombocytopenic purpura: an update. Transfusion Medicine Reviews 17: 69–80. Reproduced with permission of Elsevier.
310 Molecular Hematology

With regard to genetic abnormalities in ITP, most atten- Antibodies and their target antigens
tion has focused on the identification of MHC susceptibil-
Antiplatelet antibodies
ity genes, given their role in determining the nature and
specificity of the adaptive immune response. However, HLA Many patients with ITP show elevated levels of platelet-
association studies in ITP have not produced clear results. associated IgG, which is believed to be the autoantibody, but
A large analysis focusing on HLA-DR4 gene variations in for unknown reasons platelet-associated IgG may be elevated
more than 100 Japanese patients with ITP reported that the in other non-immunological causes of thrombocytopenia.
DRB1*0410 allele was significantly increased in ITP patients The autoantibodies involved in ITP are generally IgG, but
compared with controls. Moreover, this allele was signifi- IgA and IgM autoantibodies have been reported. Opsonized
cantly decreased in patients who showed a good response to platelets are removed prematurely by the reticuloendothe-
prednisolone. MHC may therefore play a role in some cases lial system through an Fc-dependent mechanism, primarily
of ITP, but there are clearly other genes implicated. These within the spleen. However, many patients fail to respond to
include genetic polymorphisms within cytokine and other therapies aimed at inactivation of the reticuloendothelial sys-
immune regulatory genes. tem, suggesting that other mechanisms of platelet destruction
exist.

Clinical features
Antigenic targets
The ITP phenotype is heterogeneous: some patients suffer
Using antigen-specific assays, such as the monoclonal
major bleeding from the outset, while others have few
antibody-specific immobilization of platelet antigens,
problems apart from an increased tendency to bruise. This
platelet-associated IgG, and antigen capture assays, several
may partly be explained by the acquired platelet dysfunction
platelet autoantigens have been characterized. These include
that is seen in some patients with ITP, which in turn may
epitopes on GPIIb/IIIa (αIIIbβ3, the fibrinogen receptor)
be related to the target antigen involved in the autoimmune
and GPIb/IX (the von Willebrand receptor), which appear to
process (this is discussed later; see Antibodies and Their
be the most frequently involved. Less commonly, GPIa/IIa,
Target Antigens). Autoantibodies reacting with GPIIb/IIIa
GPIV, and GPV are involved. Recent reports suggest that
affect platelet aggregation, and anti-GPIb/IX autoantibodies
possibly 80% of autoantibodies are reactive to both GPIIb/
impair platelet adhesion to the subendothelial matrix,
IIIa and GPIb/IX, possibly due to the serum in some
causing unexpectedly severe bleeding for the level of platelet
patients with ITP containing two different IgG antibodies.
count. In contrast, however, to the thrombocytopenia due to
In terms of disease chronicity, GP-specific autoantibodies
marrow infiltration (e.g. leukemias, lymphomas, and other
may be important in the pathogenesis of chronic ITP; from
malignancies) or aplasia, patients with ITP are able to tolerate
available data, GPIIb/IIIa appear to play a major role in the
remarkably low platelet counts and to maintain an adequate
development of chronic ITP in approximately 40% of cases.
quality of life. The degree of bleeding is largely dependent on
the platelet count, and patients with counts below 10 × 109 l−1
(and usually below 5 × 109 l−1 ) are at greatest risk of bleeding, Which epitopes are involved?
including intracranial bleeding.
Previous investigators have looked for autoantigenic epi-
topes on the GPIIb/IIIa molecule using competitive binding
between human autoantibodies and mouse monoclonal anti-
Diagnosis
bodies. In addition, enzyme-cleaved IIb or IIIa fragments and
The diagnosis of ITP remains clinical, and one of exclu- synthesized peptides corresponding to different sequences of
sion. Secondary causes include SLE, lymphoproliferative dis- GPIIIa have been used to localize epitopes on the respective
ease, and HIV infection. Standard investigation includes a glycoprotein.
full blood count (isolated thrombocytopenia), blood film (to Kekomaki et al. have shown that the 33-kDa chymotryp-
ensure no red cell fragments, leukemia, parasitic infections), tic core fragment of IIIa is a frequent target. Fujisawa et al.,
and autoimmune profile (to exclude secondary cause). A using synthetic peptides corresponding to IIIa sequences,
bone marrow examination is often carried out in adults but showed that in 5 of 13 sera from patients with chronic ITP,
not usually in children, and will typically show normal or binding was to residues 721–744 or 742–762, correspond-
increased MK in an otherwise normal marrow. Immunolog- ing to the carboxy terminal of IIIa. Recently, Nieswandt et al.
ical assays have been devised, including platelet-associated have examined the pathogenic effects of IgG monoclonal
IgG or IgM and monoclonal antibody immobilization of antibodies of different IgG subclasses against murine GPIIb/
platelet antigens, but these do not alter the management and IIIa, Ibα, Ib/IX, V, and CD31. Their data suggest that, at
are of debatable value. least in mice, the antigenic specificity of the antiplatelet
Autoimmune hematological disorders 311

antibodies determines the pathogenic effects rather than Chronic refractory ITP
the IgG subclass. They also demonstrated that antibodies
This defines those patients who fail to respond to first-line
against GPIb/IX caused thrombocytopenia through an Fc-
treatment or require unacceptably high doses of corticos-
independent mechanism, while that from autoantibodies
teroids to maintain a safe platelet count. A number of agents
against GPIIb/IIIa involved the Fc system. Further work
have been used as second-line therapy for ITP, including
is clearly needed in order to determine the significance of
high-dose steroids, high-dose IVIg, intravenous anti-D, vinca
all these findings, which may translate into stratification of
alkaloids, danazol, azathioprine, combination chemotherapy,
patients into those in whom Fc receptor blockade or inacti-
and dapsone.
vation is a useful option, and those in whom it is not.
At the T-cell level, it has been demonstrated that T cells
from Japanese patients with chronic ITP could proliferate
in vitro in response to disulfide-reduced GPIIb/IIIa or the Targeted versus untargeted therapies
molecule’s tryptic peptides. This suggested that autoreactive for autoimmune disease
CD4+ Th cells in chronic ITP need to recognize a modified
GPIIb/IIIa molecule, implying that antigen-processing Until now, most of our treatments for autoimmune dis-
mechanisms within recipient APCs may be required to ease have been untargeted and unselective in their modes of
present GPIIb/IIIa autoantigens in the context of self HLA- action. In disorders such as ITP, the therapeutic aim has been
DR molecules. Subsequently, mapping studies using six large to induce global immunosuppression in the hope that, as part
(~200 amino acids) recombinant fragments encoding differ- of this process, the ITP-related component of the immune
ent portions of the GPIIbα and GPIIIa chains showed that system will be suppressed and that this will help reduce the
the T cells from patients with ITP recognized primarily the quantity of autoantibody produced. For antibody-mediated
amino-terminal portion of the two chains (GPIIbα 18–259 autoimmune diseases, what remains unclear is whether the
and GPIIIa 22–262) and that these molecules also stimulated B-cell population that is generating the antiplatelet autoanti-
the production of antiplatelet autoantibodies. Ultimately, the body is the primary problem, or whether events downstream,
GPIIIa molecule has been mapped for CD4+ T-cell specifici- such as those involving antigen presentation or T-cell regu-
ties by using 15-mer peptides of the GPIIIa chain, and this has lation, are disturbed, and simply driving the passive B cells,
revealed several immunodominant peptides spanning the resulting in the autoantibody phenotype.
entire breadth of the molecule. What was particularly intrigu- Now that we have a clearer understanding of the immuno-
ing was that despite a lack of HLA association observed in logical mechanisms involved in autoimmune disease, we
many studies of patients with ITP, some patients apparently have started to develop more targeted therapies. We are now
recognize common elements on the GPIIIa molecule. This developing treatments designed to target T cells, B cells, and
may suggest that either a host (e.g. antigen processing) or other effectors within the immune system. For ITP these
environmental (e.g. infection) factor could be responsible for include Campath-1H and anti-CD20. Although these agents
generating a common autoepitope that is presented to T cells are not entirely specific because they deplete the B-cell com-
across different HLA. If this were true, it would be an effica- partment, they should reduce the quantity of autoantibody
cious way of developing peptide antigen-specific T-cell ther- produced. Other therapies which may be of benefit in ITP
apies for autoantibody production and subsequent platelet are mycophenolate mofetil and anti-CD40 ligand.
destruction in ITP.
Campath-1H
Standard treatment
Campath-1H is a humanized IgG monoclonal antibody that
There is a lack of clinical trial data to help guide treatment, targets the CD52 antigen, present on mature human lym-
and our energies should now be focused on constructing phocytes (T and B cells) and monocytes. Campath-1H is
high-quality randomized trials in order to determine the effective in the treatment of malignant B-cell disorders, espe-
most effective therapy in this disorder. Therapy is seldom cially B-cell chronic lymphocytic leukemia, in which it has
necessary for patients whose platelet counts exceed 30 × been shown to be effective in clearing lymphocytes from both
109 l−1 and in whom there are few spontaneous bleeding blood and bone marrow.
episodes, unless they are undergoing any procedure likely Campath-1H has been used in a variety of autoimmune
to induce blood loss. Standard treatments, including oral diseases, including rheumatoid arthritis, vasculitis, and
prednisolone, IVIg, and splenectomy, will elevate the platelet Wegener granulomatosis. There is ongoing interest in its
count sufficiently in most adults (Figure 22.8). However, use for the treatment of autoimmune hematological disease
some 20–25% of adults with ITP are refractory to first-line that is refractory to first- and second-line therapies. One
therapy. recent study of the use of Campath-1H in autoimmune
312 Molecular Hematology

Corticosteroids,
intravenous immune globulin,
anti-D immune globulin, Splenectomy
danazol, vinca alkaloids Macrophage

Platelet

FCγ
receptor

Platelet
transfusion
Plasmapheresis

Bone marrow

Antibody
against CD20,
intravenous
immune globulin(?)
staphylococcal
protein A(?)

Thrombopoietin
B-cell
corticosteroids

Antibody Azathioprine,
against CD154 cyclophosphamide,
T-cell cyclosporin,
corticosteroids,
danazol
Fig. 22.8 Standard and novel treatment strategies in immune thrombocytopenia (ITP). Standard treatments include corticosteroids,
intravenous immunoglobulin, danazol, and vinca alkaloids. Their sites and modes of action are illustrated. New therapies include monoclonal
antibodies against CD154 (CD40 ligand) and CD20 (on B cells, leading to transient B-cell depletion).
Source: Cines D.B., Blanchette V.S. (2002). Immune thrombocytopenic purpura. New England Journal of Medicine 346: 995–1008. Reproduced
with permission of New England Journal of Medicine.

neutropenia, autoimmune hemolytic anemia, pure red cell sequences. The antigen-binding domain binds to the CD20
aplasia, ITP, and combined hemolytic anemia and ITP (Evans antigen on B cells, while the Fc domain mediates B-cell lysis
syndrome) has shown responses in 15 of 21 patients treated; through recruitment of immune effector cells. Because of its
in 6 patients the response was sustained. Campath-1H specificity for B cells, rituximab has been viewed as a poten-
therefore appears to be an effective agent in severe refractory tial treatment for autoimmune disease, the rationale being
autoimmune disease. The drug is well tolerated, but because the reduction or elimination of autoantibody-producing B
it can precipitate bleeding during administration, it should cells, with concomitant improvement of the autoimmune
not be given in the presence of active bleeding (or active disease. A recent study by Stasi et al. reports on the efficacy
infection). of rituximab in the treatment of 25 patients with chronic
refractory ITP. Patients were treated if their platelet counts
were below 20 × 109 l−1 irrespective of symptoms, or at
Anti-CD20 monoclonal antibody therapy
higher platelet counts if bleeding or bruising was problem-
Rituximab, a genetically engineered chimeric human/mouse atic. All patients had received between two and five previous
anti-CD20 monoclonal antibody, has been developed treatments; eight had failed splenectomy. Rituximab was
as a treatment for B-cell lymphoproliferative disease administered in the same manner and dose as that used
(non-Hodgkin lymphoma). The antibody is an IgG κ in non-Hodgkin lymphoma. After four courses, 40% of
immunoglobulin comprising murine light- and heavy-chain patients achieved a platelet count of at least 50 × 109 l−1 ; five
variable-region sequences and human constant-region achieved complete remission (platelets >100 × 109 l−1 ); and
Autoimmune hematological disorders 313

Cytokine proteins orchestrate

Fc receptor

Y
Y XY Y
CTLA-4

Antigen-presenting T cell B cell Antibody


cell

Fig. 22.9 Costimulatory blockade may be beneficial in some autoimmune diseases. CTLA-4 linked to human immunoglobulin Fc
(CTLA-4-Ig) blocks the critical second signal between antigen-presenting cells and T cells, resulting in T-cell anergy. This should result in a reduction
in antibody production and amelioration of disease if the autoimmune disease is antibody mediated. CTLA-4-Ig treatment has been shown to be
of benefit to patients with psoriasis. Similarly, anti-CD40 ligand also blocks the second signal with similar results, and has been shown to be of
value in refractory immune thrombocytopenia (ITP).

five partial remission (platelets 50–100 × 109 l−1 ). Responses Costimulatory blockade
were seen during treatment with rituximab, with a peak
Therapies such as Campath-1H and anti-CD20 may not pro-
response up to four weeks after the end of treatment; 28%
duce lasting remission if the autoimmune B cells are driven by
had responses that lasted for more than six months.
dysregulated T cells, and a novel agent, CTLA-4-Ig, has been
The results suggest that the use of rituximab resulted in
evaluated in psoriasis in an attempt to block T-cell costimu-
responses similar to those given by other second-line agents
lation, thereby inducing anergy in the T-cell compartment.
used in ITP (including vinca alkaloids, cyclophosphamide,
CTLA-4-Ig, a fusion protein between CTLA-4 and the Fc
and azathioprine), around 40–50%, but sustained responses
portion of human immunoglobulin, binds to B7-1 and B7-2,
to these agents is usually seen in fewer than 20% of patients
blocking T-cell costimulation (Figure 22.9). This small trial
(i.e. lower than for rituximab). Rituximab would appear to be
showed that, at least within this group of patients, CTLA-4-Ig
useful for some patients with chronic symptomatic refractory
was able to improve the disorder and was shown to be safe.
ITP in whom there is a definite need to elevate the platelet
CTLA-4-Ig may have applications within other autoimmune
count to a safe level.
disorders, including ITP. If a drug such as CTLA-4-Ig were
The mechanism of action of rituximab in ITP has been
shown to be effective in ITP, not only would it provide an
assumed to be due to selective depletion of CD20+ B cells that
additional targeted treatment modality, it would also provide
subsequently affects autoantibody development. This con-
useful evidence of T-cell dysfunction in this disease. Interest-
cept was recently shattered when it was demonstrated, using a
ingly, the CTLA-4 gene has been mapped as a susceptibility
variety of sophisticated techniques to analyze T-cell parame-
gene in autoimmune thyroid disease and type 1 diabetes in
ters, that only when the abnormal T-cell subsets were normal-
humans.
ized was rituximab therapy effective. The reasons for these
results are not clear, but may relate to how B-cell popula-
Other options: Helicobacter pylori
tions may be either important in maintaining autoreactive
eradication
T-cell activation patterns or, by decreasing the total mass of B
cells, may cause a collapse of autoreactive T-cell stimulation Helicobacter pylori is the main cause of gastritis and peptic
and normalization of the T-cell repertoire, even as the B cells ulcer disease. It has also been implicated in the development
begin to return months after the therapy. What is perhaps of gastric adenocarcinoma and mucosa-associated lymphoid
more enlightening is the demonstration that the abnormal tumors, and in some autoimmune disorders. Previous stud-
T-regulatory (Treg) populations are indirectly targeted by rit- ies in ITP showed improvement in platelet counts after erad-
uximab therapy; the anti-CD20 treatment reverses the Treg ication of the bacterium in patients shown to be positive for
deficiency in patients with ITP and normalizes the autoim- H. pylori. More recently, Emilia et al. looked for the presence
munity. Taken together, these studies truly lend credence to of H. pylori in 30 patients with chronic refractory ITP. It was
the notion that attacking T cells in ITP, even indirectly by found in 13 of 30 patients (43.3%). Standard triple therapy
the destruction of B cells, is perhaps the only way to reduce for H. pylori eradication resulted in a complete response in 4
platelet destruction effectively. of 12 patients in whom the bacterium was eradicated, and a
314 Molecular Hematology

Thrombopoietin Nplate Promacta


O OH

TPO-R binding
Fc
domain
domain
(including spacer regions)

OH

N NH
H3C O
N N
CH3

(a) (b) (c) CH3

Fig. 22.10 Structures of thrombopoietin (TPO) and synthetic c-mpl ligands. (a) Native TPO is a 332-amino-acid glycoprotein with a
molecular mass of 60–70 kDa. It is the major humoral regulator of platelet production. (b) Nplate (romiplostim) is a 60 kDa synthetic “peptibody”
that does not share any amino acid homology to native TPO. It comprises a human immunoglobulin Fc domain linked via polyglycine to two
divalent mpl-binding peptide regions. The Fc component extends the half-life of the drug in the circulation, while the peptide “warhead” binds to
the TPO receptor, c-mpl, and activates signaling. (c) Promacta (eltrombopag) is an orally bioavailable hydrazone small molecule with a molecular
mass of 546 Da. Unlike TPO and Nplate, which bind the extracellular domain of c-mpl, Promacta is reported to bind to the transmembrane region
of c-mpl.
Source: Kuter D.J. (2007). New thrombopoietic growth factors. Blood 109: 4607–4616. © American Society of Hematology. Reproduced by
permission.

partial response in 2 of 12 (16.6%). The responses were main- covalently linked to a human Fc fragment. The Fc protein
tained for a median of 8.33 months. In addition, there are prolongs the half-life of the drug and the drug stimulates
other anecdotal reports of improvements in platelet counts megakaryopoiesis after subcutaneous injection. In contrast,
in adults and children with ITP after eradication of H. pylori. eltrombopag is a small molecule naphthalenesulfonic acid of
Larger studies are required to confirm these earlier findings, 564.6 Da molecular weight; it has similar potency to native
but from the available data triple therapy appears to offer a TPO and can be administered as an oral tablet (Figure 22.10).
non-immunosuppressive therapy for patients with refractory The two drugs have proven to be highly effective in raising
ITP and possibly other autoimmune diseases. platelet counts in patients with chronic ITP. Of interest is that
On the other hand, platelets express Toll-like receptor it now appears that approximately a third of romiplostim-
(TLR)4, and this has been shown to be responsible for the treated patients remain in remission even after 24 weeks off
thrombocytopenia induced by lipopolysaccharide adminis- TPO treatment. How this occurs is unknown, but it appears
tration in vivo. It now appears that bacterial products such that romiplostim rescues the Treg deficiency observed during
as lipopolysaccharide together with IgG bound to platelets active disease. It was shown that Treg function was increased,
can significantly enhance Fc-mediated platelet phagocytosis and the platelet counts correlated with circulating trans-
by mononuclear phagocytes. This suggests that infectious forming growth factor (TGF) levels, which may be due the
agents, in combination with antiplatelet antibodies, could increase in the platelet mass. Thus, the TPO-receptor ago-
affect platelet destruction in vivo, and may be at least one nists may not only directly induce thrombopoiesis, but also
explanation of why thrombocytopenia worsens in some modulate the immune system, perhaps by modulating Treg
patients with ITP during infections and, alternatively, reso- patients with ITP.
lves in other patients with ITP who are treated with bacterial
eradication therapy.
Novel therapies for the treatment
Thrombopoietin (TPO) mimetics
of other autoimmune diseases
Patients who fail first-line therapy can be treated with
thrombopoietin (TPO)-receptor agonists. Both eltrombopag Human and animal studies have been helpful in learning how
(Novartis) and romiplostim (Amgen) activate TPO recep- the immune system works in both health and disease, but can
tors on MK and induce platelet production via the JAK2 and such information be translated into better patient care? Until
STAT5 kinase pathways. These drugs stimulate MK prolifer- recently, the treatment strategy for autoimmune diseases
ation and differentiation, with a resulting increase in platelet has been to induce global immunosuppression in the hope
counts. Romiplostim is classed as an Fc peptide fusion pep- that the autoimmune process may be abrogated or stopped.
tibody and consists of two 14-amino-acid peptide dimers In some cases treatment is effective, but it is clear from
Autoimmune hematological disorders 315

longitudinal follow-up studies of patients with ITP that there used in small clinical trials include IL-10 and CTLA-4-Ig;
is very significant morbidity and mortality associated with the latter molecule is a recombinant protein comprising
our current treatments. Infection plays a major role in the the extracellular domain of CTLA-4 linked to the constant
death of patients with autoimmune disease, and such fatal- region of IgG1. CTLA-4-Ig downregulates activated T cells
ities are usually induced by immunosuppression. Now that and prevents activation of naı̈ve T cells. CTLA-4-Ig was well
we have a better understanding of the components of the tolerated in a small trial conducted by Bristol-Myers-Squibb
immune system and how these interact in disease, we should and the patients’ skin condition improved by more than 50%.
be able to develop targeted therapies that aim to modify spe- Further studies are required to confirm safety and efficacy.
cific components of the immune system, while leaving most There are studies of other agents used in the treatment of
of the immune system intact and able to fight infection. autoimmune disease, but there is insufficient space to dis-
Such therapeutic advances are in fact being made, and cuss these here. However, it would appear that our knowl-
many of these have been developed through knowledge edge of immunity and autoimmunity is now being used to
concerning specific components of the immune system in design more subtle and specific treatments for patients with
disease. We have now been able to develop targeted therapies autoimmune disease. It is expected that these treatments will
for rheumatoid disease, multiple sclerosis, psoriasis, diabetes, reduce immunosuppression, morbidity, and mortality and
and systemic lupus. We discuss each of these briefly in turn. offer modern solutions to these otherwise intractable dis-
eases. No doubt the future will see even more designer drugs
being developed for this fascinating group of diseases.
Rheumatoid arthritis and blockade
of TNF-α
Disease-modifying therapies, designed to reduce deformity Conclusions
in rheumatoid disease, have been around for a number of
years. Methotrexate is one of the main agents in this category. Autoimmune diseases are complex immunological disorders
Bone destruction in rheumatoid disease is partly mediated affecting 5% of the population. Until recently our under-
by macrophages through an inflammatory process. With our standing of the pathogenesis and treatment of these disorders
increased understanding of cytokines and their interactions, was severely limited. However, with greater understanding
specific modulators of the immune response have been devel- of the immune system in health and autoimmune disease
oped. Because TNF-α plays a key role in the inflammatory we are able to identify underlying abnormalities leading to
response, antibodies against TNF-α have been developed the develop of autoimmunity. With this new knowledge we
and shown to be of value. To date, two antibodies appear have been able to modify our therapies by replacing non-
promising: the first is a TNF-α receptor–IgG1 fusion protein, selective immunosuppressive treatment with more subtle
etanercept; the second, infliximab, is a monoclonal antibody targeted therapies.
directed against TNF-α itself. This form of therapy also
appears to have a place in the management of other autoim-
mune diseases, such as Crohn’s disease, psoriatic arthropathy,
Further reading
and ankylosing spondylitis.
Another cytokine involved in the pathogenesis of rheuma-
General
toid disease is IL-1. Blockade of the IL-1 receptor using a
recombinant antagonist may slow down the development of Janeway, C.A., Travers, P., Walport, M., and Capra, J.D. (1999). Immuno-
erosive bone disease, but from available data this appears to biology: The Immune System in Health and Disease, 4e. New York:
be less effective than TNF-α. Current Biology.
Roitt, I.M. and Delves, P.J. (2001). Roitt’s Essential Immunology, 10e.
Oxford: Blackwell Science.
Multiple sclerosis
IFN-β has recently been introduced for the treatment of mul- Immune system and HLA
tiple sclerosis. The available study data indicate that it may
Chien, Y.H., Gascoigne, N.R., Kavaler, J. et al. (1984). Somatic recombi-
delay the onset of the disease if started immediately after the
nation in a murine T-cell receptor gene. Nature 309: 322–326.
patient’s first attack of optic neuritis.
Delves, P.J. and Roitt, I.M. (2000a). The immune system. First of two
parts. N. Engl. J. Med. 343: 37–49.
Psoriasis Delves, P.J. and Roitt, I.M. (2000b). The immune system. Second of
two parts. N. Engl. J. Med. 343: 108–117.
This autoimmune skin disorder has been treated by TNF-α Klein, J. and Sato, A. (2000a). The HLA system. First of two parts.
blockade with and without methotrexate. Other agents N. Engl. J. Med. 343: 702–709.
316 Molecular Hematology

Klein, J. and Sato, A. (2000b). The HLA system. Second of two parts. Immune thrombocytopenia
N. Engl. J. Med. 343: 782–786.
Parkin, J. and Cohen, B. (2001). An overview of the immune system. Aslam, R., Speck, E.R., Kim, M. et al. (2006). Platelet Toll-like receptor
Lancet 357: 1777–1789. expression modulates lipopolysaccharide-induced thrombocytope-
Tonegawa, S. (1983). Somatic generation of antibody diversity. Nature nia and tumor necrosis factor-production in vivo. Blood 107: 637–
302: 575–581. 641.
Bussel, J.B. (2002). Novel approaches to refractory immune thrombocy-
topenic purpura. Blood Rev. 16: 31–36.
Tolerance Chow, L., Speck, E.R., Kim, M. et al. (2010). A murine model of severe
immune thrombocytopenia is induced by antibody- and CD8+ T
Dario, A., Vignali, A., Collison, L.W., and Workman, C.J. (2008). How cell–mediated responses that are differentially sensitive to therapy.
regulatory T cells work. Nat. Rev. Immunol. 8: 523–532. Blood 115: 1247–1253.
Kamradt, T. and Mitchison, N.A. (2001). Tolerance and autoimmunity. Cines, D.B. and Blanchette, V.S. (2002). Immune thrombocytopenic
N. Engl. J. Med. 344: 655–664. purpura. N. Engl. J. Med. 346: 995–1008.
Mackay, I.R. (2000). Science, medicine, and the future: tolerance and Coopamah, M.D., Garvey, M.B., Freedman, J., and Semple, J.W. (2003).
autoimmunity. Br. Med. J. 321: 93–96. Cellular immune mechanisms in autoimmune thrombocytopenic
Sarzotti, M. (1997). Immunologic tolerance. Curr. Opin. Hematol. 4: purpura: an update. Transfus. Med. Rev. 17: 69–80.
48–52. de Luis, D.A., Varela, C., de La Calle, H. et al. (1998). Helicobacter
pylori infection is markedly increased in patients with autoimmune
atrophic thyroiditis. J. Clin. Gastroenterol. 26: 259–263.
Autoimmunity Emilia, G., Longo, G., Luppi, M. et al. (2001). Helicobacter pylori erad-
ication can induce platelet recovery in idiopathic thrombocytopenic
Davidson, A. and Diamond, B. (2001). Autoimmune diseases. N. Engl.
purpura. Blood 97: 812–814.
J. Med. 345: 340–350.
Fujisawa, K., Tani, P., O’Toole, T.E. et al. (1992). Different specificities of
Maloy, K.J. and Powrie, F. (2001). Regulatory T cells in the control of
platelet-associated and plasma autoantibodies to platelet GPIIb-IIIa
immune pathology. Nat. Immunol. 2: 816–822.
in patients with chronic immune thrombocytopenic purpura. Blood
Sinha, A.A., Lopez, M.T., and McDevitt, H.O. (1990). Autoimmune
79: 1441–1446.
diseases: the failure of self tolerance. Science 248: 1380–1388.
Gasbarrini, A., Franceschi, F., Tartaglione, R. et al. (1998). Regression
Taneja, V. and David, C.S. (2001). Lessons from animal models for
of autoimmune thrombocytopenia after eradication of Helicobacter
human autoimmune diseases. Nat. Immunol. 2: 781–784.
pylori. Lancet 352: 878.
Kekomaki, R., Dawson, B., McFarland, J. et al. (1991). Localization of
Role of genetic and other factors in human platelet autoantigens to the cysteine-rich region of glycopro-
tein IIIa. J. Clin. Invest. 88: 847–854.
autoimmune disease
Kuwana, M., Kaburaki, J., and Ikeda, Y. (1998). Autoreactive T cells to
Albert, L.J. and Inman, R.D. (1999). Molecular mimicry and autoimmu- platelet GPIIb-IIIa in immune thrombocytopenic purpura. Role in
nity. N. Engl. J. Med. 341: 2068–2074. production of anti-platelet autoantibody. J. Clin. Invest. 102: 1393–
Benoist, C. and Mathis, D. (2001). Autoimmunity provoked by infec- 1402.
tion: how good is the case for T cell epitope mimicry? Nat. Immunol. McKenzie, C.G., Guo, L., Freedman, J., and Semple, J.W. (2013). Cellu-
2: 797–801. lar immune dysfunction in immune thrombocytopenia (ITP). Br. J.
Choy, E.H. and Panayi, G.S. (2001). Cytokine pathways and joint inflam- Haematol. 163: 10–23.
mation in rheumatoid arthritis. N. Engl. J. Med. 344: 907–916. Nieswandt, B., Bergmeier, W., Rackebrandt, K. et al. (2000). Identifica-
Criswell, L.A., Moser, K.L., Gaffney, P.M. et al. (2000). PARP alleles and tion of critical antigen-specific mechanisms in the development of
SLE: failure to confirm association with disease susceptibility. J. Clin. immune thrombocytopenic purpura in mice. Blood 96: 2520–2527.
Invest. 105: 1501–1502. Portielje, J.E., Westendorp, R.G., Kluin-Nelemans, H.C. et al. (2001).
Ermann, J. and Fathman, C.G. (2001). Autoimmune diseases: genes, Morbidity and mortality in adults with idiopathic thrombocytopenic
bugs and failed regulation. Nat. Immunol. 2: 759–761. purpura. Blood 97: 2549–2554.
Gianani, R. and Sarvetnick, N. (1996). Viruses, cytokines, antigens, and Provan, D. and Newland, A. (2002). Fifty years of idiopathic thrombo-
autoimmunity. Proc. Natl. Acad. Sci. U.S.A. 93: 2257–2259. cytopenic purpura (ITP): management of refractory ITP in adults. Br.
Marron, M.P., Zeidler, A., Raffel, L.J. et al. (2000). Genetic and phys- J. Haematol. 118: 933–944.
ical mapping of a type 1 diabetes susceptibility gene (IDDM12) to Semple, J.W. and Freedman, J. (2006). Mechanisms underlying autoim-
a 100-kb phagemid artificial chromosome clone containing D2S72- munity in hematology. Drug Discovery Today: Dis. Mech. 3: 231–
CTLA4-D2S105 on chromosome 2q33. Diabetes 49: 492–499. 235.
Spielman, R.S. and Ewens, W.J. (1996). The TDT and other family-based Stasi, R., Pagano, A., Stipa, E. et al. (2001). Rituximab chimeric anti-
tests for linkage disequilibrium and association. Am. J. Hum. Genet. CD20 monoclonal antibody treatment for adults with chronic idio-
59: 983–989. pathic thrombocytopenic purpura. Blood 98: 952–957.
Wanstrat, A. and Wakeland, E. (2001). The genetics of complex autoim- Sukati, H., Watson, H.G., Urbaniak, S.J. et al. (2007). Mapping helper
mune diseases: non-MHC susceptibility genes. Nat. Immunol. 2: 802– T-cell epitopes on platelet membrane glycoprotein IIIa in chronic
809. autoimmune thrombocytopenic purpura. Blood 109: 4528–4538.
Autoimmune hematological disorders 317

Zentilin, P., Savarino, V., Garnero, A. et al. (1999). Is Helicobacter pylori Kremer, J.M. (2001). Rational use of new and existing disease-
infection a risk factor for disease severity in rheumatoid arthritis? modifying agents in rheumatoid arthritis. Ann. Intern. Med. 134:
Gastroenterology 116: 503–504. 695–706.
Kuwana, M., Kawakami, Y., and Ikeda, Y. (2003). Suppression of
autoreactive T-cell response to glycoprotein IIb/IIIa by blockade
Therapeutic advances of CD40/CD154 interaction: implications for treatment of immune
thrombocytopenic purpura. Blood 101: 621–623.
Abrams, J.R., Lebwohl, M.G., Guzzo, C.A. et al. (1999). CTLA4Ig- Maini, R.N. and Taylor, P.C. (2000). Anti-cytokine therapy for rheuma-
mediated blockade of T-cell costimulation in patients with psoriasis toid arthritis. Ann. Rev. Med. 51: 207–229.
vulgaris. J. Clin. Invest. 103: 1243–1252. Mease, P.J., Goffe, B.S., Metz, J. et al. (2000). Etanercept in the treat-
Bell, S. and Kamm, M.A. (2000). Antibodies to tumour necrosis factor ment of psoriatic arthritis and psoriasis: a randomised trial. Lancet
alpha as treatment for Crohn’s disease. Lancet 355: 858–860. 356: 385–390.
Brandt, J., Haibel, H., Cornely, D. et al. (2000). Successful treatment Provan, D. and Newland, A. (2015). Current management of primary
of active ankylosing spondylitis with the anti-tumor necrosis factor immune thrombocytopenia. Adv. Ther. 32: 875–887.
alpha monoclonal antibody infliximab. Arthritis Rheum. 43: 1346– Semple, J.W., Aslam, R., Kim, M. et al. (2007). Platelet-bound
1352. lipopolysaccharide enhances Fc receptor-mediated phagocytosis of
Cines, D.B., Wasser, J., Rodeghiero, F. et al. (2017). Safety and efficacy IgG opsonized platelets. Blood 109: 4803–4805.
of romiplostim in splenectomized and nonsplenectomized patients Stasi, R., Cooper, N., Del Poeta, G. et al. (2008). Analysis of regulatory
with primary immune thrombocytopenia. Haematologica 102: 1342– T cell changes in patients with idiopathic thrombocytopenic purpura
1351. receiving B-cell depleting therapy with rituximab. Blood 112: 1147–
Kapur, R., Aslam, R., Guo, L. et al. (2016). Thymic-derived tolerizing 1150.
dendritic cells are up-regulated in the spleen upon treatment with Zufferey, A., Kapur, R., and Semple, J.W. (2017). Pathogenesis and thera-
intravenous immunoglobulin in a murine model of immune throm- peutic mechanisms in immune thrombocytopenia (ITP). J. Clin. Med.
bocytopenia. Platelets 25: 1–4. 6: 16–37.
Chapter 23 Molecular therapeutics in
hematology: gene therapy
William M. McKillop1 & Jeffrey A. Medin2
1 Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
2
Departments of Pediatrics and Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA

Introduction, 319 Adeno-associated virus gene transfer, 330


General comments on gene transfer/therapy, 319 Genetic immunotherapy, 331
Viral vectors for gene transfer, 321 Methods to improve gene therapy safety and suicide gene therapy, 334
Oncoretroviral gene transfer, 321 Conclusions, 335
Lentiviral gene transfer, 327 Further reading, 336
Adenoviral gene transfer, 329

(ALL). Several other CAR-T therapies are following close


Introduction behind this trajectory in what has become a fast-evolving
field in modern medicine.
Approved clinical investigation of gene transfer into humans
Since hematology has contributed so much to the gene-
began with the seminal trial of Rosenberg and colleagues,
sis and progression of human gene therapy, it is the purpose
which involved transplantation of genetically altered lym-
of this chapter both to reiterate the inherent promise and to
phocytes into patients. This landmark trial was closely fol-
revisit some of the remaining obstacles posed by the applica-
lowed by studies of therapeutic gene transfer using other
tion of gene transfer into humans employing hematopoietic
hematopoietic cells. Studies involving the blood system have
cells.
therefore been central to the development of human gene
Pluripotent hematopoietic stem cells (HSCs) are attrac-
therapy. Gene marking or gene therapy protocols are under
tive targets for gene therapy in humans because of their
increasingly intensive investigation worldwide, with the field
capacity for self-renewal and the systemic multilineage dis-
having grown to over 2900 approved clinical trials by the
tribution of their progeny (Figure 23.1). Sustained expres-
beginning of 2019.
sion of transgenes at clinically relevant levels in the progeny
Importantly, some of the first successes in the entire
of HSCs would result in novel and potentially curative treat-
field of gene therapy have recently been realized. Shenzhen
ments for a wide range of blood diseases, including, for exam-
SiBiono GeneTech Co. Ltd. obtained a drug license from the
ple, hemophilia A and B, hemoglobinopathies, hereditary
State Food and Drug Administration of China for Gendicine,
immune deficiencies, and some lysosomal storage disorders
an adenovirus-based therapy designed to treat head and
(LSDs). Even the partial correction of such blood disorders
neck squamous cell carcinoma in 2003. However, it was not
would have a substantial impact on the transfusion needs of
until 2012 and the approval of UniQure’s Glybera, an adeno-
the affected populations. Other hematopoietic cell subpopu-
associated virus-based therapy designed to treat lipoprotein
lations are also important targets for gene therapy. This dis-
lipase deficiency, that a gene therapy treatment strategy
cussion examines some of the targets of gene therapy involv-
was approved in the West. In 2016 Strimvelis, a lentiviral
ing the hematopoietic system and outcomes mediated by a
vector used to treat severe combined immune deficiency,
variety of gene transfer mechanisms.
became the second gene therapy approved in Europe. In
August 2017, the US Food and Drug Administration (FDA)
approved Kymriah (CTL019, tisagenlecleucel), a chimeric
antigen receptor T cell (CAR-T) therapy targeting CD19, General comments on gene
for the treatment of pediatric and young adult patients with transfer/therapy
relapsed/refractory B-cell acute lymphoblastic leukemia
Gene therapy has the potential to revolutionize medical treat-
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. ment by selectively repairing or augmenting the expression
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. of defective genes by the insertion of new genetic material.

319
320 Molecular Hematology

Remove
hematopoietic
stem cells

Add genetically Stem cells proliferate


modified virus Harvest and return and gene is distributed
to patient

Fig. 23.1 Ex vivo transduction of hematopoietic stem cells (HSCs). HSCs can be harvested from patients and vector transduced. The
transduced cells are returned to the patient, where all blood cells maturing from the gene-modified hematopoietic stem cells retain a copy of the
transgene. In theory, this gene transfer method could help distribute the gene product throughout the body at clinically relevant levels.

To date, selective repair of genetic defects within the host are still possible, as the short recognition sequences used
genome has proven to be a very difficult endpoint; however in the system are rarely unique in the genome. Although
genome editing technologies capable of such repair are tremendously promising, protocols using these gene editing
now being investigated. Targetable DNA cleavage reagents strategies are just beginning to enter the clinic. Thus, the
including zinc finger nucleases (ZFN) and transcription great majority of gene therapy studies conducted to date have
activator-like effector nucleases (TALENs) cut specific been gene augmentation protocols, involving co-expression
sequences of DNA, allowing for homologous recombination or even amplified overexpression of wild-type cDNA
with recombinant exogenous DNA and the introduction of sequences.
an edited sequence at the target site. However, ZFN-based Let us take a step back. An array of techniques has been
strategies appear to suffer from context-dependent recog- described to facilitate gene transfer into blood cells. A
nition site binding. There is also some degeneracy in the discussion of some of those methods is warranted. Such
TALEN-DNA binding specificity, both nucleases are highly techniques can be broadly grouped as either physical methods
sensitive to DNA methylation, and they both require very or viral vector methods. Physical methods of gene transfer
specific protein–DNA interactions. A third nuclease-based are generally of lower efficiency and usually provide only
strategy dependent solely on nucleic acid base pairing transient gene expression in the absence of selection. The
is the clustered regularly interspaced short palindromic advantages of physical methods of delivery are that genes
repeats/CRISPR associated protein 9 (CRISPR/Cas9) are transferred without viral sequences, which may affect
system. CRISPR/Cas9 offers an easy-to-engineer and partic- the biology of the target cell or the host. In addition, large
ularly efficient system for gene editing, but off-target effects or multiple genes can be transferred, and gene transfer
Molecular therapeutics in hematology: gene therapy 321

is independent of the proliferative status and cell-surface Viral gene transfer methods take advantage of facets of
receptor profile of the target cell. Those methods of physical the normal virus life cycle to facilitate the transfer of genetic
gene transfer that have been used in the clinical setting material into target cells. For synthesis of recombinant LV-
include electroporation (electrical fields that create channels based gene transfer systems, for example, the wild-type viral
in cell membranes, allowing passage of DNA into the cell), genome is modified by deletions of most incumbent viral
particle bombardment (microscopic gold beads labeled genes and insertion of therapeutic or marker sequences in
with DNA, for example, which are forced through the cell their place. The viral gene products necessary to produce
membrane by carbon dioxide (CO2 )-driven pressure), and recombinant virions are then provided in trans by trans-
liposomal encapsulation of DNA. Liposomal-mediated gene fections and replication-incompetent virions are produced.
delivery has gained most acceptance in the clinical arena, Replication-incompetent virions are deemed safe for clini-
driven by the higher gene transfer efficiencies obtained with cal applications because they transduce target cells only once;
newer liposomal formulations and by safety considerations they are unable to cause a subsequent infection because of the
as an alternative to viral vector-mediated gene delivery. absence of secondary expression of viral genes required for
Other non-viral gene delivery systems include the use of replication and virion packaging (Figure 23.2).
plasmid DNA alone (so-called naked DNA), the use of Retrovirus-based vectors are attractive for the treatment
synthetic polymers, transposons, and the adaptation of of conditions requiring stable long-term expression engen-
bacterial gene delivery systems. The use of plasmid DNA dered by genomic integration. Retroviruses also display a
alone is particularly efficacious in specific tissues such as fairly wide tropism, as well as minimal toxicity and minimal
muscle. Thus, non-viral DNA-based gene delivery is gaining adverse immune responses in human patients. Two retro-
in popularity for use in malignancy and infectious disease, viruses commonly used in clinical gene therapy protocols tar-
and in applications directed toward the vascular bed. In geting hematopoietic cells are oncoretroviruses and LVs.
cancer, DNA-based vaccines have been used in clinical trials
where the immunogenic antigen is encoded by the plasmid
itself, with or without an added immunostimulatory gene. Oncoretroviral gene transfer
Yet, while this approach is promising, clearly more work on
modulating the biology of the immune response is needed. Oncoretroviruses are double-stranded RNA viruses belong-
Physical methods such as electroporation, direct DNA ing to the gammaretrovirus genera of the family Retroviridae.
transfer, and liposome-mediated DNA transfer have been The viral RNA genome is reverse-transcribed into double-
used with varying levels of success for transferring genes stranded DNA, which integrates into the genome of tar-
into hematopoietic cells. However, transgenes rarely integrate get cells. Currently, oncoretroviral vectors derived from the
with physical methods of transfer, except transposons, and Moloney murine leukemia virus (MMLV) and other murine
the vectors are diluted in dividing daughter cells, and thus oncoretroviruses are used for clinical gene transfer protocols
are generally of little value to long-term stable gene transfer targeting hematopoietic cells.
applications. Nevertheless, when transient gene expression For gene transfer vectors, the wild-type oncoretroviral
is sufficient, such as in cancer immunotherapy applications, genome is modified by deleting most of the gag, and all
physical methods of gene transfer may find a niche. pol and env sequences. Viral sequences that are retained
in the oncoretroviral vector include the packaging signal
and the long terminal repeats (LTRs), which are necessary
Viral vectors for gene transfer for viral integration and often drive transcription of the
marking or therapeutic transgene in the absence of an added
As an alternative to the physical methods just described, heterologous promoter. These deletions render the vectors
protocols requiring higher-efficiency gene transfer or stable replication-incompetent while making approximately 6–8 kb
long-term gene expression make use of recombinant viral of space available for the insertion of desired transgene
vector transduction. The recombinant vectors used are mod- sequences. Stable oncoretroviral packaging cell lines have
ified from their wild-type state. The most commonly used been engineered to minimize the chance of accidental
viral backbones in clinical gene therapy trials were previously replication-competent retrovirus (RCR) production. Plas-
based on murine oncoretroviruses and human adenoviruses; mids used to create these stable packaging cell lines engineer
lately recombinant adeno-associated viruses (AAVs) and expression of key components of the parental virus, but have
lentiviruses (LVs) are preferred. The relative merits and dis- extensive deletions along with split genes and promoters.
advantages of some of these vector systems are outlined in These modifications have minimized recombination-prone
Table 23.1. The particular nuances of the acquired or inher- homologous sequences between the added gene transfer
ited disorder to be corrected determines which delivery sys- vector encoding the transgene of interest and the stable
tem is more appropriate. packaging cells, thereby significantly reducing the chance of
322 Molecular Hematology

Table 23.1 Some viral vectors used in the application of gene therapy

Helper-dependent Adeno-associated
Oncoretrovirus Adenovirus adenovirus virus Herpes virus Lentivirus Pox viruses

Advantages r Well-studied r Very efficient r Large cloning r Cell-cycle r Very efficient r Integrates r Efficient gene
r Integrates gene transfer capacity independent gene transfer r Efficient gene transfer
r High transgene r Not immunogenic r Safe r Cell-cycle transfer
expression independent r Large capacity

Disadvantages r Cell-cycle r Immunogenic r Technically r Limited cloning r Transient r Production r Transient

dependent r Transient cumbersome capacity r Cytotoxic r Public expression


r Promoter expression r Helper-virus r Helper-virus perception r Immunogenic

silencing r Pre-existing contamination contamination


r Safety issues immunity r Transient r Production

expression

Potential r Many involving r Cancer r Gene r Cancer r Cancer r Many involving r Cancer

applications hematopoietic immunotherapy augmentation immunotherapy immunotherapy hematopoietic immunotherapy


cells therapy r Gene r Pain cells
augmentation management r Non-dividing

therapy via muscle targets


r Brain delivery
Molecular therapeutics in hematology: gene therapy 323

Infectious Replication incompetent


HIV virus lentiviral vector

Transfer Packaging Envelope


Therapeutic plasmid plasmid plasmid
gene
Infectious
HIV virus HIV genes

HIV genes
Host nucleus
Host DNA

Replication
incompetent
lentiviral vector

Infectious
HIV virus

Target cell

Therapeutic
Therapeutic transgene
No virions
transgene released
No viral
proteins

Host nucleus

Host DNA
Fig. 23.2 Biology of the HIV retrovirus and the production of HIV-derived, replication-incompetent lentiviral vectors. For wild-type
retroviruses such as HIV, after binding and entry the provirus integrates into the genome of infected cells. Viral proteins and new mRNA molecules
are produced. Virions are then assembled for the next round of infection. For HIV-derived recombinant replication-incompetent lentiviral vectors,
the gag, pol, and env genes have been removed to allow space for subcloning of the transgene of interest. To make a recombinant lentiviral
vector, the transfer plasmid containing the therapeutic gene flanked by the HIV long terminal repeats (LTRs) is introduced into a production cell
line, along with separate plasmids engineering expression of gag, pol, and env genes in trans. The resulting replication-incompetent lentiviral
vector can then bind to, and be internalized by, a target cell wherein integration into the host genome occurs and the vector transgene product is
synthesized. Since the infected target cell lacks the genes necessary to form a new virion, the vector is not infectious secondarily.

RCR being generated. Another measure of safety is naturally (see later), investigators have also generated recombinant
present in oncoretroviral gene transfer systems as these oncoretroviruses with self-inactivating (SIN) 3′ LTRs. Thus,
vectors, when packaged in murine-based packaging cell lines upon reverse transcription, this promoter unit is modified,
at least, are rapidly inactivated by human serum. Taking a making it unable to drive transcription itself if productive
lesson from commonly used LV-based gene transfer systems recombination should occur, and also making it unable
324 Molecular Hematology

to drive transcription in the presence of viruses that may 3 Do malignant cells or their precursors contribute to the
encode cross-promoting functions. Lastly, along with the high relapse rates observed after myeloablative therapy and
inherent safety features described, all viral supernatants and autologous HSC transplantation?
patient cell samples that have been exposed to clinical-grade A number of groups have reported the consequences of
oncoretroviral supernatants are extensively tested for the infusing gene-marked bone marrow cells into humans and
presence of RCR and other possible contaminants prior to the contribution of contaminating tumor cells in the graft
infusion into patients. to disease recurrence. The first important observation from
Oncoretroviral vectors were one of the preferred gene these studies was that oncoretrovirus-mediated gene transfer
transfer vectors for early clinical gene therapy protocols. appears relatively safe. No detrimental effects, either on the
Their main advantage over other delivery systems was sta- autograft or in patients, have been reported in these marking
ble integration of the vector into the host cell genome, allow- studies. Replication-competent oncoretrovirus has also not
ing long-term transgene expression in the target cell and been detected at appreciable levels in patients participating in
its progeny. These vectors also generated minimal immune clinical trials. That said, in two clinical gene therapy protocols
responses themselves due to the extensive deletions of wild- that will be further described, T-cell leukemias developed in
type coding subunits that they have undergone. Disadvan- five patients and appear to have resulted from the integration
tages of oncoretroviral vectors include the requirement of tar- of the therapeutic provirus into the regulatory region of a cer-
get cells to be in cycle for genomic integration, the specificity tain oncogene (see later). The second important observation
of virion/receptor binding, which can reduce infection rates from earlier gene-marking studies was that multiple HSCs
into some target cells depending on the env pseudotyping contributed to long-term hematopoiesis, albeit at relatively
employed, the extensive periods of ex vivo culturing that may low levels. Interestingly, rather than all daughter hematopoi-
be required to obtain efficient gene transfer into key target etic cells being derived from a single HSC, it thus appears that
cells such as human HSCs, and the randomness of the inte- multiple stem cells contribute to the formation of the renew-
gration event itself. ing blood system.
Some of the earliest work with oncoretroviral vectors In the first gene-marking studies in children reported by
involved gene-marking studies in hematopoietic cells. By Brenner and colleagues, 2–15% of clonogenic hematopoietic
marking transduced cells, the long-term distribution and progenitor cells were marked after autologous bone marrow
survival of transplanted cells could be followed in vivo. transplantation. The marker gene was detectable for up to
The reporter gene that was used most often in early clin- four years after transplant and was found in granulocytes,
ical studies was the bacterial neomycin phosphotransferase B cells, and T cells, at least by PCR. In the earlier adult
(neoR ) gene which, when expressed, confers resistance to gene-marking studies, however, oncoretroviral transduction
the neomycin analog G418. Other marker genes have also of marrow or peripheral blood HSCs has resulted in detec-
been used and include those sequences encoding for the tion of integrated vector in only a very low percentage of
murine heat-stable antigen, the human CD24 and CD25 anti- peripheral blood cells. For example, in one study, although
gens, the truncated nerve growth factor receptor, modified the marker gene persisted for up to two years, neoR-positive
CD4 or CD19 or CD34 antigens, and an array of fluores- cells could only be detected intermittently with analyses
cent proteins. The stable and unique integration pattern of employing PCR. Work by a number of groups suggested
proviral DNA in the genome of marked cells can provide a that modification of the transduction protocols would
permanent marker for individual hematopoietic or malig- result in a significant improvement in the engraftment
nant cells and their clonal descendants. This marking pat- of genetically modified HSCs. Along these lines, since
tern can be established using a polymerase chain reaction preclinical experience indicated that the use of bone marrow
(PCR)-based analysis that has been developed called ligation- stroma enhances gene transfer into HSCs, this approach was
mediated PCR, which provides information on the actual site evaluated in some early clinical trials. However, the results
of integration of the provirus. Clinical applications in which of this adaptation proved to be no better than those just
gene marking has provided new and important information discussed and, in general, are very similar to those observed
include the infusion of oncoretrovirally marked, autologous, in the pediatric gene transfer studies.
tumor-infiltrating lymphocytes into patients with advanced In total, data on more than 40 patients enrolled in gene-
melanoma and the infusion of oncoretrovirally marked bone marking studies during bone marrow transplantation for
marrow or peripheral blood into patients with myeloid acute myeloid leukemia, neuroblastoma, chronic myeloid
leukemia, myeloma, and neuroblastoma. The study of such leukemia, breast cancer, myeloma, acute leukemia, or non-
patients offers three important lines of investigation: Hodgkin lymphoma have been presented. Of the relapsed
1 Is retroviral-mediated gene transfer relatively safe? patients, gene-marked tumor cells have been detected in
2 Do genetically altered bone marrow or blood stem cells a high percentage in many studies. In one patient with
contribute to long-term hematopoiesis? acute myeloid leukemia, the simultaneous detection of a
Molecular therapeutics in hematology: gene therapy 325

Table 23.2 Strategies for optimization of stable gene transfer into hematopoietic stem cells

Strategy Method

Inducing recipient cells to cycle Optimization of ex vivo cytokine stimulation


Collection of cells during recovery phase after myeloablation or mobilization
Culture on stromal layers
Increased cell–virus contact Centrifugation of cells and virus during transduction (spinoculation)
Viral supernatant flow-through systems
Coat dishes with fibronectin fragment
Higher viral titers and multiple exposures
Increase viral receptor levels on target cells Increase levels of amphotropic receptor by phosphate depletion
Transfer viral receptor into cell by adenovirus or adeno-associated virus
Target subpopulations of cells that have high levels of receptors
Alternatively pseudotyped recombinant retroviruses Exploiting GALV, RD114, 10A1 receptors for entry
VSV-G envelope to expand tropism and allow virion concentration
Modified retroviral vectors to increase efficiency and Lentivirus- and foamy virus-based vectors
infect non-cycling cells
Positive selection of transduced cells Add positive selectable marker to vector: metabolic, fluorescent, cell-surface

cytogenetic marker along with the neoR gene confirmed that against the toxicity of chemotherapeutic drugs, i.e. chemo-
gene-marked cells contributed to relapse. A second critical protection studies, are under exploration. The gene-marking
observation was made a number of years ago when Rill and studies just mentioned suggest that normal bone marrow
colleagues reported that a multiplicity of neuroblastoma cells cells could be removed, transduced with a vector engineering
in the graft contributed to relapse. The high frequency of expression of a drug resistance gene, and returned to patients.
gene-marked relapse, despite the very low frequency of trans- Theoretically, such protected cells would then expand clon-
fused malignant cells, strongly suggests that a large percent- ally after chemotherapy treatment and confer relative resis-
age of tumor cells in the graft contribute to relapse or, alterna- tance to a cytotoxic or cytostatic agent, allowing further dose
tively, that tumor cells susceptible to retroviral gene marking escalation and a potential cure of some patients. Further-
are uniquely capable of engraftment and clonal expansion. more, such drug resistance genes, if expressed in a multi-
The gene-marking studies mentioned set the stage for cistronic format with a second therapeutic gene product, may
the investigation of multiple maneuvers designed to increase even serve as selectable markers allowing ex vivo or in vivo
long-term gene transfer efficiency into HSCs. Some of the enrichment or “preselection” of functionally gene-marked
strategies being pursued are described in Table 23.2. These cells, which may further enhance observed clinical outcomes.
include increasing the true direct target cell-to-virus contact The original chemoprotection studies made use of
ratio by incorporating prior HSC enrichment, altering retro- the human multidrug resistance 1 (MDR-1) gene (P-
viral envelope utilization, or increasing co-localization of glycoprotein). The approach used for gene transfer there
vector and cell in gene transfer protocols. Newer vector sys- was generally similar to that used for the human gene-
tems improving on both oncoretroviral and lentiviral back- marking trials. MDR-1 transduced and transplanted cells
bones and incorporating alternative envelope pseudotyping engrafted and conferred drug resistance to bone marrow
may also increase gene transfer efficiency, while refinement cells in vivo, and also allowed for positive selection of
of growth factor combinations may more efficiently induce MDR-1 transduced cells by chemotherapy. Infusion of
HSC cycling in shorter time periods, thus improving retrovi- oncoretroviral MDR-1-modified HSCs was found to be safe,
ral integration without compromising engraftment. Many of with no deleterious effects of infusing such gene-modified
these approaches have now been incorporated into clinical cells observed, but only minimal levels of gene marking
protocols and have likely contributed to some of the recent were noted following transplantation. Multiple studies have
successes in this field, as described shortly. investigated an alternative strategy aiming to overexpress
Oncoretroviral vectors have also been used in chemopro- mutant methylguanine methyltransferase (MGMT) in HSCs
tection studies. Cancer chemotherapy often involves induced to allow them to survive multiple rounds of chemotherapy
hematopoietic system toxicity, leading to myelosuppression. targeting glioblastoma, for example. The mutant MGMT was
Gene therapy strategies aiming to protect hematopoietic cells safe, allowed an increase in the mean number of tolerated
326 Molecular Hematology

chemotherapeutic cycles, and resulted in improved clinical transfer protocols mentioned were adapted in that protocol,
outcome for the seven patients treated. and higher levels of gene marking (up to 25% of CFU-C ini-
The first human clinical gene transfer trials for inher- tially) and functional correction were demonstrated. Long-
ited single-gene disorders used oncoretroviral vectors and term multilineage hematopoietic cell marking was also found
focused on adenosine deaminase (ADA) deficiency, a con- in both patients. This study demonstrated, without the con-
dition that leaves the body unable to produce leukocytes founding implications of ERT, that stable long-term correc-
and thus predisposed to infection. Recombinant oncoretro- tion of this immune deficiency could be accomplished using
viruses containing the normal human ADA cDNA were this therapeutic approach.
transferred into either peripheral blood T lymphocytes, Impressive results have also been demonstrated in efforts
bone marrow cells, or cord blood cells from ADA-deficient to correct another inherited immunodeficiency, X-linked
patients. T-lymphoid cells expressing the normal ADA gene SCID (SCID-X1). This is caused by a deficiency of the com-
have a selective growth and survival advantage over ADA- mon γ-chain subunit of the receptors for the cytokines inter-
deficient cells, even though patients were maintained on leukin (IL)-2, IL-4, IL-7, IL-9, and IL-15, and is thereby not
pegylated-ADA enzyme replacement therapy (ERT) for ethi- a candidate for soluble factor augmentation therapy. Expres-
cal reasons. In those early studies, patients who had received sion of the γ-chain was also expected to offer a growth advan-
multiple infusions of autologous ADA-transduced blood cells tage to productively transduced cells. This was indeed the
had increased levels of enzyme in their serum, and up to 20% case and in a first report in 2000, two patients were shown to
of their peripheral blood T cells were found to carry provirus have fully corrected immune function as a result of the gene
for some period. Indeed, long-term follow-up has revealed therapy. Since this landmark first description of this benefi-
that one patient has maintained this level of marking for over cial outcome, other SCID-X1 patients have also been treated
10 years. In two of the three initial trials in which patients by this method. The original clinical trials treated 20 patients
received autologous marrow or cord blood cells transduced and resulted in the majority of these individuals retaining
with ADA cDNA-containing oncoretroviruses, between 12% improved and stable immune function. Yet even with these
and 40% of colony-forming units (CFUs) were transduced, impressive results, this study highlights areas where the field
and genetically marked cells were found for greater than of gene therapy must still progress in its understanding. This
one year after infusion. In the one study, by Hoogerbrugge is because of reports that five of the patients receiving the
and colleagues, in which provirally marked cells were not corrective 𝛾-chain gene in two separate clinical gene ther-
maintained for greater than six months, there was lower in apy protocols for SCID-X1 employing similar vectors and
vitro gene transfer efficiency of 5–12% CFU prior to trans- transduction protocols went on to develop acute T-cell lym-
plant. In the cord blood study, there was evidence for a phoblastic leukemia-like disease approximately three years
selective growth advantage of T cells, as there were higher after the transplantation of oncoretrovirally transduced cells.
levels of marked T cells than myeloid cells, even while the In at least two of these patients the development of this prolif-
patients were maintained on progressively lower doses of erative disorder has debatably been ascribed to a deleterious
PEG-ADA therapy. ERT was withdrawn from one patient integration event that may have caused dysregulated expres-
and the number of T cells carrying the provirus increased to sion of a proto-oncogene called LMO2. However, it should
30%; however, the total number of B lymphocytes and natu- be emphasized that since analogous vector backbones and
ral killer cells dropped, and the patient had reduced immune transduction conditions have been used for other studies,
function. That patient subsequently resumed PEG-ADA such as the ADA-SCID trials mentioned earlier, without the
treatment. emergence of adverse events, the possibility exists that these
Inherited immunodeficiencies, such as severe combined leukemia-like diseases are a specific consequence of over-
immunodeficiency (SCID) of ADA (see earlier) and X-linked expression of the 𝛾-chain gene itself, since it impacts many
SCID (see later), have become focal points for the potential diverse signaling pathways that affect a number of cellular
benefits and some of the potential hazards of gene therapy. functions in vivo.
The studies in ADA-SCID likely represent the first tangible Still, a second attempt at SCID-XI gene therapy was
correction of an inherited disorder by stable transfer of a launched in 2008, with results published in 2012 detailing
therapeutic gene into primitive hematopoietic cells and their follow-up on the nine patients enrolled at sites in France
progeny, although it is difficult to state this unequivocally and the USA. The oncoretroviral vector was modified to be
due to the simultaneous administration of ERT. However, a SIN vector and to contain deletions in the viral enhancer
those initial results have been surpassed recently for ADA- sequences that may have activated LMO2. Of the eight
SCID in a study published by Aiuti and colleagues, where patients who received treatment, seven displayed immune
productive transfer of the ADA gene was effected into CD34+ system reconstitution, and analysis of vector insertion dis-
cells of non-myeloablated recipients for whom ERT was not played significantly reduced clustering within LMO2 or other
available. Many of the incremental improvements in the gene proto-oncogenes. Although very promising, these children
Molecular therapeutics in hematology: gene therapy 327

will be followed for 15 years and longer to further assess the II, and III, metachromatic leukodystrophy, neuronal ceroid
efficacy and safety following treatment. lipofuscinoses, and Pompe disease.
Results from another study involving the hematopoietic Collectively, the results from the gene transfer studies for
system have also generated some close scrutiny. Clonal dom- genetic diseases described here (and others not directly men-
inance of cells with specific integration events was observed tioned) illustrate several points that will likely impact on the
in a recombinant oncoretrovirus-based clinical gene therapy clinical success of gene transfer protocols for other single
trial for the X-linked form of chronic granulomatous dis- gene inherited disorders.
ease. There patients have impaired immune function and 1 The presence in patients of cells carrying the provirus for
cannot sufficiently resist bacterial and fungal infections, due greater than one year has demonstrated the feasibility of gene
to an inability of neutrophils to generate superoxide ions; therapy and newer vector systems may further enhance this.
expression of the CYBB gene can abrogate this. Interest- 2 The transfer of genes that provide a growth or survival
ingly, the amplified integration locus involved the MDS1 advantage can provide long-term expression and mainte-
and EVI1 genes in that case, which differs from that men- nance of transduced cells.
tioned in the SCID-X1 trials and may reflect alterations 3 A selective advantage of transduced cells can compen-
in the activity of myeloid cells post-transduction with this sate for a modest gene transfer efficiency and yet, if too
cDNA, rather than a specific sequence preference for vector strong, may lead to amplification of deleterious transforma-
integration. tion events.
Another particular group of inherited disorders currently 4 Using the incrementally optimized protocols, gene deliv-
undergoing gene therapy preclinical and clinical trials are the ery using recombinant viruses along with their subsequent
LSDs. LSDs are genetic diseases that result from the loss of transgene expression levels may presently be sufficient to cor-
metabolic genes that normally work in the lysosome to break rect a number of disorders that are directly or indirectly man-
down particular cellular molecules. Over time, the result of ifested in the hematopoietic system.
the disrupted enzyme activity is an accumulation of substrate, Although still used in groundbreaking clinical stud-
leading to a broad spectrum of debilitating symptoms. As ies tracking toward FDA approval, enthusiasm about
most LSDs are the result of the loss or malfunction of a single oncoretroviral vectors was somewhat reduced following the
enzyme, and as even a small fraction of cells producing work- leukemogenesis reports post-X-SCID treatment described
ing copies of the malfunctioning enzymes may be sufficient earlier. We now know that oncoretroviral vectors have a
to cross-correct lysosomes and positively impact the disease, predisposition to integrate at certain genetic “hotspots,”
LSDs are promising targets for gene therapy. including within genes, and in regions immediately sur-
Gaucher disease is an LSD resulting from a deficiency rounding transcriptional start sites, leading to the potential
in the enzyme glucocerebrosidase that is manifested mainly for dysregulation of nearby endogenous genes. In an effort to
in macrophages, and it has been proposed that this defect overcome these obstacles and improve therapeutic safety and
may be especially amenable to treatment by therapeutic gene efficiency, replication-incompetent complex retroviruses
transfer into HSCs. Results from two earlier clinical gene based on lenti-retroviruses (LVs) and human foamy virus
transfer studies targeting this disorder have been reported. have been developed. These vectors can integrate into
Mobilized peripheral blood or marrow CD34+ cells from these and other key non-dividing target cells and are less
Gaucher patients were transduced with an oncoretroviral predisposed to integration near active genes.
vector that engineered expression of glucocerebrosidase and
then infused into non-myeloablated autologous recipients. In
both studies, transduced cells were detected at low levels in Lentiviral gene transfer
blood and/or marrow leukocytes. One patient who received
cells transduced with an MFG-based oncoretroviral vector LVs and human foamy virus are also members of the family
manifested increased levels of enzyme activity corresponding Retroviridae. They are sometimes termed “complex” retro-
to 50% of normal, which was maintained for 12 months after viruses, in contrast to oncoretroviruses, which are termed
infusion. No therapeutic benefit or increased enzyme activity “simple” retroviruses. Foamy viruses have a large genome
was detected in other patients or in the other study. Develop- (and hence a large transgene carrying capacity), do not cause
ment of viable murine models for this LSD have been very any known disease, and can be used for in vivo gene trans-
beneficial for the testing and implementation of novel gene fer because they are not effectively inactivated by human
therapy strategies involving some of the conditional manip- serum. LVs also show significant promise as gene transfer
ulations already mentioned or employing newer vectors. In vehicles and many laboratories around the world and subse-
addition to Gaucher disease, gene therapy clinical trials are quent clinical trials have adopted this alternative delivery sys-
underway investigating a number of other LSDs, including tem. Current LVs contain less than 25% of the HIV-1 genome
Fabry disease, mucolipidosis, mucopolysaccharidosis type I, and seem to be much less susceptible to “shutdown” effects
328 Molecular Hematology

compared with recombinant oncoretroviruses. To further patient blood. Many gene therapy trials using LVs have been
increase safety, many laboratories now use third-generation completed to date and many more are underway.
recombinant LVs that have a SIN 3′ LTR. Much like SIN As previously mentioned, Kymriah, the first gene therapy
oncoretroviruses, SIN LV minimize the risk that replication- approved in the USA, utilizes the LV delivery system. Kym-
competent lentivirus will be created. SIN vectors also reduce riah is a CAR-T cell therapy based on infusion of autolo-
the chance that the expression of endogenous genes located gous T cells transduced with an LV engineering expression
near the insertion site will be enhanced by viral LTR activity, of a CD19-directed CAR for the treatment of ALL. Kym-
and reduce or eliminate transcriptional interference between riah has been evaluated in a number of clinical trials. One
the LTR and the promoter driving the transduced transgene. trial was based on the treatment of 30 children and adults;
To further minimize the chance of recombination lead- 27 patients experienced complete remission, including many
ing to the production of replication-competent lentivirus, patients in whom stem cell transplantation had failed. The
some laboratories even use up to seven separate plasmids CAR-T cells were detectable in the blood, bone marrow, and
in transient transfections of packaging cell lines to gener- cerebrospinal fluid of patients who displayed a response. All
ate recombinant virions (although three separate plasmids patients experienced cytokine-release syndrome, but this was
is more the norm). LVs can be readily pseudotyped with controllable with tocilizumab. Positive outcomes in a subse-
alternative envelope proteins, such as VSV-g, which expands quent global trial, ELIANA, helped secure FDA approval. A
the tropism of the virions and also allows stable concentra- phase II trial investigating Kymriah’s effect on diffuse large
tion of effective viral titer by ultracentrifugation. LVs can B-cell lymphoma is underway (JULIET). Kymriah has also
also transduce non-dividing cells. There is thus considerable been used in the clinic to treat chronic lymphocytic leukemia
optimism that recombinant LVs will overcome some of the (CLL). In one study, 14 patients were treated with autolo-
shortcomings of MMLV-based oncoretroviruses by facilitat- gous T cells transduced with Kymriah LV and infused at
ing delivery of transgenes of interest to a wide spectrum of varying doses. More than 50% of patients displayed a posi-
cell types, including HSCs. Furthermore, due to a marked tive response. The in vivo expansion and persistence of the
increase in LV-mediated transduction efficiencies over previ- CAR-T cells correlated with responses, no patient displaying
ous methods, it may be possible now to better manage other a complete remission relapsed, and minimal residual disease
secondary components of the ex vivo gene therapy proce- was not detectable in patients displaying a complete remis-
dure, such as enhancing engraftment of transduced cells in sion. All responding patients developed B-cell aplasia and
recipients by minimizing culture periods and exposure to experienced cytokine release syndrome.
differentiation-inducing cytokines, for example. Alterations Although the first LV-based gene therapy won FDA
have also been made to LV backbones such as the addi- approval in 2017, the first protocol in humans to employ
tion of a central polypurine tract and a woodchuck hepati- recombinant LVs was conducted in the early 2000s by
tis virus post-transcriptional regulatory element, to possibly Levine and colleagues. That trial aimed to use the VRX496
enhance nuclear import or enhance transgene expression lev- (Lexgenleucel-T) LV to treat HIV infection. VRX496 is an
els, respectively. LV engineering expression of an antisense transgene against
As previously mentioned, LV genomic integration the HIV envelope. The LV is delivered to autologous CD4
patterns differ from oncoretroviral integration patterns. T cells, which are then reinfused into the patient. Infection
Unlike oncoretroviral vectors, which show a propensity with HIV results in antisense expression and disruption of
to integrate into promoter-proximal regions, LVs tend to HIV replication. In one clinical study, a single IV infusion
distribute throughout open chromatin and thus present less of VRX496-containing CD4 T cells into five patients was
of an insertional mutagenesis risk. That said, due to their found to be safe. Engraftment was observed. No evidence of
increased efficiency, LVs tend to integrate into permissive insertional mutagenesis was noted, nor was recombination
cells in higher copy numbers than oncoretrovirus-based between the gene transfer vector and wild-type HIV. Patients
vectors. This increase can be good in order to generate displayed improved CD4 counts, and in one patient a sig-
higher levels of transgene products for correction, but it nificant decrease in viral load was seen. A subsequent study
also necessitates careful planning concerning the effective investigated multiple VRX496 infusions into 17 patients
multiplicity of infections (MOIs) and the tolerance of and noted a significant decrease in viral load. No evidence
the infected cell population to multiple proviral copies. of clonal selection of LV-transduced T cells or integration
Indeed, it may turn out that MOIs with LVs actually need enrichment near oncogenes was detected. Several other
to be reduced to minimize this multiplicity effect in some clinical trials have used gene therapy to target HIV. One
protocols. Importantly, in a landmark first LV study in study involved treatment of four HIV patients with an LV
HIV-AIDS patients, no detectable recombination occurred delivering three RNAs: a shRNA targeting an exon shared by
between the gene transfer vector and wild-type HIV-1, even the HIV genes tat and rev, an RNA hairpin transactivating
when the latter was present in very high copy numbers in region decoy that antagonizes viral transactivation, and
Molecular therapeutics in hematology: gene therapy 329

an anti-CCR5-specific hammerhead ribozyme intended Furthering the LSD clinical work presented earlier, Luigi
to block viral entry. The therapy was well tolerated and Naldini and Alessandra Biffi and colleagues have targeted
the shRNA was detected in primary blood mononuclear metachromatic leukodystrophy, a demyelinating lysosomal
cells and/or primary blood granulocytic cells at least six storage disease caused by arylsulfatase A deficiency. Patients
months post-treatment in all four patients. Another ongoing received autologous HSCs transduced with a LV-encoding
study is testing an LV engineering expression of an anti- arylsulfatase A; 18 months post-treatment the great major-
CCR5 shRNA and C46 peptide to block HIV entrance into ity of patients displayed stable engraftment of gene-corrected
T cells. cells, increased arylsulfatase A activity in circulating HSCs
LVs have also been used to treat X-linked adrenoleukodys- and in cerebrospinal fluid, a reduction of the glycosphin-
trophy (ALD), a severe brain demyelinating disorder result- golipid buildup normally associated with disease pathology,
ing from a deficiency in ALD protein. Autologous HSCs were and improved functional outcome.
removed from two patients and engineered to express ALD Direct bilateral injection of an LV into the putamen
by LV vector transduction. Between 24 and 30 months post- was used to restore dopamine production in patients with
treatment, polyclonal reconstitution had occurred, with 9– advanced Parkinson’s disease; 15 patients received the LV, 3
14% of granulocytes, monocytes, and T and B lymphocytes at a low dose, 6 at a mid dose, and 6 at a high dose. During the
expressing the ALD protein. Beginning 14–16 months after first 12 months of follow-up, 51 mild and 3 moderate adverse
treatment, cerebral demyelination had stopped. events were reported; however, a significant improvement
Marina Cavazzana-Calvo’s group has published on their in mean motor scores off medication was reported in all
first patient treated with a LV delivering an anti-sickling β- patients.
globin gene into autologous HSCs. Fifteen months after treat- Attempts to use vectors other than traditional HIV-1-
ment, therapeutic anti-sickling β-globin was still detectable derived LVs have been made and are now entering the clinic.
and sickle crises, as well as other phenotypic hallmarks One such strategy is based on equine infectious anemia virus
of sickle cell disease, were absent in the patient. No (EIAV) and has been used to treat neovascular age-related
adverse events unrelated to busulfan conditioning were macular degeneration (NVAMD). NVAMD is an eye disease
reported. Cavazzana-Calvo’s group also used LVs to treat β- leading to vision loss. Endostatin and angiostatin were deliv-
thalassemia, a form of thalassemia resulting from reduced ered by EIAV LV to patients with advanced NVAMD by sub-
or absent hemoglobin β chain synthesis that can result in retinal injection in three dosing cohorts. Each dose was well
severe anemia. More than 30 months after LV β-globin gene tolerated with no toxicities. Long-term transgene expression
transfer, an adult patient with severe β-thalassemia who had and reduced disease progression were noted.
required monthly transfusions since childhood had been
transfusion independent for the previous 21 months.
Several trials have been conducted using LV-based gene Adenoviral gene transfer
therapy treatments for Wiskott–Aldrich syndrome, a rare
immunodeficiency associated with severe microthrombocy- Recombinant adenoviral gene transfer systems are com-
topenia, eczema, recurrent infections, and susceptibility to monly used for some gene therapy applications, especially
autoimmunity and lymphomas. In one study, autologous in immunotherapy protocols. Adenoviruses infect cells effi-
HSCs corrected by LV transduction were infused in seven ciently in vitro and in vivo, express high levels of trans-
patients; 24 months post-treatment six of the seven patients gene products, can infect both cycling and stationary cells,
were alive and showed sustained clinical benefit, with sub- and exhibit wide tissue tropisms. They can also be pro-
stantially reduced disease-related days of hospitalization. The duced at very high titers and have a large carrying capac-
patient who died succumbed to a pre-existing drug-resistant ity for foreign cDNAs. Adenoviral vectors have been used
herpes virus infection. In a separate study, three patients to efficiently transfer genes into a variety of human tumors
were treated analogously and showed stable engraftment with by direct injection, and into circulating hematopoietic pro-
improvements in platelet counts, immune functions, and genitor and malignant cells in ex vivo transduction proce-
clinical scores. Importantly, in these three patients, the LV- dures. Nevertheless, circulating B and T lymphocytes have
mediated therapy did not induce integrations near onco- generally proven relatively resistant to adenoviral-mediated
genes, and no aberrant clonal expansion was observed 20– gene transfer. So had CD34+ hematopoietic cells, until it was
32 months following the study. In a second cohort of four found that serotype 35 fiber knob domains can help over-
patients treated by this same protocol, LV-transduced HSCs come this blockade. Furthermore, many patients have pre-
repopulated both the bone marrow and the periphery, with existing immunity to common adenoviral serotypes used
a normal distribution of B-cell subsets, and led to normal in clinical protocols. This can lead to a strong immune
immunoglobulin and autoantibody expression in all treated response to the vector and severely restricts readministration
patients. efficacy.
330 Molecular Hematology

The receptor for most adenoviruses used in gene trans- useful only for the delivery of relatively small transgenes.
fer applications has been identified as the coxsackie-and- As usually produced, AAVs have titers of approximately
adenovirus receptor protein, and has been useful in deter- 106 particles ml−1 and can be concentrated to greater than
mining the mechanism of adenovirus binding and cell entry. 109 particles ml−1 , although in each preparation many
Adenoviral vectors do not integrate into the genome at an non-infectious particles are generated. Human CD34+
appreciable frequency and are lost from most cycling tar- hematopoietic stem/progenitor cells have been transduced
get cells within weeks of transduction. Thus, in comparison with recombinant AAV vectors, with up to 80% of CFU
to integrating vectors, adenovirus may appear comparatively carrying the transgene. Interestingly, the optimal use of
safe; however, they are not without their own set of risks. In recombinant AAV-based gene transfer vectors to impact the
1999 teenager Jesse Gelsinger died due to a massive immune hematopoietic system may be in secondary manifestations.
response generated to the adenovirus vector he had been In a landmark study, Kay and colleagues injected recom-
treated with in a trial at the University of Pennsylvania. Ade- binant AAV vectors that engineered expression of factor IX
noviral constructs have since been refined, with many potent (F.IX) into skeletal muscles of severely afflicted hemophilia
immuno-stimulatory viral sequences having been eliminated B patients. Long-term vector persistence was observed in
so as to leave only the necessary inverted terminal repeats, that study, as were slight increases in the circulating levels
and thus reduce the immunogenicity of the vector. Oncolytic of the corrective proenzyme. Subsequent studies aimed to
adenovirus vectors, which replicate only in permissive cancer improve on the limited F.IX expression seen in this early AAV
cells, have been developed and hold promise for the treat- work. Using a new AAV serotype (AAV8) and an improved
ment of solid tumors and leukemic cells. For example, one transgene design, a dose-escalation study infusing F.IX AAV8
such vector system only replicates in cells harboring defective into peripheral veins was conducted. F.IX expression was
p53 tumor-suppressor gene function. With these modifica- stable three years after infusion in most patients and aver-
tions, adenoviruses may be ideal vectors if the generation of aged 5.1% of normal in the high-dose group. The treatment
an immune response and/or short-term high levels of trans- was well tolerated. However, it should be noted that some
gene expression are desired; however, they are not suitable for of these patients were given an immunosuppressive corticos-
therapies that require long-term expression or integration of teroid four to eight weeks post-treatment. Importantly, most
the transgene. of the high-dose patients were able to reduce their exogenous
F.IX treatment following gene therapy.
Considerable work has also gone into investigating the use
Adeno-associated virus gene transfer of AAV to treat type 2 Leber congenital amaurosis (LCA).
LCA is an inherited retinal dystrophy that causes loss of
Versions of the human parvovirus adeno-associated virus vision at an early age. In three independent stage I/II trials
(AAV) are also being used more frequently as vehicles using Luxturna (voretigene neparvovec, hRPE65), an AAV2
for therapeutic gene transfer. AAVs are non-pathogenic in construct, a total of 20 patients were treated by subretinal vec-
humans and establish a latent infection in the absence of ade- tor administration. In all three trials, all patients experienced
novirus itself or helper functions provided by other viruses. improvements in visual sensitivity, peaking roughly a year
Wild-type AAV integrates at some frequency into a specific post-treatment. In two of the three trials, the patients began
site on human chromosome 19q13.3-qter, which appears to to lose the AAV-confirmed benefit after two to three years.
be a relatively benign location. Only the AAV inverted termi- However, sustained improvement in their condition was
nal repeats are required as transcriptional units for recombi- noted in the third trial. A following phase III study reported
nant AAV vectors, which allows about 4 kb for foreign inserts that 65% of participants displayed maximal improvement in
given the size of the parental viral genome. However, both the the trial’s primary endpoint: bilateral multiluminance mobil-
efficiency and specificity of integration are lost without the ity testing, one year after treatment. On the back of this study,
wild-type genes; recombinant AAV integrates at a very low Luxturna is a front-runner to be the first AAV gene therapy
frequency. Due to the risk of insertional mutagenesis posed approved by the FDA.
by integrating vectors, this, along with low immunogenicity, In α1-antitrypsin (AAT) deficiency, mutations in the SER-
has become a major selling point for AAV gene therapy. PINA1 gene led to reduced liver secretion of AAT and
With that said, there have been reports of AAV delivery impaired anti-protease activity in the lung, leading to early-
resulting in hepatocellular carcinoma formation in mice, onset pulmonary emphysema. In an early trial, AAV2 encod-
and wild-type AAV2 has been found to integrate near and ing the AAT cDNA sequence was injected intramuscularly
potentially dysregulate oncogenes in human hepatocellular into the upper arm of 12 patients at various doses, all of which
carcinomas. Thus, further study on recombinant AAV were below comparable levels used in rodent studies. Serum
gene therapy vector integration is warranted. AAV vectors AAT levels were found to be above background in only 1 of
also carry a small genetic payload, leaving them clinically the 12 subjects dosed, which was fairly unsurprising given
Molecular therapeutics in hematology: gene therapy 331

the relatively low dosage used. A follow-up trial made use of these encouraging allogeneic responses require a haploiden-
an AAV1 vector, as it displayed 100-fold higher hAAT serum tical T-cell donor, which is not available to the vast majority
levels than that seen with AAV2 constructs in murine mus- of patients. Another more widely applicable approach is the
cle transduction models. This follow-up AAV1 study resulted use of autologous tumor cells that have been genetically engi-
in detectable AAT in the serum of almost all patients, and neered to express immunostimulatory cytokines. Trials using
they maintained subtherapeutic levels of this factor for at this strategy in myeloma, low-grade lymphoma, leukemia,
least 90 days and up to one year post-treatment. A phase 2 and CLL are being pursued.
trial followed aiming to employ a dose escalation to achieve Many tumor cells express unique antigens on their cell
therapeutic transgene levels. To reach the maximum dose, surface, either alone or as proteolytically cleaved peptides in
AAV was delivered in 100 separate intramuscular injections. association with major histocompatibility complex (MHC)
A linear relationship between dose and serum AAT levels was class I molecules. The specificity and sheer quantity of these
noted; however, even at the maximum dosage serum AAT tumor-associated antigens (TAAs) may allow immune effec-
levels were still below a therapeutically relevant level. tor cells to distinguish tumor from normal tissue (Fig-
Another AAV-based gene therapy trial investigated treat- ure 23.3). Some of these TAAs have been isolated and shown
ment of spinal muscular atrophy type 1 (SMA1) in 15 infants. to be recognized by T-cell receptor (TCR) complexes on
SMA1 is caused by low levels of the survival motor neuron human cytotoxic T lymphocytes. This has engendered gene
protein and affects all muscles in the body. That study was a therapy strategies to augment T-cell-mediated eradication of
dose-escalation trial delivering an AAV9-based (AVXS-101) tumors. Indeed, while to date cancer has been the primary
construct intravenously through a peripheral limb vein. Thus target of the bulk of clinical gene therapy protocols, this area
far, results appear positive, as this treatment schema has led has seen limited success. That is until the results of a land-
to improved survival as well as improved motor skills, espe- mark study were published in 2006 by Morgan and colleagues
cially in the higher-dose cohorts. demonstrating definitive success in this field. The cDNAs for
Attempts at using DNA-editing ZFNs (see earlier) have the α and β chains of a TCR against a melanoma antigen were
begun in the clinic, with one study using AAV to deliver subcloned into an oncoretroviral vector and used to trans-
an anti-CCR5 ZFN into 12 HIV patients. Years following duce peripheral blood lymphocytes of melanoma patients.
treatment, the gene-modified T cells could be detected in Following infusions of the transduced cells into 17 patients,
circulation, but only at low levels. Many other AAV vectors two patients demonstrated actual sustained tumor regres-
are currently being evaluated in both preclinical and clinical sions as determined by standard criteria. Furthermore, one
proof-of-concept studies. year after infusions both of the responding patients had high
levels of gene-transduced cells in their circulation (20–70%
of peripheral blood mononuclear cells).
Genetic immunotherapy A further evolution of this immune augmentation strategy
has led to the creation of chimeric antigen receptors (CARs).
A number of potential approaches using gene therapy for As described earlier, Kymriah, the first gene therapy approved
hematological malignancies can be considered (Table 23.3). in the USA, is a CAR-based strategy. CARs use engineered
Proof of concept for immunotherapy as a valid approach to receptors endowing a patient’s own immune effector cells
the treatment of hematological malignancy has been pro- with specificity against a particular TAA. A TCR specific to
vided by earlier clinical studies that demonstrated that infu- the TAA is not required; rather, monoclonal antibodies raised
sion of allogeneic T cells can eradicate minimal disease in against the TAA are used as the platform for CAR approaches.
patients relapsing after allogeneic transplant. Unfortunately, The single-chain variable fragment of a monoclonal antibody

Table 23.3 Approaches to gene therapy targeting hematological malignancies

Strategy Genes employed Problems and merits

Gene replacement p53, p16, Rb Gene delivery to every cell required


Specific to defective cancer cell
Gene inhibition BCR-ABL, MYC, CCND1 (cyclin D1), BCL-2 Gene delivery to every cell required
Suicide genes Thymidine kinase, cytosine deaminase Immunogenicity and bystander effect contribute
Drug resistance genes MDR-1, DHFR, MGMT Stem cell gene delivery required
Immunotherapy IL-2, IL-12, B7-1, CD40L, GM-CSF, Systemic response, autoimmunity a theoretical problem
tumor-associated antigens (TAAs) For TAAs, requires that tumors express foreign antigen
332 Molecular Hematology

Antigen-presenting cell T cell Effect

Adhesion
Peptide
Adhesion receptor T-cell recruitment
and killing of
MHCI T-cell receptor IL–2 target cells
expressing
Costimulatory receptor peptide
Costimulation
γ interferon
IL–12

Tumor cell
Adhesion
Peptide
Signaling
Adhesion receptor
defective

MHCI T-cell receptor Results in anergy

Costimulatory receptor

Gene-engineered tumor cells

Insert Adhesion
gene of Adhesion receptor
interest Restoration
of cell killing
MHCI T-cell receptor IL–2
B7–1
Costimulatory receptor

IL–12

Fig. 23.3 Some facets of drug resistance immunotherapy. Efficient signaling of T cells occurs following adhesion, engagement of the major
histocompatibility complex (MHC) class I/T-cell receptor signal, and after a costimulatory signal is received. Under normal circumstances,
professional antigen-presenting cells (APCs) present processed peptides in the groove of the MHC class I to T cells. Tumor cells are often deficient
in one or all of the components required to function as APCs, because they lack an appropriate tumor antigen, cannot process the antigen, or are
deficient in the adhesion molecules, MHC class I, or costimulatory molecules required to generate a T-cell response. These missing components can
be provided or expression enhanced using gene transfer techniques. By overcoming the deficiencies of the tumor cell, the gene-engineered cells
can serve as autologous cancer vaccines presenting foreign antigen to the host T cells.

against a TAA is fused to the intracellular signal components 2015 over 200 patients had undergone CD19-targeted CAR-
of a TCR and other costimulatory signaling domains. These modified T cell clinical trials for the treatment of ALL, with
constructs can be transduced into the patient’s own effector reported positive response rates hovering over 80%.
immune cells and, when reintroduced into the patient, pro- As more CAR-T cell trials enter the clinic, variations of
vide transduced cells with the ability to recognize cancer cells the core strategy have emerged (as have playful adapta-
expressing that TAA. Although still in their infancy, CAR-T tions of the CAR acronym). Multiplexing TCR complexes to
cell clinical trials have shown considerable promise treating broaden the immune response and overcoming antigen drift
blood cancers. Clinical trials are underway for ALL, CLL, dif- and subsequent tumor evasion, adaptation of recombinant
fuse large B-cell lymphoma (DLBCL), follicular lymphoma, LVs to increase gene transfer frequencies, and incorporation
and multiple myeloma, among other indications. CD-19, a of methods to overcome immune dampening mechanisms
B-cell TAA, is the most well-studied CAR antigen. By driven by the tumors themselves may increase the potency
Molecular therapeutics in hematology: gene therapy 333

of these strategies in future protocols. Examples of the new patients have sufficient immune capacity for this kind of
CARs undergoing preclinical work will be listed here. manipulation, this can cause upregulation of the immune
T cells redirected for universal cytokine killings (TRUCKs) response against that specific TAA. The most immuno-
co-express both a CAR and an antitumor cytokine. Cytokine logically powerful (so-called professional) APCs are bone
production can be induced by T-cell activation and thereby marrow–derived dendritic cells. Dendritic cells express
recruit immune cells to tumor sites. Universal CAR-T cells MHC class I and II, B7-1, B7-2, CD40, ICAM-1, and LFA-3.
are also engineered from allogeneic T cells to eliminate They are capable of presenting processed antigen for days,
endogenous TCR and/or HLA molecules, and thus prevent and are potent stimulators of immunity when administered
graft-versus-host disease (GVHD) and transplant rejection. as vaccines to animals. Dendritic cells modulate immune
Self-driving CARs co-express both a CAR and a tumor lig- responses in part by secretion of IL-12 (hence the notion
and chemokine receptor to enhance tumor homing. Armored of converting tumor cells into APCs by introduction of
CARs secrete active cytokines or express ligands that improve IL-12 and B7-1 sequences). Dendritic cells can be readily
CAR-T cell efficacy and persistence. The “armor” agent is expanded from bone marrow progenitors in vitro using
chosen based on knowledge of the tumor microenviron- cytokine-supplemented medium (useful cytokines include
ment to further enhance CAR-T cell efficacy and persis- Flt3L, TNF-α, GM-CSF, and IL-4). Such cells may also be
tence. Self-destructing CARs can be engineered by transient genetically engineered by a variety of methods to express
CAR expression obtained by electroporation of RNA into TAAs, thereby presenting peptides in a proper context; this
the cell, or by inducing apoptosis with cell suicide systems. holds promise for the immunotherapy of hematological
Conditional CAR-T cells are inactive until binding an exoge- malignancies and for some solid tumors. Clinical trials
nous small molecule that activates the CAR. Marked CAR-T have been performed in this area targeting a number of
cells express both a CAR and a known tumor epitope that TAAs. Results have indicated that immune responses can be
existing therapies can clear. If adverse events result from generated in patients. Current work is focused on enhancing
treatment, administration of the anti-epitope therapy will that immune outcome and capitalizing on that facet to
clear the marked CAR-T cells and hopefully alleviate symp- decrease tumor burdens and/or treat metastatic disease.
toms. Tandem CAR-T cells express a CAR consisting of two As alluded to earlier, tumor cells known to express poten-
linked recognition domains that both must interact with a tially antigenic peptides manage to evade host immuno-
target on the tumor cell. A dual CAR-T cell expresses two surveillance and proliferate in vivo. Thus, tumor cells may
separate CARs with different targets: one CAR is fused to lack or downmodulate expression of the necessary acces-
only the CD3ζ intracellular domain, and the second CAR sory signals required to induce expression by immune effec-
is fused to only the costimulatory intracellular domain(s). tor cells of cytokines that are necessary for activation and
Thus, dual CAR-T cell activation also requires co-expression directed in vivo expansion of cytotoxic T lymphocytes. The
of two targets on a tumor. A safety CAR contains both a end result is anergy, a failure of T cells to respond to the tumor
CAR and an antigen-binding domain linked to an intracel- antigen. In addition to optimal presentation of antigen to the
lular inhibitory domain. If the tumor expresses antigens for TCR, efficient activation of naı̈ve T cells requires a second
both the CAR and the inhibitory domain scFv, the CAR costimulatory signal. It is now appreciated that molecules of
will be inactive. Thus, these CAR-T cells only become acti- the B7 family (B7-1/CD80, B7-2/CD86) on APCs engaging
vated when tumors display the CAR target, but lack the CD28/CTLA-4 receptors on T cells play a key role in this pro-
inhibitory receptor target. Bionic CARs combine CAR-T cells cess, inducing autocrine IL-2 production and T-cell prolifera-
with bispecific antibodies (antibodies featuring the benefits tion. Murine models have demonstrated that T-cell-mediated
of two monoclonal antibodies and differing binding specifici- rejection of tumors can be induced by transduction of tumor
ties in one construct) to improve killing efficiency. Many of cells with such costimulatory molecules. In the absence of
these novel CAR-based systems may make their way into the costimulatory signals, it is possible to bypass this require-
clinic soon. ment by ectopic expression of cytokines, and thus overcome
To date, the completed CAR trials are encouraging, but or prevent anergy of the immune effector cells. Proof of prin-
as these studies have occurred only recently, a considerably ciple that cytokine gene-transduced tumor cells can prevent
longer follow-up period will be required to provide data on tumor engraftment has been obtained. Such models have also
long-term patient safety/efficacy responses. shown that transduction of the genes for various cytokines,
Another facet of immunotherapy for cancer that has such as IL-2, IL-4, IL-6, IL-7, IL-12, IFN-γ, GM-CSF, and
received substantial interest is the application of gene TNF-α, into murine tumors not only led to primary rejec-
delivery techniques to induce specific immune responses tion of the modified cells, but often elicited protective immu-
in patients by directing efficient antigen-presenting cells nity against subsequent tumor challenge with unmodified
(APCs) to present peptides from such TAAs to T cells. Where tumor cells. Furthermore, in such models, synergy has been
334 Molecular Hematology

demonstrated between molecules with varying mechanisms population. The antibody–immunotoxin conjugate is admin-
of action, for example IL-2, IL-12, and B7-1. istered in vivo to specifically eradicate transduced trans-
planted cells. As this strategy relies on the administration of
toxic substances in vivo, a significant understanding of the
Methods to improve gene therapy physiological response to the toxin is required to minimize
safety and suicide gene therapy toxicity and maximize bioavailability of exogenous effector
molecules.
With the obvious importance of stably integrating vectors Another suicide strategy is the use of molecular switches
in gene therapy protocols involving the hematopoietic sys- that engage apoptotic pathways in transduced cells. One
tem, and given the deleterious outcomes in clinical trials for such strategy is the inducible caspase 9 (iCasp9) system.
one inherited disorder (see earlier), it is appropriate that the The iCasp9 transgene encodes a chimeric receptor engi-
field has directed attention to studying the safety of such gene neered to be activated by the addition of an exogenous small
delivery agents. As already discussed, extensive sequenc- molecule, referred to as a chemical inducer of dimeriza-
ing analyses have revealed that LVs and oncoretroviral vec- tion (CID). Induced dimerization of iCasp9 results in activa-
tors have different integration patterns. Yet, in both cases, tion of the mitochondrial apoptosis pathway in transduced
such integrations are still fairly random. Efforts are under- cells. Clinically, the iCasp9 system has been used in five
way to tether the viral integrases to specific sequences in the patients who received stem cell transplantation for relapsed
genome, thereby directing proviral integration into specific leukemia and DLI. On addition of the CID, a rapid elim-
areas of chromatin; however, to date these strategies have ination of transplanted T cells resulted. Thus, this promis-
yielded limited success, with the specificity of integration ing suicide system is currently being evaluated in several
being only minimally altered. CAR-T-based clinical trials. However, it is important to note
Another strategy is the incorporation of insulator elements that iCasp9-based cell fate control requires transgene expres-
into the viral 3′ LTR. As the 3′ LTR of retroviral vectors is sion above a certain threshold level to induce apoptosis, and
copied to the 5′ LTR during the intracellular generation of thus transduced cells remaining post-CID application may
provirus, insulator elements provided in the 3′ LTR should prove difficult to eliminate, rendering this strategy less use-
yield inserted retrovirus surrounded by insulator motifs, and ful for applications requiring a more complete elimination of
thereby reduce the likelihood of viral activation of host genes engineered cells.
near the site of insertion. A problem here is that such manip- Perhaps the best-studied suicide genes are enzymes which,
ulations usually reduce vector titer dramatically. Perhaps the when expressed by transduced target cells, confer suscepti-
most studied safety system is the usage of gene transfer bility to drug-induced cell death by specifically converting
to endow target cells of interest with “suicide” factors that normally non-toxic prodrugs into potent cytolytic or cyto-
allow their selective eradication should deleterious outcomes static molecules. The gene most commonly employed in clin-
arise. This approach certainly has applications in hemato- ical trials in this context is the human herpes simplex virus
logical transplantation, and also in numerous other devel- (HSV) type 1 thymidine kinase transgene, which confers sen-
oping research fields employing different candidate popula- sitivity to the drugs ganciclovir and aciclovir, among others.
tions, such as embryonic stem cells and induced pluripotent Use of thymidine kinase is further enhanced for some direct
stem cells. tumor applications by diffusion of the converted prodrug into
At least three general applications of suicide gene therapy neighboring cells, and thus a bystander effect occurs (Fig-
can be envisioned. One is direct tumor therapy, where the ure 23.4). The obvious limitations of this treatment approach
vector is injected into the tumor mass and patients are given are that not all cells targeted will be successfully gene mod-
the prodrug, which is activated only in the tumor. The sec- ified and thus, even with the bystander effect, only a frac-
ond application is in reduction of GVHD after donor lym- tion of malignant cells will be destroyed. Nevertheless, appli-
phocyte infusion (DLI). Here, if symptoms of GVHD appear, cations of this type are in clinical trials for treatment of
productively transduced and transplanted cells can be selec- solid tumors. Another proven clinical application of this sui-
tively removed by the addition of prodrug. Lastly, if a truly cide approach, which may actually have a higher proba-
portable system exists, such safety elements could conceiv- bility of success, is in the prevention of GVHD following
ably be incorporated into any cell transplanted out of its nor- transplantation. These trials were first performed by Bonini
mal context or into any gene therapy vector, in order to pro- and colleagues in patients who received T-cell-depleted allo-
tect transduced cells from genotoxicity and the development geneic bone marrow transplantation, followed by infusions
of leukemias should such outcomes occur. of oncoretrovirally transduced lymphocytes. The transduced
One class of suicide genes makes use of antibody– cells demonstrated antitumor activity in five patients. Three
immunotoxin conjugates engineered to specifically bind patients developed GVHD, which was controlled by the addi-
unique cell surface markers transduced into the target cell tion of ganciclovir.
Molecular therapeutics in hematology: gene therapy 335

Tumor

Gap
junction
between
cells

TK TK
protein protein
TK
protein

Ganciclovir
Vector carrying prodrug
herpes simplex thymidine
kinase (TK) gene

TK
TK

TK

Death of TK
expressing cells
Fig. 23.4 Basis of suicide gene therapy. The metabolic product of the activity of the thymidine kinase suicide gene appears capable of diffusing
into neighboring cells via gap junctions, which join the cells together. This non-specific diffusion allows a greater effective cell killing percentage
than might be predicted using direct gene transfer efficiency alone. Because cells are dying and releasing tumor antigen into the local milieu of the
tumor, it is possible that such suicide gene therapies will synergize with immune-based treatment strategies.

The HSV thymidine kinase system relies on a slow and


fairly inefficient mechanism to convert ganciclovir into its
Conclusions
toxic form, and the enzyme itself is a foreign protein, which
While it is still a relatively young field, some successes have
has resulted in antithymidine kinase immune responses
been observed in clinical gene therapy trials. Some concerns
being reported in patients receiving transduced cells. This
have also been raised. Development and implementation
can lead to premature clearing of the transplanted cell popu-
of gene delivery systems have led to the accumulation
lation. It should also be noted that many transplant patients
of important secondary knowledge. For example, gene
are already on prophylactic ganciclovir to reduce the possi-
delivery methods are now routinely used in most basic
bility of cytomegalovirus infection.
biology laboratories to study many different processes.
For the reasons noted, Sato and colleagues have attempted
Viral entry mechanisms are much better understood and
to improve on the HSV thymidine kinase system. They use a
immune responses initiated following viral infections are
human enzyme (thymidylate kinase) that has been minimally
better characterized. As a result of the use of integrating
modified to efficiently convert azidothymidine monophos-
vectors, it has also been demonstrated that some T cells in
phate to azidothymidine diphosphate. This system allows
humans are actually very long-lived and that multiple HSCs
for killing of non-cycling cells through mitochondrial mem-
contribute to hematopoiesis. Much more has been learned
brane disruption. In vitro and in vivo testing of such a sui-
concerning mechanisms that contribute to the development
cide safety system has been completed, and recombinant LV
of leukemias. Some concepts and approaches to molecular
that engineers expression of this variant enzyme is being pro-
therapy of diseases that interface with the blood system
duced for eventual clinical trials.
336 Molecular Hematology

have been described in this chapter; many other therapeutic Miller, D.G., Adam, M.A., and Miller, A.D. (1990). Gene transfer by
strategies are being developed. Indeed, the advent and retrovirus vectors occurs only in cells that are actively replicating at
success of human gene therapy trials combined with rapid the time of infection. Mol. Cell. Biol. 10: 4239–4242.
advances in gene transfer technology and implementation of Mitchell, R.S., Beitzel, B.F., Schroder, A.R. et al. (2004). Retroviral DNA
differential delivery systems, along with the full sequencing integration: ASLV, HIV, and MLV show distinct target site prefer-
ences. PLoS Biol. 2: E234.
of the human genome, are setting the stage for a new era in
Mulligan, R.C. (1993). The basic science of gene therapy. Science 260:
human medicine. The field of human molecular medicine 926–932.
will surely continue to expand and make inroads in a thera- Naldini, L. (2015). Gene therapy returns to centre stage. Nature 526:
peutically relevant way. It seems likely that study of the blood 351–360.
system, both in the laboratory and in patients, will continue Naldini, L., Blomer, U., Gallay, P. et al. (1996). In vivo gene delivery and
to play a pivotal role in the further development of this area. stable transduction of nondividing cells by a lentiviral vector. Science
272: 263–267.
Pawliuk, R., Bachelot, T., Wise, R.J. et al. (1999). Long-term cure of the
photosensitivity of murine erythropoietic protoporphyria by prese-
Further reading lective gene therapy. Nat. Med. 5: 768–773.
Qin, G., Takenaka, T., Telsch, K. et al. (2001). Preselective gene therapy
General for Fabry disease. Proc. Natl. Acad. Sci. U.S.A. 98: 3428–3433.
Thornhill, S.I., Schambach, A., Howe, S.J. et al. (2008). Self-inactivating
Brenner, M.K. (2001). Gene transfer and the treatment of hematological
gammaretroviral vectors for gene therapy of X-linked severe com-
malignancy. J. Intern. Med. 249: 345–358.
bined immunodeficiency. Mol. Ther. 16: 590–598.
Jinek, M., Chylinski, K., Fonfara, I. et al. (2012). A programmable dual-
RNA–guided DNA endonuclease in adaptive bacterial immunity. Sci-
ence 337: 816–821.
June, C.H. (2007). Adoptive T cell therapy for cancer in the clinic. J. Clin. Oncoretroviral gene transfer
Invest. 117: 1466–1476. Abonour, R., Williams, D.A., Einhorn, L. et al. (2000). Efficient
Melchiorri, D., Pani, L., Gasparini, P. et al. (2013). Regulatory evaluation retrovirus-mediated transfer of the multidrug resistance 1 gene into
of Glybera in Europe – two committees, one mission. Nat. Rev. Drug autologous human long-term repopulating hematopoietic stem cells.
Discovery 12: 719–723. Nat. Med. 6: 652–658.
Miller, A.D. (1992). Human gene therapy comes of age. Nature 357: 455– Aiuti, A., Slavin, S., Aker, M. et al. (2002). Correction of ADA-SCID by
460. stem cell gene therapy combined with nonmyeloablative condition-
Rosenberg, S.A., Aebersold, P., Cornetta, K. et al. (1990). Gene transfer ing. Science 296: 2410–2413.
into humans: immunotherapy of patients with advanced melanoma, Blaese, R.M., Culver, K.W., Miller, A.D. et al. (1995). T lymphocyte-
using tumor-infiltrating lymphocytes modified by retroviral gene directed gene therapy for ADA-SCID: initial trial results after 4 years.
transduction. N. Engl. J. Med. 323: 570–578. Science 270: 475–480.
Rosenberg, S.A. and Restifo, N.P. (2015). Adoptive cell transfer as per- Cavazzana-Calvo, M., Hacein-Bey, S., de Saint Basile, G. et al. (2000).
sonalized immunotherapy for human cancer. Science 348: 62–68. Gene therapy of human severe combined immunodeficiency (SCID)-
Williams, D.A. and Smith, F.O. (2000). Progress in the use of gene trans- X1 disease. Science 288: 669–672.
fer methods to treat genetic blood diseases. Hum. Gene Ther. 11: Deisseroth, A.B., Zu, Z., Claxton, D. et al. (1994). Genetic marking
2059–2066. shows that Ph+ cells present in autologous transplants of chronic
myelogenous leukemia (CML) contribute to relapse after autologous
Viral vectors for gene transfer bone marrow transplant in CML. Blood 83: 3068–3076.
Dunbar, C.E., Cotter-Fox, M., O’Shaughnessy, J.A. et al. (1995). Retro-
Bergelson, J.M., Cunningham, J.A., Droguett, G. et al. (1997). Isolation virally marked CD34-enriched peripheral blood and bone marrow
of a common receptor for Coxsackie B viruses and adenoviruses 2 cells contribute to long term engraftment after autologous transplan-
and 5. Science 275: 1320–1323. tation. Blood 85: 3048–3057.
Crystal, R.G. (1995). Transfer of genes to humans: early lessons and Fischer, A., Hacein-Bey-Abina, S., and Cavazzana-Calvo, M. (2010). 20
obstacles to success. Science 270: 404–410. years of gene therapy for SCID. Nat. Immunol. 11: 457–460.
Dilloo, D., Rill, D., Entwistle, C. et al. (1997). A novel herpes vector Hesdorffer, C., Ayello, J., Ward, M. et al. (1998). Phase I trial of
for the high-efficiency transduction of normal and malignant human retroviral-mediated transfer of the human MDR1 gene as marrow
hematopoietic cells. Blood 89: 119–127. chemoprotection in patients undergoing high-dose chemotherapy
Horn, P.A., Topp, M.S., Morris, J.C. et al. (2002). Highly efficient and autologous stem-cell transplantation. J. Clin. Oncol. 16: 165–
gene transfer into baboon marrow repopulating cells using GALV- 172.
pseudotype oncoretroviral vectors produced by human packaging Hoogerbrugge, P.M., van Beusechem, V.W., Fischer, A. et al. (1996).
cells. Blood 100: 3960–3967. Bone marrow gene transfer in three patients with adenosine deam-
Miller, A.D. and Buttimore, C. (1986). Redesign of retrovirus packaging inase deficiency. Gene Ther. 3: 179–183.
cell lines to avoid recombination leading to helper virus production. Howe, S.J., Mansour, M.R., Schwarzwaelder, K. et al. (2008). Insertional
Mol. Cell. Biol. 6: 2895–2902. mutagenesis combined with acquired somatic mutations causes
Molecular therapeutics in hematology: gene therapy 337

leukemogenesis following gene therapy of SCID-X1 patients. J. Clin. Saar, G. and June, C.H. (2014). Going viral: chimeric antigen receptor T-
Invest. 118: 3143–3150. cell therapy for hematological malignancies. Immunol. Rev. 263: 68–
Kohn, D.B., Weinberg, K.I., Nolta, J.A. et al. (1995). Engraftment of 89.
genemodified umbilical cord blood cells in neonates with adenosine Zielske, S.P., Reese, J.S., Lingas, K.T. et al. (2003). In vivo selection of
deaminase deficiency. Nat. Med. 1: 1017–1023. MGMT (P140K) lentivirus-transduced human NOD/SCID repop-
Mossoba, M.E. and Medin, J.A. (2006). Cancer immunotherapy using ulating cells without pretransplant irradiation conditioning. J. Clin.
virally transduced dendritic cells: animal studies and human clinical Invest. 112: 1561–1570.
trials. Expert Rev. Vaccines 5: 717–732.
Onodera, M., Ariga, T., Kawamura, N. et al. (1998). Successful periph-
Adeno-associated virus gene transfer
eral T-lymphocyte-directed gene transfer for a patient with severe
combined immune deficiency caused by adenosine deaminase defi- Kay, M.A., Manno, C.S., Ragni, M.V. et al. (2000). Evidence for gene
ciency. Blood 91: 30–36. transfer and expression of factor IX in haemophilia B patients treated
Ott, M.G., Schmidt, M., Schwarzwaelder, K. et al. (2006). Correction of with an AAV vector. Nat. Genet. 24: 257–261.
X-linked chronic granulomatous disease by gene therapy, augmented Manno, C.S., Pierce, G.F., Arruda, V.R. et al. (2006). Successful transduc-
by insertional activation of MDS1-EVI1, PRDM16 or SETBP1. Nat. tion of liver in hemophilia by AAV-factor IX and limitations imposed
Med. 12: 401–409. by the host immune response. Nat. Med. 12: 342–347.
Rill, D.R., Santana, V.M., Roberts, W.M. et al. (1994). Direct demon- Nathwani, A.C., Reiss, U.M., Tuddenham, E.G. et al. (2014). Long-term
stration that autologous bone marrow transplantation for solid safety and efficacy of factor IX gene therapy in hemophilia B. N. Engl.
tumors can return a multiplicity of tumorigenic cells. Blood 84: 380– J. Med. 371: 1994–2004.
383. Russell, S., Bennett, J., Wellman, J.A. et al. (2017). Efficacy and safety of
Sorrentino, B.P., Brandt, S.J., Bodine, D. et al. (1992). Selection of drug- voretigene neparvovec (AAV2-hRPE65v2) in patients with RPE65-
resistant bone marrow cells in vivo after retroviral transfer of human mediated inherited retinal dystrophy: a randomised, controlled,
MDR1. Science 257: 99–103. open-label, phase 3 trial. Lancet 390: 849–860.
Tebas, P., Stein, D., Tang, W.W. et al. (2014). Gene editing of CCR5 in
Lentiviral gene transfer autologous CD4 T cells of persons infected with HIV. N. Engl. J. Med.
370: 901–910.
Aiuti, A., Biasco, L., Scaramuzza, S. et al. (2013). Lentiviral hematopoi-
etic stem cell gene therapy in patients with Wiskott–Aldrich syn-
Genetic immunotherapy
drome. Science 341: 1233151.
Biffi, A., Bartolomae, C.C., Cesana, B. et al. (2011). Lentiviral vector Fesnak, A.D., June, C.H., and Levine, B.L. (2016). Engineered T cells: the
common integration sites in preclinical models and a clinical trial promise and challenges of cancer immunotherapy. Nat. Rev. Cancer
reflect a benign integration bias and not oncogenic selection. Blood 16: 566–581.
117: 5332–5339. Morgan, R.A., Dudley, M.E., Wunderlich, J.R. et al. (2006). Cancer
Biffi, A., Montini, E., Lorioli, L. et al. (2013). Lentiviral hematopoietic regression in patients after transfer of genetically engineered lympho-
stem cell gene therapy benefits metachromatic leukodystrophy. Sci- cytes. Science 314: 126–129.
ence 341: 1233158.
Brenner, M.K., Rill, D.R., Holladay, M.S. et al. (1993). Gene marking to
Methods to improve gene therapy safety,
determine whether autologous marrow infusion restores long term
haemopoiesis in cancer patients. Lancet 342: 1134–1137.
and suicide gene therapy
Brenner, M.K., Rill, D.R., Moen, R.C. et al. (1993). Gene-marking to Bonini, C., Ferrari, G., Verzeletti, S. et al. (1997). HSV-TK gene trans-
trace origin of relapse after autologous bone-marrow transplantation. fer into donor lymphocytes for control of allogeneic graft-versus-
Lancet 341: 85–86. leukemia. Science 276: 1719–1724.
Levine, B.L., Humeau, L.M., Boyer, J. et al. (2006). Gene transfer in Montini, E., Cesana, D., Schmidt, M. et al. (2006). Hematopoietic stem
humans using a conditionally replicating lentiviral vector. Proc. Natl. cell gene transfer in a tumor-prone mouse model uncovers low geno-
Acad. Sci. U.S.A. 103: 17372–17377. toxicity of lentiviral integration. Nat. Biotechnol. 24: 687–696.
Maude, S.L., Frey, N., Shaw, P.A. et al. (2014). Chimeric antigen recep- Sato, T., Neschadim, A., Konrad, M. et al. (2007). Engineered human
tor T cells for sustained remissions in leukemia. N. Engl. J. Med. 371: tmpk/AZT as a novel enzyme/prodrug axis for suicide gene therapy.
1507–1517. Mol. Ther. 15: 962–970.
Chapter 24 Pharmacogenomics
Leo Kager1 & William E. Evans2
1 Department of Pediatrics, St. Anna Children’s Hospital, Medical University, Vienna, Austria
2
St Jude Children’s Research Hospital, Memphis TN, USA

Introduction, 339 Pharmacogenomics to optimize oral antithrombotic therapy, 348


Principles of pharmacogenomics, 340 Summary and challenges for the future, 350
Pharmacogenomics to improve childhood ALL therapy, 342 Further reading, 351

monogenic loci for drug-metabolizing enzymes that strongly


Introduction influence the effects of medications. Interestingly, two
of the most important clinical examples of Mendelian
It has long been recognized that there is great heterogene-
pharmacogenetics – polymorphisms in the thiopurine
ity in the way people respond to medications. For exam-
S-methyltransferase (TPMT) gene that affect the efficacy
ple, when a standard dose of a certain drug is given to a
and toxicity of thiopurines, and polymorphisms in the
cohort of patients, some will respond and some will not
cytochrome P4502C9 (CYP2C9) and vitamin K epoxide
respond, some will respond only partially, and some will
reductase complex 1 (VKORC1) that affect the efficacy and
experience adverse drug reactions (ADRs) that can be life-
toxicity of warfarin – were again discovered in the field of
threatening. This variation in both host toxicity and treat-
hematology, namely in the treatment of acute lymphoblastic
ment efficacy can have many different causes, including envi-
leukemia (ALL) and oral anticoagulant therapy, respectively.
ronmental (e.g. nutrition, drug interactions), physiological
However, it is well recognized that most pharmacologi-
(e.g. age, gender, nutritional status, organ functions), patho-
cal effects result from the interplay of numerous gene prod-
physiological (e.g. pathogenesis and severity of the disease
ucts, and since the human genome has been sequenced and
being treated), and genetic factors. Overall, genetic factors
the human haplotypes of the most common form of genetic
are estimated to account for 15–30% of interindividual dif-
variation, namely single-nucleotide polymorphisms (SNPs),
ferences in drug metabolism and response. For certain drugs,
have been mapped, genome-wide approaches (i.e. pharma-
however, genetic factors are of the utmost importance and
cogenomics) are often used to elucidate the genomic con-
can account for up to 95% of interindividual variability in
tributors of variability in drug effects. The terms “pharmaco-
drug disposition and effects.
genetics” and “pharmacogenomics” are synonymous for all
The science of pharmacogenomics, which aims to define
practical purposes, and we herein use the term pharmacoge-
the genomic determinants for drug disposition and effects,
nomics. Besides pharmacogenomics, also the field of phar-
has a long tradition in the field of hematology. In the 1950s,
macoepigenomics, which focuses on the identification of
the relationship between hemolysis after antimalarial ther-
pharmacologically relevant epigenetic variants, has evolved.
apy and the inherited glucose-6-phosphate dehydrogenase
Recent advances in genome, transcriptome, and
(G6PD) activity in erythrocytes was identified. This discov-
epigenome interrogation technologies (e.g. arrays for
ery explained why the ADR of hemolysis is observed mainly
genome-wide SNP, mRNA, and DNA-methylation anal-
in Africans, where up to 10% of individuals are deficient in
yses, “next-generation” DNA sequencing technologies
G6PD, but is rarely seen in other ethnic groups like Euro-
like whole-exome sequencing – WES, coding regions and
peans, in whom G6PD deficiency is uncommon. In 1959,
“untranslated regions” only – and whole-genome sequenc-
Friedrich Vogel defined pharmacogenetics as “the study of
ing – WGS, coding and non-coding regions) and in silico
the role of genetics in drug response.” Until 2000, efforts
analytical approaches have become sufficiently robust
were mainly concentrated on mapping highly penetrant
and cost-effective and allow essentially agnostic genome-
wide investigations to identify pharmacologically relevant
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. relationships between genomic variants and well-defined
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. pharmacological endpoints, for example in genome-wide

339
340 Molecular Hematology

association studies (GWAS). Recent advances in single target or targets), which harbor germline and also acquired
cell RNA and DNA sequencing provide the technology to somatic variants. Many variants in the human genome and
interrogate the clonal nature of somatic variants, to identify in cancer cells have been identified that influence the expres-
sub-clones that may be resistant to cancer chemotherapy. sion and activity of pharmacologically relevant proteins.
Once discovered, such pharmacological relationships have
first to be validated and then translated into clinical practice,
Variation in the human genome
the latter of which is the most difficult task.
In this short chapter, validated, clinically relevant exam- The human genome consists of about 3 × 109 bp (3 Gb).
ples are presented to illustrate how pharmacogenomics can Although any two humans are thought to be up to 99.9%
be used to rationally improve current drug therapy in hema- identical in their DNA sequence, the remaining small frac-
tological diseases and to identify novel targets for developing tion of the genome, which constitutes the genetic diversity
new therapeutic approaches in hematological diseases. among individuals, contains many forms of variation, rang-
ing from large, microscopically visible chromosome anoma-
lies to single-nucleotide changes. Variants that are about
Principles of pharmacogenomics 3 Mb or more in size are referred to as microscopic vari-
ants. The smaller, and much more abundant, variants include
The effects of drugs are determined by the interplay of SNPs, small (<50 bp) insertions and deletions (indels) of
many gene products that influence the pharmacokinetics and nucleotides, variation in the number of repeats of a spe-
pharmacodynamics of medications. Whereas pharmacoki- cific motif (i.e. variable-number tandem repeats, minisatel-
netics describes the absorption, distribution, metabolism, lites, and microsatellites), duplications, and variants of DNA
and excretion of drugs (so-called ADME), pharmacodynam- segments 50 bp or larger that are collectively defined as
ics studies the relationship between the pharmacokinetic copy number variations (CNVs). Common DNA variants,
properties of drugs and their pharmacological effects, which often defined as greater than 1% in a given population,
can be desired or adverse. The ultimate goal of pharmacoge- are named polymorphisms, whereas rare DNA variants are
nomics is to elucidate functionally relevant genomic determi- named mutations.
nants for drug disposition (germline variants) and response
(variants in drug targets) to select medications and dosage
Single-nucleotide polymorphisms
of medications on the basis of each patient’s inherited ability
to metabolize, eliminate, and respond to specific drugs (Fig- The most common and most extensively studied inherited
ure 24.1). In hematological malignancies, for example, the genomic variations are SNPs, positions in the genome where
genomic architecture differs between the normal host cells individuals have inherited a different nucleotide. SNPs are
(i.e. germline) and the malignant cells (i.e. the therapeutic found approximately every 300 bp in both the coding and

DRUGS
(germline)
Host cells

GENOTYPE
Delivery

Absorption
Distribution
PK
Genomic variants

Single-nucleotide polymorphisms (SNPs) Metabolism


insertions/deletions (Indels <50 bp) Excretion
Varying number of tandem repeats (VNTR)
Copy number variants (CNV, <50 bp)
Epigenetic variants
Target Fig. 24.1 General principles of
PD Mechanism of action
Drug responses
pharmacogenomics. The goal of
pharmacogenomics is to elucidate genomic
Somatic variants
determinants that influence the pharmacokinetics
(PK) and pharmacodynamics (PD) of delivered
Target
cells

GENOTYPE Pharmacological effects drugs, thereby influencing both efficacious and


toxic effects. Investigating these functionally
EFFICACY TOXICITY important variants in the human population can
help to explain some differences in drug effects.
Pharmacogenomics 341

non-coding regions of the human genome. About 660 million common variants in the human genome (e.g. structural vari-
SNPs have been identified (of which about 130 million are ants), and the largest public catalogue of human variation and
currently validated) and are available in the public domain at genotype data is the database of the 1000 Genomes Project
http://www.ncbi.nlm.nih.gov/projects/SNP/snp_summary. (http://www.1000genomes.org/about).
cgi?view+summary=view+summary&build_id=151.
SNPs that cause amino acid changes in the encoded
Copy number variants
protein are named non-synonymous coding SNPs, whereas
SNPs that do not change the amino acid composition are DNA segments 50 bp or larger and which are present at vari-
named synonymous (or silent) SNPs. Amino acid substitu- able copy number in comparison with a reference genome are
tions have the potential to change the function of a protein, defined as CNVs. It is estimated that up to 9.5% of the genome
and have more relevance in functionally important domains contributes to CNVs (losses > gains); any variation in copy
of the protein (e.g. the catalytic cleft of an enzyme). However, number will affect a wide spectrum of genomic sequences,
it has been demonstrated for the adenosine triphosphate and possibly many pharmacologically relevant genes. CNVs
(ATP)-binding cassette transporter ABCB1 that a silent have been identified to influence the activity of major drug-
or synonymous SNP can also affect in vivo protein folding metabolizing enzymes like CYP isoenzymes and glutathione
and function. Moreover, SNPs in genomic regions that S-transferases. For example, fully functional gene duplication
influence the expression of functionally relevant genes and multiplication of the cytochrome P450 2D6 (CYP2D6),
like transcription factors or microRNA binding sites can which is responsible for the metabolism of more than 30%
also have pharmacological consequences. For example, a of all orally administered drugs (e.g. opioids, antidepressants,
so-called microRSNP (defined as a functional SNP that can etc.) can result in ultra-rapid drug metabolism and therapeu-
interfere with microRNA function and results in loss of the tic failure, or excessive response in patients.
microRNA-mediated regulation of a drug target gene) was One example, which illustrates the potential importance of
found to be associated with antifolate resistance by influ- CYP2D6 copy numbers on drug effects, is severe intoxication
encing the expression of the antifolate target dihydrofolate with codeine (which is often used to treat children suffering
reductase (DHFR) gene. In addition, a promoter polymor- from pain crisis in sickle cell disease, SCD), an analgesic pro-
phism in the gene encoding centrosomal protein 72kD drug that is metabolized by CYP2D6 to the active metabo-
(CEP72), which creates a binding site for a transcriptional lite morphine. High levels of morphine can be caused by
repressor, leading to lower expression of CEP72 mRNA, was ultra-rapid activation due to three copies of CYP2D6, exem-
found to be significantly associated with vincristine-induced plified by a woman who had taken codeine for pain shortly
peripheral neuropathy in children with ALL; and these after she had given birth. Whereas the woman survived the
results were recently confirmed in adults with ALL. intoxication, her child died from a lethal dose of morphine
via her breast milk. Though very important, analytical test-
ing for CNVs is non-trivial and more laborious than for
Haplotypes, linkage disequilibrium, and haplotype map
SNPs, and current efforts focus on the development of clini-
SNPs and other genomic variants are not inherited inde- cally useful assays for pharmacogenomic CNV analyses. The
pendently, but rather belong to segments of DNA that are major catalogues for CNVs are the Database of Genomic
inherited as units, with each unit referred to as a haplotype. Variants (http://dgv.tcag.ca/dgv/app/home) and the database
Genome-wide haplotypes can be constructed by linkage dis- of genomic structural variation (dbVar, http://www.ncbi.nlm.
equilibrium (LD) analysis, a statistical measure of the extent nih.gov/dbvar).
to which particular alleles or SNPs at two loci are associated
with each other in the population. LD occurs when haplotype
Somatic variations
combinations of alleles or SNPs at different loci occur more
frequently than would be expected from random association. Non-random genetic abnormalities, including gains and
SNPs and alleles of interest are presumably inherited together losses of chromosomes, can be found in the majority
if they are physically close to each other (typically 50 kb apart of hematological malignancies. This can create differences
or closer), producing strong LD. between normal host cells (i.e. germline genotype) and can-
The international HapMap consortium created a genome- cer cells (acquired somatic variants), and such differences can
wide map of haplotypes, which revealed a block-like struc- have pharmacologically relevant consequences. For example,
ture of LD, as well as the existence of areas of low or high resistance mechanisms against thiopurines, which are key
recombination rate, leading to the identification of so-called components in the successful therapy of childhood ALL, have
tagging (tag) SNPs. Tag SNPs can be used to predict with high recently been identified in ALL cells, and include somatic
probability the alleles at other cosegregating “tagged” SNPs. deletions of genes encoding proteins that regulate the sta-
It was also found that common SNPs are in LD with other bility of the DNA mismatch repair enzyme MSH2, acquired
342 Molecular Hematology

activating mutations in the genes encoding the cytosolic Approaches for establishing
nucleotidase NT5C2, and the purine biosynthesis enzyme pharmacogenomic models
phosphoribosyl pyrophosphate synthetase 1 (PRPS1). Col-
Pharmacogenomics is a broad strategy for establishing mod-
lectively, such findings can help to establish mechanistic
els by integrating information from functional genomics,
models for drug resistance in cancer cells, can help to pre-
high-throughput molecular analyses, pharmacokinetics, and
dict relapse (and to refine stratification of patients into dif-
pharmacodynamics. These models can be used to further
ferent treatment arms), and can be used to develop strategies
optimize existing drug therapy or identify novel therapeu-
to circumvent drug resistance mechanisms in hematological
tic targets. As already described, there is a bewildering
malignancies.
array of human genomic variation, and a central issue in
pharmacogenomics is to elucidate those genomic determi-
Epigenetic variations and epidrugs nants that are pharmacologically relevant, namely which
genomic variants are associated with certain drug-related
Besides genome sequence variation, epigenetic regulation of
phenotypes. Pharmacogenomic models can be established
gene expression is increasingly recognized to contribute to
using two broad strategies: candidate gene or agnostic
differences in the pharmacological effects of many medica-
genome-wide strategies. More details including the advan-
tions, referred to as pharmacoepigenomics. Epigenetic signal-
tages and disadvantages of these approaches are provided in
ing is mediated through DNA methylation, DNA hydrox-
Figure 24.2.
ymethylation, and post-translational histone modifications.
We herein provide selected examples of how candidate
Epigenetic biomarkers for drug responses have been identi-
gene and genome-wide approaches have been used to opti-
fied in the therapy of hematological malignancies. One exam-
mize therapies in hematological diseases, and focus on the
ple is somatic hypomethylation of the promoters of caspase 1
improvement of childhood ALL therapy and oral antithrom-
(CASP1) and its activator NLR family, pyrin domain contain-
botic therapies.
ing 3 (NLRP3) in ALL cells, leading to overexpression of both
genes, and higher caspase 1 activity in leukemia cells. This has
been shown to lead to glucocorticoid resistance in ALL cells,
due to caspase 1 cleavage of the glucocorticoid receptor. Pharmacogenomics to improve
Small molecules that interfere with the epigenetic control childhood ALL therapy
of gene expression, for example inhibitors of DNA methyl-
transferase (DNMT) or histone deacetylase (HDAC), are Childhood ALL is the most common cancer in children.
often referred to as “epidrugs.” Of note is that epidrugs were Insights from genome profiling and sequencing have
initially developed to treat hematological malignancies, and improved our understanding of ALL pathogenesis and have
the first US Food and Drug Administration (FDA) approved further proven that ALL is a heterogeneous and often poly-
epidrugs were the DNMT1 inhibitor vidaza for the treat- clonal disease. There are more and more ALL subtypes iden-
ment of myelodysplastic syndromes, and the HDAC inhibitor tified, each of which has distinctive somatic genomic alter-
vorinostat for the treatment of advanced cutaneous T-cell ations, which propel tumorigenesis and influence response to
lymphoma. antileukemic medications. Although about 85% of children
with ALL can be cured with current risk-adapted treatment
protocols in industrialized countries – stratification in these
Genetic variation among ethnic groups
protocols is based for example on the patient’s age (infants
It is well known that differences in the frequency and nature versus older), leukemia cell immunophenotype (B-lineage
of genetic variants among ethnic groups must also be recog- versus T-lineage), and genetics (“low-risk” chromosomal
nized when attempting to extrapolate research from one pop- abnormalities like t(12/21)/ETV6-RUNX1, high hyper-
ulation to another. For example, allele frequencies and types diploidy, t(1;19)/TCF3-PBX1, versus “high-risk” chromoso-
of polymorphisms in the TPMT and NUDT5 genes, which mal abnormalities like t(9;22)/BCR-ABL1, low hypodiploidy,
influence the efficacy and toxicity of thiopurine therapy, vary t(17;19)/TCF3-HLF, etc.), as well as response to therapy; and
greatly among different ethnic groups. Whereas pharmaco- patients are stratified based on their risk of relapse into differ-
logically relevant variants in TPMT are frequent in Euro- ent regimens like “low-risk” or “high-risk” regimens – ALL
peans and Africans, these variants are rare in Asians. Con- remains a leading cause of death from disease in children.
versely, actionable NUDT15 variants are frequently found in Major causes of death in childhood ALL are treatment fail-
East Asians and Hispanics, but they are rare in Europeans and ure (i.e. relapse or less frequent, progressive disease during
not observed in Africans. Therefore, pharmacogenomic rela- therapy) and ADRs, like bone marrow toxicity-associated
tions must be validated for each therapeutic indication within lethal infections. The significant reduction in treatment
different ethnic groups. failures during the last decades, which resulted in significant
Pharmacogenomics 343

Single-gene approach Pathway-gene approach

Drug-related
• Analyzes the possible phenotype • Analyzes several
candidate gene functionally related
candidate genes
• Requires a large
cohort to test the • Requires a smaller
association cohort to test the
Genome-wide approach association
− Carries the risk of not
finding an association − Carries the risk of
missing important
• Analyzes the whole genome genes
+ Provides proof of
(expression and SNP)
principle if an
association is found + Provides a more
• Requires smaller cohorts to test biologically meaningful
associations association

− Difficult assessment of biological


meaning (i.e. risk of false positives)

+ May identify new associations


Fig. 24.2 Comparison of single-gene, candidate pathway-gene, and genome-wide pharmacogenomic approaches to the analysis of
drug-related phenotypes. The main characteristics (•), disadvantage (−), and advantage (+) of each approach are indicated. The arrows between
the three approaches indicate that any approach can lead to another and that a combination of approaches might be considered. SNP,
single-nucleotide polymorphism.
Source: Cheok M.H., Evans W.E. (2006). Acute lymphoblastic leukaemia: a model for the pharmacogenomics of cancer therapy. Nature Reviews.
Cancer 6: 117–129. Reproduced by permission of Springer Nature.

improvements in outcomes, was mainly achieved by shifting to develop strategies to avoid the potentially lethal ADR
a higher proportion of patients into more intensive treatment leukopenia in “at-risk” patients treated with thiopurines. Per-
arms (i.e. “high-risk” ALL therapies) based on novel iden- tinent information on other germline genetic variants, which
tified “high-risk” profiles, such as minimal residual disease have been reported to be associated with ADRs of ALL
(MRD) load on day 15 of induction therapy in recent Berlin– medicines, including the non-lethal ADRs glucocorticoid-
Frankfurt–Münster (BFM) protocols. As a higher proportion induced osteonecrosis and vincristine-induced neuropathy,
of patients had received more intensive therapies in recently are summarized in Table 24.1. We furthermore provide
completed treatment trials, for example the Associazione examples on how genome-wide interrogation investigations
Italiana di Ematologia e Oncologia Pediatrica (AIOP) and have helped to identify thiopurine resistance mechanisms
BFM group’s childhood AIOP-BFM ALL 2009 protocol, the (somatic variants in NT5C2 and PRPS1) in ALL cells from
number of deaths due to ADRs have increased. Therefore, relapse, and how this information can be used to develop
there is an urgent need to identify those patients who are at strategies to overcome these drug resistance mechanisms.
high risk for lethal ADRs or for death due to treatment fail- In addition, we describe how an integrated drug response
ure, in order to tailor their supportive and/or antileukemia profiling approach has been successfully used to identify
therapy accordingly, or to develop novel therapeutic novel therapies for patients with TCF3-HLF-positive ALL, a
strategies and design novel innovative treatment protocols. rare lethal subtype when treated with current treatment pro-
We herein provide selected clinically relevant examples tocols; and we have summarized in Table 24.2 the results
to demonstrate the potential of pharmacogenomics in this of pharmacogenomic investigations, which have identified
context. We describe how testing for G6PD deficiency can potential actionable somatic genomic variants in patients
help to prevent the occurrence of potentially lethal hemoly- with other “high-risk” ALL subtypes; that is, mixed-lineage
sis after administration of rasburicase (which is the standard leukemia (MLL)-rearranged infant ALL, t(9;22)/BCR-ABL
medication to treat hyperuricemia), and how results from ALL, and “BCR-ABL1-like” ALL (which is a subgroup of ALL
candidate gene (variants in TPMT) and genome-wide inves- that has a similar gene expression signature and outcome
tigations (which identified functionally relevant variants in as t(9;22)/BCR-ABL1 ALL, but lacks the t(9;22)/BCR-ABL1
NUDT15) have provided important insights that can be used aberration).
344 Molecular Hematology

Table 24.1 Selected examples of germline genetic variants (for TPMT and NUDT15 see text) associated with adverse drug reactions (ADRs) of
acute lymphoblastic leukemia medications, identified via genome-wide association studies

Gene (variant SNP ID) and function(s)


of the encoded protein Drug(s) Possible mechanism(s) of action ADR

SLCO1B1 (rs11045879), solute carrier Methotrexate (MTX) MTX transporter, altered MTX Gastrointestinal
organic anion transporter family clearance toxicity
member 1B1, drug transporter
Near the GRIN3A locus (rs10989692), Drugs that contribute Glutamate is involved in osteoblast Osteonecrosis in
glutamate ionotropic receptor NMDA to osteonecrosis activation and impairs endothelial children and
type subunit 3A, glutamate regulated (dexamethasone, barrier function adults
ion channels asparaginase, MTX)
BMP7 (rs75161997), bone morphogenic Drugs that contribute Variants in BMP7 may alter bone Osteonecrosis in
protein 7, member of the to osteonecrosis metabolism and formation; BMP7 is children
transforming growth factor-beta (dexamethasone, also toxic to smooth muscles; may <10 years
family of proteins, plays a role in bone, asparaginase, MTX) induce vascular injury in bone
kidney, and brown adipose tissue vasculature
development and bone homeostasis
CEP72 (rs924607), centrosomal protein Vincristine (VCR) rs924607 T promotor variant creates VCR neuropathy
72kD, involved in the recruitment of a binding site for a transcription
key centrosomal proteins to the repressor, leading to lower CEP72
centrosome and microtubule mRNA expression
formation
HAS3, (rs2232228), hyaluron synthase 3, Anthracyclines Hyaluran is enriched in tissues Cardiotoxicity
enzyme that produces undergoing remodeling after
low-molecular-weight hyaluran injury – impaired remodeling after
cardiac injury
CPA2 (rs199695765), carboxypeptidase Asparaginase rs199695765, non-sense variant; Pancreatitis
A2, secreted pancreatic enzyme loss-of-function variants in CPA1
were found to be associated with a
risk for chronic pancreatitis

SNP, single-nucleotide polymorphism.

Table 24.2 “High-risk” acute lymphoblastic leukemia (ALL) subtypes and selected examples of druggable targets that have been identified via
pharmacogenomics investigations

ALL subtype Identified drug targets Drugs

MLL-rearranged ALL (affects mainly infants) FLT3 was found to be overexpressed in MLL-rearranged ALL FLT3 inhibitor: lestaurtinib
t(9;22)/BCR-ABL1 or Ph-ALL (more frequent Chimeric fusion protein BCR-ABL1 has tyrosine kinase TKIs (ABL1 inhibitors):
in adolescents and adults) activity, and can be selectively blocked via TKIs imatinib, dasatinib
“BCR-ABL1-like” ALL, exhibits a gene Genetic key subgroups with therapeutic implications have
expression profile similar to that of been identified:
a. TKIs: imatinib, dasatinib
Ph-ALL but lacks the BCR-ABL1 fusion a. ABL-class rearrangements targeting ABL1, ABL2, CSF1R,
b. JAK inhibitor: ruxolitinib
protein and PDGFRB, which are sensitive to imatinib and
dasatinib
b. JAK2 fusions, EPOR rearrangements, and IL7R/SH2B3
alterations are sensitive to JAK inhibitors

ABL, ABL proto-oncogene 1; CSF1R, colony stimulating factor 1 receptor; EPOR, erythropoietin receptor; FLT3, FLT3 Fms-Related Tyrosine Kinase
3; IL7R, interleukin 7 receptor; JAK2, Janus kinase 2; MLL, mixed-lineage leukemia; PDGFRB, platelet-derived growth factor receptor beta; Ph,
Philadelphia; SH2B3, SH2B adaptor protein 3; TKI, tyrosine kinase inhibitor; VEGFR2, vascular endothelial growth factor receptor 2.
Pharmacogenomics 345

Rasburicase and G6PD deficiency competition with direct drug inactivation (S-methylation)
via TPMT and (hydrolysis of the triphosphates TGTP
Tumor lysis syndrome (TLS) is the most common disease-
and TdGTP to monophosphates TGMP and TdGMP) via
related emergency in hematological cancers. TLS occurs
NUDT15. TPMT and NUDT15 activities determine how
when tumor cells release their contents into the blood-
much thiopurine is inactivated and how much remains for
stream (either spontaneously or due to therapy), and this
incorporation into DNA.
causes electrolyte and metabolic disturbances (i.e. hyper-
uricemia, hyperkalemia, etc.), which, untreated, will result
in renal insufficiency, cardiac arrhythmias, and ultimately Variants in TPMT
death due to multiorgan failure. Hyperuricemia, which con-
Variations in TPMT activity are regulated primarily by
tributes to TLS, can be prevented/treated via rasburicase, an
variants in the TPMT gene. Three non-synonymous SNPs
enzyme which catalyzes the cleavage of uric acid to hydro-
account for more than 95% of the relevant TPMT vari-
gen peroxide, and which is then further metabolized via glu-
ants, namely TPMT*2 (dbSNP identification number rs
tathione peroxidase (GSHPX). In patients with G6PD defi-
1800462, nucleotide change c.238G > C, hereafter referred to
ciency, however, the activity of GSHPX is markedly reduced
as amino acid change p.Ala80Pro), TPMT*3C (p.Tyr240Cys),
in red blood cells (RBCs), and treatment with rasburicase
and TPMT*3A (p.Ala154Thr + p.Tyr240Cys). One in 300
results in oxidative damage to RBCs (i.e. acute hemolytic ane-
persons carries two variant TPMT alleles and does not
mia, AHA). Rasburicase administration is an essential ele-
express functional TPMT activity (due to lesser stabil-
ment of supportive therapy in children with ALL at diagnosis
ity of the variant protein); about 5–10% are heterozygous
and during initial induction therapy.
and have intermediate levels of enzyme activity, whereas
However, fatalities have been reported after rasburicase
95% of individuals are homozygous for the wild-type allele
administration in ALL patients with G6PD deficiency. G6PD
(TPMT*1/TPMT*1) and have normal TPMT activity (Fig-
deficiency is a very common disorder, and about 5% of the
ure 24.3). Population studies have shown significant differ-
world population (highest incidence in the subtropical girdle
ences in TPMT pharmacogenetics among ethnic groups. For
with high malaria prevalence) is affected. As of August 2016,
example, TPMT*3A is the most common variant allele in
about 180 functionally relevant variants had been identified
Europeans, whereas TMPT*3C accounts for more than 50%
in the G6PD gene, most of which are missense mutations
of variants in Africans. TMPT*3C is also the major variant
that alter G6PD stability. Complete loss of G6PD is lethal;
allele in East Asian populations, which generally lack the
and only a few rare, complex variants that cause significant
TPMT*3A allele.
reduction in enzyme activity result in severe transfusion-
Patients who carry two non-functional TPMT alleles expe-
dependent chronic non-spherocytic hemolytic anemia. As
rience severe hematotoxicity if treated with conventional
rasburicase administration can cause lethal ADRs, the use
doses of thiopurines. Depending on the dose (e.g. 60 mg/m2
of this drug has been contraindicated by the FDA and the
daily) and the comedications, many patients with one non-
European Medicines Agency (EMA) in patients with G6PD
functional TPMT allele can tolerate MP therapy at full doses.
deficiency. More details, including information on G6PD
However, these patients might be at higher risk of dose-
genetic and activity tests, as well as genetic variant classifica-
limiting hematotoxicity with slightly higher mercaptopurine
tion (classes I–IV), are available at the Clinical Pharmacoge-
doses (e.g. 75 mg/m2 daily), but may experience better
netics Implementation Consortium (CPIC) guideline website
leukemia control than do those who have two wild-type
(https://www.pharmgkb.org/molecule/PA10176).
TPMT alleles. Moreover, patients with one non-functional
TPMT allele may be at increased risk of epipodophyllotoxin-
related acute myeloid leukemia and irradiation-induced
Optimization of thiopurine therapy
brain tumors as a result of thiopurine therapy, and of veno-
The thiopurine antimetabolites mercaptopurine (MP) and occlusive disease of the liver following thioguanine therapy.
thioguanine (TG) are essential components in childhood Most importantly, results from the St Jude Children’s
ALL treatment protocols. Thiopurines have narrow thera- Research Hospital (SJCRH) Total XIIIB childhood ALL
peutic indices, which explain the frequently observed toxici- treatment trial provide proof of principle that prospective
ties, including severe myelosuppression (mainly leukopenia) MP dose adjustment based on TPMT genotypes can decrease
with the risk of life-threatening infections. To exert cytotox- toxicity without a compromise in treatment efficacy. Based
icity, the prodrugs MP and TG have to undergo anabolism on an FDA advisory committee recommendation, a change
to form active cytotoxic thioguanine nucleotides (TGNs). in labeling for MP, with TPMT testing and dosage rec-
These multistep anabolic reactions, which include conver- ommendations provided for TPMT-deficient patients, was
sion into thioguanosine triphosphate (TGTP) and reduc- implemented in 2004, and more details on TPMT-guided
tion to deoxythioguanosine triphosphate (TdGTP), are in preemptive thiopurine dose adjustment, which is considered
346 Molecular Hematology

100 TGN
(HPRT) wt/wt
(active)
80 MP Inherited
Percent of population

MeMP differences in
(TPMT) (inactive) metabolism
60

40

20
wt/var
var/var
0
5 10 15 20 25 30
(a) Fig. 24.3 Thiopurine methyltransferase
TPMT activity
(TPMT). (a) Mercaptopurine (MP) can be activated
Protein to thioguanine nucleotides (TGN) by hypoxanthine
(b) level
phosphoribosyltransferase (HPRT) or inactivated to
methylmercaptopurine (MeMP) via TPMT. The
genetic polymorphism in TPMT results in a trimodal
population frequency distribution in TPMT activity,
TGN concentration

Inherited differences with deficient activity caused by inheritance of two


2000 in drug levels variant (var) alleles, intermediate activity caused by
heterozygosity, and high activity associated with
homozygote wild-type (wt) genotypes. (b) TPMT
activity is directly proportional to the amount of
1000
TPMT protein and (c) is inversely related to
intracellular concentrations of active TGN
metabolites in hematopoietic cells following MP
(c) var/var wt/var wt/wt
therapy. (d) The biochemical basis for low protein
Toxicity Risk of relapse conferred by the most common var. polymorphism
100 (719A → G) is illustrated by the longer half-life for
Percent remaining

t1/2 = 18 h in vitro expressed wt TPMT when compared with


719 wt the rapidly degraded var protein.
10 Source: Jones T.S., Yang W., Evans W.E., Relling
M.V. (2007). Using HapMap tools in
t1/2 = 0.25 h
719 var pharmacogenomic discovery: the thiopurine
1 methyltransferase polymorphism. Clinical
0 5 10 15 20 25 Pharmacology and Therapeutics 81: 729–734.
(d) Time (h) Reproduced by permission of John Wiley & Sons.

a prototype of pharmacogenomic precision medicine thiopurine-induced myelotoxicity, however, is considerably


approaches, is available at the CPIC guideline website higher in Asians. In order to identify other genetic variants
(https://www.pharmgkb.org/guideline/PA166104945). that contribute to thiopurine-induced myelotoxicity in
Patients who share the same TPMT genotypes still exhibit Asians, a case–control GWAS study was performed in
considerable variations in their response to thiopurines, and Korean patients who were treated with thiopurines, used as
other genetic variations that contribute to the toxicity and an immunosuppressant, for chronic inflammatory bowel dis-
response to thiopurines have recently been identified via ease. This research identified a non-synonymous SNP in the
GWAS studies. The major finding was the identification NUDT15 gene (rs116855232, c.415C>T, hereafter referred to
of four loss-of-function germline NUDT15 coding variants, as p.Arg139Cys) that encodes nudix hydrolase 15 (or nucleo-
which play an important role in thiopurine intolerance, espe- side diphosphate-linked moiety X-type motif 15), which was
cially in East Asian populations. strongly associated with thiopurine-induced leukopenia.
Researchers from the SJCRH and from the Children’s
Oncology Group (COG) confirmed the importance of this
Variants in NUDT15
variant to causing MP intolerance in a GWAS study on
It is well known that the frequency of TPMT deficiency more than 1000 children treated for ALL. In addition,
causing variants is lower in Asians compared to individuals it was confirmed that NUDT15 deficiency (low- or
of European descent (~3% versus ~10%). The frequency of intermediate-activity diplotypes) was most common in
Pharmacogenomics 347

East Asians (22.6%) and Hispanics (for example, 21.2% in variant proteins. In vitro experiments confirmed that variants
Peruvians), rare in Europeans, and not observed in Africans. with high NT5C2 activity conferred resistance against thiop-
Subsequent targeted sequencing of NUDT15 identified a urines, but not against other antileukemic agents. Ultra-deep
total of four functionally relevant loss-of-function cod- sequencing revealed that in 2 out of 7 patients, the minor
ing variants (p.Arg139Cys, p.Arg139His, pVal18Ile, and mutant clone already existed at ALL diagnosis. It is assumed
p.Val18_Val19insGlyVal) that were associated with thiop- that NT5C2 gain-of-function harboring clones exist at
urine intolerance. Five NUDT15 haplotypes (*1–*5) were diagnosis or evolve early during therapy, and can undergo
inferred from distinct combinations of these four variants, selective outgrowth in treatment phases, when thiopurines
and functional analyses showed that NUDT15 deficiency play a major role, like in oral maintenance therapy, which is
results in excessive levels of active thiopurine metabo- given for >1 year as a final element of ALL therapy. Indeed,
lites and host toxicity. Of note is that ALL cells carrying there was a significant association between activating
intermediate-activity NUDT15 diplotypes were shown to NT5C2 variants and early ALL relapse (within 36 months
be significantly more sensitive to thiopurines compared to from diagnosis); and this early outgrowth of resistant clones
NUDT15 wild-type diplotypes. Therefore, thiopurine dose during or shortly after completion of maintenance therapy
reduction in NUDT15-deficient patients, in order to avoid supports this assumption. Strategies to overcome the drug
the ADR leukopenia, will most likely not result in a compro- resistance somatic phenotype gain of function in NT5C2
mise of therapeutic effect, because NUDT15-deficient ALL can focus either on the inhibition of NT5C2 via small
cells are more sensitive to thiopurines. Prospective clinical molecules, or on the use of drugs that are not inactivated
trials, however, are necessary to prove this hypothesis. To via NT5C2.
further personalize thiopurine therapy in different ethnic
populations, a polygenic dosing algorithm that incorporates
Variants in PRPS1
both TPMT and NUDT15 variants should be implemented.
Functionally relevant gain-of-function mutations in another
mediator of purine metabolism, PRPS1, were recently iden-
Relapsed ALL, thiopurine resistance, and
tified in almost 7% of relapsed BCP-ALL samples. PRPS1
variants in NT5C2 and PRPS1
encodes phosphoribosyl pyrophosphate synthetase 1, which
Up to 15% of children with ALL experience disease recur- is a key enzyme in the de novo purine synthesis (DNPS) path-
rence, and unfortunately most of them will die. Identification way. In contrast to NT5C2 mutations, ultra-deep sequencing
of the mechanisms of drug failure in resistant ALL clones of serial bone marrow samples revealed that PRPS1 muta-
is urgently needed, in order to develop strategies to over- tions were not detectable at ALL diagnosis, but increased
come these resistance mechanisms. Genomic profiling and exponentially before clinical relapse, which occurred early in
sequencing investigations of ALL samples obtained from treatment. Expression of gain-of-function variants in PRPS1
diagnosis, remission, and relapse have already helped to resulted in resistance to thiopurine-induced apoptosis. Func-
identify different pathways (e.g. transcription regulation, tional investigations revealed that the drug resistance PRPS1
lymphoid development, Janus kinase and Ras signaling, variants showed defective feedback inhibition to adenosine-
cell cycle regulation, tumor suppression, mismatch repair, and guanosine-diphosphates, which allowed for continuous
epigenetic regulation, and nucleotide metabolism), in which activation of DNPS, which in turn results in an increase
somatic mutations are enriched at relapse. We herein focus of intracellular hypoxanthine levels, which inhibit the acti-
on mutations in the nucleotide metabolism pathway and vation of the thiopurine prodrugs (MP and TG) into their
provide two examples of functionally relevant variants in active metabolites. These findings demonstrate that the rare
key genes of nucleotide metabolism (i.e. NT5C2 and PSPR1) acquired somatic PRPS1 variants in ALL relapse clones can
that have recently been identified to play a role in resistance drive thiopurine resistance via altering the feedback inhi-
to thiopurines and development of relapse in a subset of bition of nucleotide synthesis and competitive inhibition
children with ALL. of bioactivation of the prodrugs MP and TG. One logi-
cal strategy to overcome this drug resistance mechanism
would be the inhibition of the continuously activated DNPS
Variants in NT5C2
pathway. Indeed, inhibition of GART (phosphoribosyl-
Two groups of researchers found that up to 10% of B-cell glycinamide formyltransferase), another important enzyme
precursor (BCP)-ALL and up to 20% of T-ALL relapse of DNPS, via the small molecule inhibitor lometrexol,
samples harbor somatic gain-of-function variants in the reversed thiopurine drug resistance in PRPS1-mutant cells
gene NT5C2. NT5C2 encodes a 5′ nucleotidase, which can in vitro.
selectively inactivate thiopurines, thereby protecting ALL Clearly, much work is needed to further elucidate the
cells against thiopurine-induced apoptosis. NT5C2 enzyme genomic drivers for treatment failure in ALL. The examples
activity was measured to be increased up to 48-fold in the of NT5C2 and PRPS1, however, provide evidence that in
348 Molecular Hematology

the near future the routine use of genomic interrogation 6.7 million deaths, coronary heart disease: 7.4 million deaths;
techniques could help to identify patients with emergent http://www.who.int/mediacentre/factsheets/fs317/en). Arte-
relapse at an early stage, and this may allow the tailoring rial or venous thrombotic events are major fatal complica-
of therapy accordingly, e.g. by drugging DNPS in clones tions of CVDs, and can be prevented via antithrombotic med-
that develop resistance to thiopurines via PRPS1-mediated ications, like oral anticoagulants and antiplatelet therapies.
DNPS activation.

Coumarins and variants in CYP2C9


Optimization of therapy in children with and VKORC1
TCF3-HLF ALL
Although treatment trials demonstrated a favorable effi-
One typical feature of childhood ALL is the subtype-defining cacy and safety profile of novel (non-vitamin K antagonist)
presence of chromosomal aberrations like translocations oral anticoagulants, which either directly inhibit thrombin
and gross aneuploidies. Translocations, such as t(12;21), (e.g. dabigatran) or factor Xa (e.g. rivaroxaban, apixaban,
t(9;22), t(1;19), or t(17;19) and the corresponding gene and edoxaban), millions of patients still take coumarins to
fusions ETV6-RUNX1, BCR-ABL1, TCF3-PBX1, or TCF3- prevent thromboembolic events in chronic conditions such
HLF, which mainly involve genes that play an important role as atrial fibrillation, deep venous thrombosis, pulmonary
in hematopoietic development, are considered to be primary emboli, acute myocardial infarction, stroke, and disease
ALL-initiating abnormalities, which comprise the leukemic and/or replacement of heart valves. The oral anticoagulants
clone and define subtype biology. These initiating lesions of the coumarin type, warfarin (used in the UK and the USA),
typically cooperate with somatic secondary abnormalities, acenocoumarol, and phenprocoumon (preferentially used in
which often involve B-cell development genes like IKAROS continental Europe), have similar pharmacodynamic prop-
zinc finger 1 (IKZF1) or paired box 5 (PAX5), which are erties but differences in half-life, and act by inhibiting the
present only in ALL subclones. activation of vitamin K–associated clotting factors. A very
Two ALL subtypes have chimeric fusion genes that narrow therapeutic index, with risk of serious hemorrhage
involve transcription factor 3 (TCF3); namely, ALL with if overcoagulated and thrombosis if undercoagulated and
t(1;19)/TCF3-PBX1 (~5% of ALL patients) and ALL with interindividual variability in response to coumarins, neces-
t(17;19)/TCF3-HLF (~1% of ALL patients). Whereas chil- sitates individualization of treatment, which is based primar-
dren with TCF3-PBX1-positive ALL have excellent outcomes ily on monitoring prothrombin time and calculation of the
with contemporary BFM and SJCRH Total therapies, all so far International Normalized Ratio (INR). Whereas INR is help-
reported children with TCF3-HLF-positive ALL have experi- ful in tailoring coumarin maintenance therapy, prospective
enced early disease relapse and died. In an attempt to iden- studies have identified coumarin induction therapy as the
tify curative treatment options for children with TCF3-HLF- period when the INR is most likely to be out of range and
positive ALL, an international collaborative study group when the rate of iatrogenic ADRs is greatest.
set up a series of genomic and functional studies, includ- Many factors have been identified as affecting the degree
ing an integrated drug–response profiling approach. They of anticoagulation achieved by coumarins, including patient
first identified that the chimeric fusion protein TCF3-HLF age (lower dose requirement in the elderly), gender, body
promotes cellular transcriptional reprogramming toward a size, ethnicity, diet (particularly vitamin K intake), cigarette
drug-resistant immature state. Subsequently, they found that smoking, disease (e.g. liver diseases), and coadministration
TCF3-HLF xenografts are highly sensitive to the BCL2 of other drugs (particularly those which inhibit the activity
(B-cell CLL/lymphoma 2)-targeting drug venetoclax. Com- of CYP2C9). Polymorphisms in genes that affect the phar-
bination of venetoclax with conventional chemotherapy macokinetics (CYP2C9, and to a lesser extent other CYP
induced durable remissions in patient-derived xenografts; enzymes) and pharmacodynamics (VKORC1) of coumarins,
and these results from preclinical investigations hold great however, have been shown to act as major determinants of
promise to offer a chance for a cure for patients with TCF3- coumarin dosage requirements.
HLF-positive ALL in future international clinical trials that Coumarins are a racemic mixture of r and s enantiomers
combine venetoclax and conventional chemotherapy. that differ in their patterns of metabolism and in their
potency of pharmacodynamic effect. For example, it has been
suggested that s-warfarin accounts for up to 70% of the over-
Pharmacogenomics to optimize oral all anticoagulation response of warfarin. After oral adminis-
antithrombotic therapy tration, warfarin is completely absorbed and bound to albu-
min (99%) in plasma. Free warfarin is taken up into liver cells,
Cardiovascular diseases (CVDs) are the most common cause where it is biologically active and either inhibits VKORC1 or
of death, accounting for ~17.5 million deaths in 2012 (stroke: is catabolized by cytochrome P450 isoenzymes (Figure 24.4).
Pharmacogenomics 349

Target Metabolism

Epoxide
Warfarin
reductase
R-warfarin
VKORC1
S-warfarin
CYP1A2
Fig. 24.4 Mechanism of action of warfarin. The Cytochrome
Reduced vitamin K Vitamin K epoxide CYP2C9
racemic mixture of r- and the more potent P450 CYP3A4
s-warfarin inhibits the reductase in the vitamin K
cycle, impairing the synthesis of active vitamin
K–dependent clotting factors in liver cells and S-OH-warfarin R-OH-warfarin
Carboxylase
causing bleeding. The cytochrome P450 isoenzyme
CYP2C9 (and to a lesser extent CYP3A4 and
CYP1A2) and vitamin K epoxide reductase complex
1 (VKORC1) genotypes influence warfarin dose Glutamic acid Gamma-Carboxyglutamic acid
requirement. Active blood clotting proteins

Variants in CYP2C9 VKORC1*1, the low-dose coumarin haplotype VKORC1*2,


and the high-dose coumarin haplotypes VKORC1*3 and
A number of CYP isoforms contribute to warfarin
VKORC1*4. The significantly higher average warfarin
metabolism; however, hydroxylation by CYP2C9 is the
requirement in Africans is in line with the significantly lower
most important inactivation pathway of the pharmacolog-
occurrence of the low-dose coumarin VKORC1*2 haplotype
ically more relevant s-warfarin. CYP2C9 is the principal
in Africans. Overall, the hereditary pharmacodynamic
CYP2C isoenzyme in the human liver, and it is involved
factor VKORC1 may explain about 25% of the variance in
in the oxidative metabolism of several clinically important
coumarin dose requirement, compared with 5–10% for the
medications, including oral anticoagulants, phenytoin, and
hereditary pharmacokinetic factor CYP2C9 alone.
various non-steroidal anti-inflammatory drugs. Numerous
CYP2C9 and VKORC1 genotypes have been incorporated
polymorphic alleles (CYP2C9*1 to CYP2C9*60) have been
into dosing algorithms in order to estimate the appropriate
identified for the known CYP2C9 gene, according to The
coumarin starting dose, and in 2010 the FDA updated the
Human Cytochrome P450 (CYP) Allele Nomenclature
warfarin drug label and suggested that VKORC1 and CYP2C9
Database (https://www.pharmvar.org/htdocs/archive/index
genotypes should be taken into consideration when the drug
original.htm), at least half of which are associated with
is prescribed. Dosing algorithms and more details are avail-
diminished enzyme activity. The two most common
able at https://www.pharmgkb.org/guideline/PA166104949
CYP2C9 variants are CYP2C9*2 and CYP2C9*3. As with
and www.warfarindosing.org. Randomized controlled
most polymorphisms, there are differences in the frequency
clinical trials, such as the European Pharmacogenetics
of polymorphic CYP2C9 alleles among different ethnic
of Anticoagulant Therapy (EU-PACT) and the US Clar-
groups. In Europeans, the overall allelic frequency of
ification of Optimal Anticoagulation through Genetics
CYP2C9*2 is about 10–20%, and that of CYP2C9*3 is about
(COAG) trials, have investigated the potential benefit of
5–10%. The *2 and *3 variants are very rare in African Amer-
genotype-based strategies for initiating coumarin therapy.
icans and Asians; 95% of these persons express the wild-type
Although the use of genotype-based algorithms has resulted
genotype *1/*1 (i.e. extensive metabolizers). Compared with
in a greater percentage of time in therapeutic range than
the wild-type enzyme activity of CYP2C9*1, the enzyme
standard-fixed dosing in the EU-PACT trial, no reductions
activity of the CYP2C9*2 variant is reduced by about 30–50%,
of severe ADRs like stroke or bleeding were reported with the
and the CYP2C9*3 variant activity is reduced by 90% in vitro.
pharmacogenomic dosing. These trials, however, were not
powered for the pharmacodynamics endpoints bleeding or
Variants in VKORC1
thromboembolic events, and the Genetics Informatics Trial
VKORC1 regenerates reduced vitamin K for another cycle (GIFT) of Warfarin to Prevent Deep Venous Thrombosis
of catalysis, essential for the γ-carboxylation of the vitamin is investigating this topic. Of note is that recently published
K–dependent clotting factors II, VII, IX, and X (Figure 24.4). results from the Effective Anticoagulation with Factor Xa
The identification of common variants in VKORC1 has Next Generation in Atrial Fibrillation–Thrombolysis in
emerged as one of the most important genetic factors Myocardial Infarction 48 (ENGAGE AF-TIMI 48) trial
determining coumarin dose requirements. Main VKORC1 (which enrolled more than 14 000 patients with atrial
haplotypes include the reference haplotype (wild-type) fibrillation) provided evidence that testing for VKORC1 and
350 Molecular Hematology

CYP2C9 was able to identify patients who are more likely clopidogrel recommends an alternative antiplatelet therapy
to bleed with warfarin therapy. This information could be (e.g. prasugrel, ticagrelor) for CYP2C19 poor (two loss-of-
used to optimize choice of oral anticoagulant therapy; that function alleles) or intermediate (one loss-of-function allele)
is, to use edoxaban in patients who are sensitive (typically metabolizers if there is no contraindication.
1–2 variant alleles) or highly sensitive (typically 3–4 variant
alleles) to warfarin.
Summary and challenges for
the future
Clopidogrel and variants in CYP2C19
Platelets play a crucial role in thrombosis and the devel- As there are numerous non-genetic factors that influence
opment of acute coronary syndromes (ACS) because a drug effects (e.g. compliance, nutritional factors, concurrent
platelet-rich thrombus forms at the site of the ruptured medications), it is clear that pharmacogenomics will never
atherosclerotic plaque. Thus, inhibition of platelet function explain all interindividual variability in drug effects. How-
is an effective strategy in the treatment and prevention of ever, there is clear evidence that pharmacogenomic mod-
thrombosis, especially after percutaneous coronary inter- els can help to improve drug treatment in hematology by
ventions (PCIs). The main classes of antiplatelet agents facilitating appropriate dose individualization and optimal
include aspirin, the thienopyridines (clopidogrel and pra- treatment selection. For example, clinically actionable vari-
sugrel), the nonthienopyridine P2Y purinergic receptor 12 ants have been identified in the TPMT, NUDT15, CYP2C9,
(P2Y12) antagonists (ticagrelor), and intravenous GPIIb/IIIa CYP2C19, and VCORC1 genes, and the TPMT/NUDT15 and
antagonists. Dual platelet inhibition via aspirin and P2Y12 CYP2C9/VCORC1 as well as the CYP2C19 models can be
receptor antagonists is the guideline-approved standard of used to individualize thiopurine and antithrombotic therapy
care in patients with ACS and PCI with stenting. The newer a priori, thereby reducing the risk of severe ADRs.
P2Y12 receptor antagonists prasugrel and ticagrelor have As of 2016, clinically actionable genomic variants had
superior efficacy compared with clopidogrel, but have a been identified in approximately 20 genes that affect about 80
higher risk for bleeding and are more expensive. drugs. However, some barriers (e.g. lack of studies that prove
Clopidogrel is an orally administered prodrug and the the cost/benefit, lack of financial reimbursement, etc.) still
response to it is heterogeneous (up to 20% of treated patients prevent the widespread use of pre-emptive genetic testing
do not respond to clopidogrel and are at risk for stent (ideally by the use of multigene panels) to guide drug therapy.
thrombosis, which usually results in sudden death or heart Initiatives like the European Pharmacogenetics Implemen-
attack, and a few patients have strong responses with bleed- tation Consortium (EU-PIC; http://www.eu-pic.net) and the
ing). Once absorbed, 85% of clopidogrel is inactivated via Implementing Genomics in Practice (IGNITE, https://www.
esterases, and only about 15% of the prodrug remains avail- genome.gov/Funded-Programs-Projects/Implementing-
able for a multistep activation via hepatic CYP enzymes. The Genomics-in-Practice-IGNITE) Network are developing
active drug selectively and irreversibly binds to the ADP- and testing pharmacogenomics implementation strategies.
dependent P2Y12 receptor on thrombocytes and thereby Moreover, the CPIC (https://cpicpgx.org) provides evidence-
inhibits platelet activation and aggregation for the platelets’ based, peer-reviewed, and publicly available guidelines that
lifespan, which is about 10 days. Candidate gene investiga- aim to support physicians to bring personalized medicine
tions identified loss-of-function variants in the activating into clinical practice, by using an individual’s genetic infor-
enzyme CYP2C19 to significantly affect drug response. As of mation to prescribe medications. Electronic clinical decision
May 2019, a total of 35 variant CYP2C19 alleles (https://www. support systems (CDSSs) can make it feasible to utilize
pharmvar.org/htdocs/archive/cyp2c19.htm) have been iden- genetic information to prescribe drugs, and CDSSs are con-
tified; the most important poor metabolizer (PM) alleles are tinuously improving and becoming more widely available.
*2 (~15% in Europeans and Africans, ~30% in Asians) and Collectively, it can be expected that pharmacogenomics will
the less frequent *3 (2–9% in Asians, less than 1% in Euro- be an important component of the announced Precision
peans and Africans). The *17 gain-of-function allele results Medicine Initiative (https://www.whitehouse.gov/precision-
in enhanced CYP2C19 enzyme activity, and can place these medicine).
ultra-metabolizing individuals at a higher risk for bleeding One example of the successful implementation of a CDSS
because of increased drug activation. evidence-based pharmacogenomics precision medicine
Based on the results of clinical investigations, the FDA approach in routine clinical hematology was recently
has issued a “black box” warning for clopidogrel in regard reported from the SJCRH. Before the administration of
to reduced effectiveness in PM individuals (i.e. two loss-of- codeine, which is given to treat pain crisis in SCD, more
function CYP2C19 alleles), and genotyping for the important than 600 patients with SCD were genotyped for CYP2D6
variants is widely available. The CPIC Dosing Guideline for variants (CYP2D6 activates the prodrug codeine to the
Pharmacogenomics 351

active morphine). Interruptive alerts recommend against Den Boer, M.L., van Slegtenhorst, M., De Menezes, R.X. et al. (2009). A
codeine for patients with high-risk CYP2D6 status (i.e. 7.1% subtype of childhood acute lymphoblastic leukemia with poor treat-
ultra-rapid metabolizers who are at risk for life-threatening ment outcome: a genome-wide classification study. Lancet Oncol. 10:
ADRs; 1.4% poor metabolizers who are at risk for insufficient 125–134.
pain control), and this example shows how safety concerns Diouf, B., Cheng, Q., Krynetskaia, N. et al. (2011). Somatic deletions of
genes regulating MSH2 protein stability cause DNA mismatch repair
can be reduced via preemptive genotyping. It should also
deficiency and drug resistance in human leukemia cells. Nat. Med. 17:
be noted that the frequency of CYP2D6 poor metabolizers 1298–1303.
would be higher (10%) and the frequency of ultra-rapid Diouf, B., Crews, C., Lew, G. et al. (2015). Association of an inher-
metabolizers lower (1–2%) when patients of European ited genetic variant with vincristine-related peripheral neuropathy in
ancestry are prescribed codeine, illustrating ancestry-related children with acute lymphoblastic leukemia. JAMA 313: 815–823.
differences in inherited genome variation. Fischer, U., Forster, M., Rinaldi, A. et al. (2015). Genomics and drug
profiling of fatal TCF3-HLF-positive acute lymphoblastic leukemia
identifies recurrent mutation patterns and therapeutic options. Nat.
Genet. 47: 1020–1029.
Further reading Karol, S.E., Mattano, L.A., Yang, W. et al. (2016). Genetic risk factors for
the development of osteonecrosis in children under age 10 treated for
Principles of pharmacogenomics acute lymphoblastic leukemia. Blood 127: 558–564.
Karol, S.E., Yang, W., van Driest, S.L. et al. (2015). Genetics of
Evans, W.E. and Relling, M.V. (1999). Pharmacogenomics: translating
glucocorticoid-associated osteonecrosis in children with acute lym-
functional genomics into rational therapeutics. Science 286: 487–
phoblastic leukemia. Blood 126: 1770–1776.
491.
Li, B., Li, H., Bai, Y. et al. (2015). Negative feedback-defective PRPS1
Evans, W.E. and Relling, M.V. (2004). Moving towards individualized
mutants drive thiopurine resistance in relapsed childhood ALL. Nat.
medicine with pharmacogenomics. Nature 429: 464–468.
Med. 21: 563–571.
Gammal, R.S., Crews, K.R., Haidar, C.E. et al. (2016). Pharmacoge-
Liu, C., Yang, W., Devidas, M. et al. (2016). Clinical and genetic risk
netics for safe codeine use in sickle cell disease. Pediatrics 138 (1):
factors for acute pancreatitis in patients with acute lymphoblastic
e20153479. https://doi.org/10.1542/peds.2015-3479.
leukemia. J. Clin. Oncol. 34: 2133–2140.
He, Y., Hoskins, J.M., and McLeod, H.L. (2011). Copy number
Ma, X., Edmonson, M., Yergeau, D. et al. (2015). Rise and fall of sub-
variants in pharmacogenetic genes. Trends Mol. Med. 17: 244–
clones from diagnostic to relapse in pediatric B-acute lymphoblastic
251.
leukemia. Nat. Commun. 6: 6604.
Hoffman, J.M., Haidar, C.E., Wilkinson, M.R. et al. (2014). PG4KDS:
Meyer, J.A., Wang, J., Hogan, L.E. et al. (2013). Relapse specific muta-
a model for the pre-emptive implementation of pharmacogenetics.
tions in NT5C2 in childhood acute lymphoblastic leukemia. Nat.
Am. J. Med. Genet. C Semin. Med. Genet. 166C: 45–55.
Genet. 45: 290–294.
Hunt, R.C., Simhadri, V.L., Indoli, M. et al. (2014). Exposing synony-
Moriyama, T., Nishii, R., Perez-Andreu, V. et al. (2016). NUDT15 poly-
mous mutations. Trends Genet. 30: 308–321.
morphisms alter thiopurine metabolism and hematopoietic toxicity.
Ivanov, M., Barragan, I., and Inglman-Sundberg, M. (2014). Epigenetic
Nat. Genet. 48: 367–373.
mechanisms of importance for drug treatment. Trends Pharmacol.
Mullighan, C.G., Su, X., Zhang, J. et al. (2009). Deletion of IKZF1 and
Sci. 35: 384–396.
prognosis in acute lymphoblastic leukemia. New Engl. J. Med. 360:
Luzzatto, L. and Seneca, E. (2014). G6PD deficiency: a classic example
470–480.
of pharmacogenomics with on-going clinical implications. British J.
Paugh, S.W., Bonten, E.J., Savic, D. et al. (2015). NALP3 inflamma-
Haematol. 164: 469–480.
some upregulation and CASP1 cleavage of the glucocorticoid recep-
Manalio, T.A. (2013). Bringing genome-wide association findings into
tor cause glucocorticoid resistance in leukemia cells. Nat. Genet. 47:
clinical use. Nat. Rev. Genet. 14: 549–558.
607–614.
Relling, M.V. and Evans, W.E. (2015). Pharmacogenetic in the clinics.
Ramsey, L.B., Panetta, J.C., Smith, C. et al. (2013). Genome-wide
Nature 526: 343–350.
study of methotrexate clearance replicates SLCO1B1. Blood 121: 898–
Rukov, J.L., Wilentzik, R., Jaffe, I. et al. (2014). Pharmaco-miR: linking
904.
microRNAs and drug effects. Brief. Bioinform. 15: 648–659.
Relling, M.V., Gardner, E.E., Sandborn, W.J. et al. (2014). Clinical Phar-
macogenetics Implementation Consortium guidelines for thiopurine
Pharmacogenomics to improve childhood methyltransferase genotype and thiopurine dosing: 2013 update.
Clin. Pharmacol. Ther. 93: 324–325.
all therapy
Relling, M.V., McDonagh, E.M., Chang, T. et al. (2013). Clinical Phar-
Annesley, C.E. and Brown, P. (2014). The biology and targeting of FLT3 macogenetics Implementation Consortium guidelines for rasburic-
in pediatric leukemia. Front. Oncol. 4: 263. https://doi.org/10.3389/ ase therapy in the context of G6PD deficiency genotype. Clin. Phar-
fonc.2014.00263.eCollection. macol. Ther. 96: 169–174.
Cheok, M.H. and Evans, W.E. (2006). Acute lymphoblastic leukaemia: a Roberts, K.G., Li, Y., Payne-Turner, D. et al. (2014). Targetable kinase-
model for the pharmacogenomics of cancer therapy. Nat. Rev. Cancer activating lesions in Ph-like acute lymphoblastic leukemia. N. Engl. J.
6: 117–129. Med. 371: 1005–1015.
352 Molecular Hematology

Tzoneva, G., Perez-Garcia, A., Carpenter, Z. et al. (2013). Activating Johnson, J.A., Gong, L., Whirl-Carrillo, M. et al. (2011). Clinical Phar-
mutations in the NT5C2 nucleotidase gene drive chemotherapy resis- macogenetics Implementation Consortium guidelines for CYP2C9
tance in relapsed ALL. Nat. Med. 19: 368–371. and VKORC1 genotypes and warfarin dosing. Clin. Pharmacol. Ther.
Wang, X., Liu, W., Sun, C.L. et al. (2014). Hyaluronan synthase 3 variant 90: 625–629.
and anthracycline-related cardiomyopathy: a report from the Chil- Mega, J.I., Walker, J.R., Ruff, C.T. et al. (2015). Genetic and the clinical
dren’s Oncology Group. J. Clin. Oncol. 32: 657–653. response to warfarin and edoxaban: findings from the randomized,
Yang, J.J., Landier, W., Yang, W. et al. (2015). Inherited NUDT15 vari- double-blind ENGAGE AF-TIMI 48 trial. Lancet 385: 2280–2287.
ant is a genetic determinant of mercaptopurin intolerance in chil- Nagalla, S. and Bray, P.F. (2016). Personalized medicine in thrombosis:
dren with acute lymphoblastic leukemia. J. Clin. Oncol. 33: 1235– back to the future. Blood 127: 2665–2671.
1242. Pirmohamed, M., Kamali, F., Daly, A.K. et al. (2015). Oral anticoagula-
Yang, S.K., Hong, M., Baek, J. et al. (2014). A common missense variant tion: a critique of recent advances and controversies. Trends Pharma-
in NUDT15 confers susceptibility to thiopurine-induced leucopenia. col. Sci. 36: 153–163.
Nat. Genet. 46: 1017–1020. Scott, S.A., Sangkuhl, K., Stein, C.M. et al. (2013). Clinical Pharmaco-
genetics Implementation Consortium guidelines for CYP2C19 geno-
type and clopidogrel therapy: 2013 update. Clin. Pharmacol. Ther. 94:
Pharmacogenomics to improve oral
317–323.
antithrombotic therapies
Stergiopoulos, K. and Brown, D.L. (2014). Genotype-guided vs clinical
Ismail, S., Lee, M.Y., Duarte, J.D. et al. (2014). Genotype- and dosing in warfarin and its analogues. Meta-analysis of randomized
phenotype-directed antiplatelet therapy selection in patients with clinical trials. JAMA Intern. Med. 174: 1330–1339.
acute coronary syndromes. Expert. Rev. Cardiovasc. Ther. 12: 1289–
1303.
Chapter 25 History and development of
molecular biology
Paul Moss
School of Cancer Sciences, University of Birmingham, Birmingham, UK

Evolution is the central tenet of biology, 353 Experimental techniques and molecular biology, 358
The understanding of monogenic and polygenic inheritance, 353 Conclusion, 362
DNA as the conduit of genetic information, 354 Further reading, 362
The rough guide to the human genome, 357

the prevailing view that all species had been placed on the
Evolution is the central tenet of earth by a divine Creator.
biology Even in Darwin’s time there were fierce arguments about
the nature of biological inheritance. Samuel Butler, later to
It is 160 years since the publication of On the Origin of Species
become a celebrated novelist, emigrated to New Zealand to
by Means of Natural Selection (Figure 25.1a). Darwin’s far-
make his fortune in sheep breeding. While there, he read Dar-
reaching insights placed natural variation and adaptation as
win’s work and, through his own highly successful breeding
the prime determinants of population change. Evolution is
experiments, came to understand the concept that “informa-
now recognized as the unifying theme of all biology, includ-
tion” was flowing from one generation to the next. His ideas
ing hematology and medicine, and these landmark observa-
were taken further by William Bateson, a Cambridge geneti-
tions can perhaps be recognized as the initiation of the mod-
cist who introduced the term genetics in 1906 to describe
ern discipline of molecular genetics.
the science of inheritance and variation. The units of this
However, despite his unique vision and imagination, Dar-
information were subsequently termed genes by Wilhelm
win was never able to understand the nature of genetic inheri-
Johannsen.
tance. It was clear that phenotypic characteristics were passed
from one generation to the next, but usually these charac-
teristics were “blended,” so that a combination of tall and
short parents would produce a child of medium height. Dar- The understanding of monogenic
win proposed his own model for heredity which he termed and polygenic inheritance
pangenesis, in which cells of the body shed gemmules that
collect in the reproductive organs. The concept was that all The modern concept of genetic inheritance began with the
the tissues in the body thus had some impact on inheri- work of Gregor Mendel, an Austrian priest who had under-
tance, but this concept has clearly been superseded. At this gone training as a science teacher and is famous for his
time there was no concept of the distinction between the pea-breeding experiments published in 1866 (Figure 25.1b).
germline and somatic tissue, and it is therefore not surprising Mendel used true-breeding varieties of pea to show that the
that even brilliant scientists such as Jean-Baptiste de Lamarck crossing of two dissimilar strains led to a uniform first gener-
believed that acquired phenotypic features could be passed, ation (F1 ). However, the crossing of the F1 generation with an
through reproduction, into the subsequent generation. How- F2 generation produced a population where three-quarters of
ever, despite his somewhat imperfect conclusions, Lamarck the peas had the same (dominant) parental characteristic and
was actually an extremely important figure in evolutionary one-quarter had the other (recessive) parental form. In the
theory, as he developed the concept that a species can change course of over 30 000 observations, Mendel went on to breed
between different generations, a view entirely in contrast to subtypes of F2 and laid the rules for dominant and reces-
sive inheritance. He postulated that each characteristic was
determined by two elements from the parents, and that these
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. were both retained and appeared in the germline as single
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. forms in equal proportions. The final expression in each plant

353
354 Molecular Hematology

(a) (b)

(d)

Fig. 25.1 (a) Charles Darwin. (b) Gregor Mendel.


(c) (c) Francis Crick and James Watson. (d) Fred Sanger.

was dependent on the combination of dominant or recessive a phenotype that displayed discontinuous properties, and
elements. These experiments were hugely influential to the the concept of polygenic inheritance of characteristics was
development of molecular biology, as they showed that her- thus born.
itable characteristics are particulate and not “blended.” His
work was largely ignored for the next 35 years, until it was
rediscovered in the early twentieth century and widely sup- DNA as the conduit of genetic
ported by Bateson and colleagues. The particulate element information
became known as the gene and the modern basis of genetics
was established. From this time, the science of genetics became firmly estab-
A huge problem in these early days was how to link lished and rooted in all aspects of biological investigation.
the clear biological results of Mendelian experiments with However, the nature of the genetic material was completely
the discontinuous phenotypes seen in everyday life. For unknown until the pioneering work of Oswald Avery in
instance, how can single pieces of information determine 1944. Avery trained as a medical doctor and was 67 at
such variables such as height or intelligence? It was left to the time of his most famous scientific contribution. He
the work of R.A. Fisher in 1918 to show that the inher- had been working on the inheritance of S and R strains
itance of multiple different single genes could produce of Streptococcus pneumoniae and was able to show that,
History and development of molecular biology 355

contrary to the current view, the use of proteases could 5ʹ end 3ʹ end
not prevent transmission of genetic information between O O–
strains. In contrast, deoxyribonuclease was highly effective in NH2 O
P
N T OH
preventing transmission, and this work therefore established –O
O N HN N
DNA as the medium for genetic inheritance. Avery died in A
O N N O
1955 and is often regarded as one of the most deserving O
scientists never to have been awarded the Nobel Prize in
Medicine or Physiology. Having established DNA as the O O–
O O
physical basis of heritability, genetic research then proceeded P NH2 O N P
to dominate biological investigation for the next 50 years. –
O O
O O
C N HN G N
O N
Elucidation of the structure of DNA O H2N O

The elucidation of the structure of DNA has become per-


OH O O–
haps the most memorable biological achievement of the last 3ʹ end P
century. The story is well known, and involves the outstand- –O
ing X-ray crystallography of Maurice Wilkins and Rosalind O
Franklyn, combined with the imagination and intelligence of 5ʹ end
James Watson and Francis Crick (Figure 25.1c). Their paper, Fig. 25.2 The structure of DNA. The deoxyribose pentose sugars are
published in 1953, took advantage of a wealth of previous joined by phosphodiester bonds at C-3 or C-5 to phosphate groups.
information from other investigators, and this was instru- The four bases, adenine (A), thymine (T), cytosine (C), and guanine
mental in allowing a final elucidation. (G), are attached to this backbone and the double helix is stabilized by
In 1919, Levene had demonstrated that DNA consisted of hydrogen bonds (represented by zig-zag lines).
three components: the bases (of which there were four types),
a sugar (which was either ribose or deoxyribose), and a phos-
phate group. He showed that these were linked together in positively charged proteins termed histones that package the
a unit that he called a nucleotide. The work of Irwin Char- DNA. The core genetic complexity of the DNA molecule
gaff defined the so-called Chargaff ratio, which relates to the comes from the composition of bases at the center of the
base composition of DNA. This showed that the amount of helix. These bases are of two types, purines and pyrim-
guanine equals that of cytosine, and the amount of adenine idines, the former comprising adenine and guanine, fused
equals that of thymine. Structural analysis revealed that the heterocyclic compounds of five- and six-membered rings,
hydrogen bonds between guanine and cytosine and between the latter cytosine and thymidine, which are more simple
adenine and thymine were of similar structure, and led to a six-membered rings. The pairing of guanine with cytosine
model in which the hydrophobic bases were on the inside of provides greater strength of interaction due to optimal
the double helix, leaving the hydrophilic components on the hydrogen bonding.
outside (Figure 25.2). The mechanism by which DNA replicates was a further
The structure of DNA has now been well characterized question that needed to be resolved. Watson and Crick had
and comprises three different subunits, a sugar, a base, and a ended their paper with the famous words: “it has not escaped
phosphate group; together these form a nucleotide. DNA is our notice that the specific pairing we have postulated
a polymer of nucleotides and it is the sequence of bases that immediately suggests a possible copying mechanism for the
determines the genetic information. Certain aspects of the genetic material.” This implied that the two helices could
nucleotide component are worth looking at in more detail. separate and allow copying of each strand. In 1958, Matthew
The backbone of the strand is made from phosphate and Meselson and Franklin Stahl showed that the replication of
sugar residues, the latter being a 2-deoxyribose five-carbon DNA was semi-conservative. That is, when a copy is made
(pentose) sugar. Importantly, these sugars are joined either at of a double-stranded DNA template, a complementary
their third or fifth carbon atom, and this asymmetry gives a sequence is made against each of the two original strands,
polarity to the DNA strand, with the 5′ end having a terminal rather than production of a completely new double strand
phosphate group and the 3′ end a terminal hydroxyl residue. with retention of the original duplex.
The phosphate groups form phosphodiester bonds, which
contain a phosphorus atom that allows strong covalent
Decoding the sequence of DNA
linkage through an ester bond. These groups are negatively
charged and this plays a role in forcing phosphates away Later work by Francis Crick predicted that “adapter” inter-
from the center of the DNA helix, as well as combining with mediates would be found which decoded the DNA sequence
356 Molecular Hematology

into the synthesis of proteins. Transfer RNA was subsequently protein translation. There are four different rRNA molecules
isolated and found to comprise a triplet anticodon attached within the ribosome, 18S, 5.8S, 28S, and 5S, and together
to a specific amino acid. In this way, the DNA code can be they constitute over 80% of RNA within the cell. The com-
translated into a protein sequence. There are 20 different sub- bination of transcription and translation can be surpris-
types of amino acid and these are encoded by a range of dif- ingly slow. It has been estimated that it can take 21 hours
ferent triplet sequences on transfer RNA. This is the genetic to produce a molecule of dystrophin, the largest gene in
code that was elucidated in the early 1960s and was set out by the genome and the one responsible for Duchenne muscular
Crick in the form in which it is now used. Translation usually dystrophy.
commences with an AUG triplet, which codes for methion- Recently, several new classes of RNA species have been
ine. Protein synthesis occurs within the cellular cytoplasm on discovered and are likely to play very important roles in the
ribosomes, which are complex mixtures of RNA and protein genetic regulation of eukaryotic cells. A particularly impor-
components. tant discovery has been the finding that RNA can have cat-
This flow of genetic information from DNA to RNA to pro- alytic activity, demonstrating that it is more than simply
tein became known as the central dogma of molecular biol- a passive scaffold. MicroRNA species are approximately 21
ogy. Although never meant to be as truly closed to critical nucleotides in length and do not code for proteins but, on
evaluation as the term implies, this observation was highly the contrary, carry complementary sequences that are able
influential in understanding the centrality of germline DNA to bind to mRNA species and regulate the rate of protein
in evolutionary processes, and essentially contradicted the production. This is done either by binding to mRNA species
concept that somatic information could flow back into the and preventing translation on ribosomes, or via recruitment
germline, as envisaged in the original Lamarckian theory of of ribonucleases that degrade double-stranded RNA species
evolution. and thus reduce the half-life of the targeted RNA species.
RNA is a major component of the cellular nucleus and Current estimates suggest that there are over 1000 micro-
three main forms were distinguished in the early days of RNA transcripts within the human genome, and it is cur-
molecular biology. Messenger RNA (mRNA) copies the DNA rently believed that mutations or deregulation of microRNA
genetic code into transcripts that travel to the cytoplasm expression can be implicated in a range of malignant dis-
for protein translation. This process of transcription allows eases; involvement of deletions of microRNAs in the etiology
different cells, all of which carry the same gene content, to of chronic lymphocytic leukemia secondary to chromosome
utilize their genome in differential ways, and transcriptional 13q deletions is one such example. The net overall effect of
regulation lies at the heart of cell differentiation programs. these systems is that the cell thus produces not only mRNA
The initial concept that one gene gives rise to a single mRNA for protein translation, but also species of RNA with com-
species, and subsequently a single protein molecule, has plementary sequence to mRNA transcripts that serve to limit
been challenged in several ways over the last few decades. information derived from each mRNA transcript. This feed-
The first observation was that genes are split into discrete back control is, of course, a commonly identified mechanism
units of expressed code, termed exons, and these DNA in many biological systems.
segments are separated by intervening sequences of intronic A range of other RNA species are now being identi-
DNA. Most of the genes within the human genome have this fied. These include small nuclear RNAs (snRNAs), which
intron–exon structure and, although the selective advantage are involved in processes such as mRNA splicing, and small
of this development is unknown, it does allow increased nucleolar RNAs. Many long non-coding RNA species of over
flexibility for decoding genetic information into a range of 200 nucleotides in length are also produced whose functions
protein transcripts. In order for an initial mRNA species to are largely unknown. They are often very highly conserved
encode an accurate representation of the exon structures, in evolution and found in gene-poor regions of the genome.
the transcript must be spliced by splicing enzymes within the It is likely that they play important and indispensable roles in
nucleus by a process that removes intervening intronic RNA embryonic development.
segments. Many genetic diseases have now been revealed
as due to aberrations in splicing mechanisms, and the tha-
Regulation of transcriptional activity
lassemia syndromes, which arguably have told us more about
the principles of molecular regulation than any other genetic The DNA genome encodes the information from which our
disease, are commonly inactivated by mutations in this complex multicellular structure can be assembled. However,
mechanism. the code is deciphered in a highly complex process involving
Ribosomal RNA (rRNA) is complexed with ribosomal pro- the interaction of many different proteins and nucleic acids.
teins to produce the large number of ribosomes that are scat- DNA is copied into RNA by the RNA polymerase II
tered around the cytoplasm and act as the principal sites for enzyme, which recognizes a promoter sequence at the 5′ end
History and development of molecular biology 357

of the gene. The organization of promoter sequences is highly This is believed to reflect the fact that the current human
disparate, although many contain the TATAAA sequence population of some 6 billion people has grown rapidly over
known as the TATA box. Promoter sequences attract a large 3000 generations from a founder size “bottleneck” of only
number of proteins called transcription factors, which serve 10 000 individuals.
to activate or repress transcription at that gene. These pro- Sequencing has also revealed that copy number variation is
teins all contain a DNA-binding domain and attract an addi- a common finding in the human genome. We seem to have
tional set of proteins involved in chromatin remodeling that a dynamic genome in which relatively large areas of deletion
also regulate transcriptional activity (see Epigenetic Regula- and insertion are seen, and this may underlie a range of disor-
tion Adds a New Dimension of Complexity). ders that may involve cryptic gene inactivation and translo-
A further set of DNA regulatory elements are enhancer cations. It may be that this high level of genomic instability
sequences that regulate the transcriptional activity of genes has contributed to our rapid evolution, but clearly there also
which are often many kilobases away. Their precise mecha- appears to be a price to pay.
nism of action is uncertain, but is likely to involve processes
such as “looping out” of DNA and colocalization of transcrip-
The packaging of genetic material
tion factors.
One fact that almost all of us remember is that each cell in the
human body contains approximately 1 m of double-stranded
The rough guide to the human genome DNA, if it were stretched out from end to end. Given that we
have some 1014 cells within our body, the whole content of
The human genome contains a sequence of 3000 million our somatic DNA would stretch for some 1014 m. It is no sur-
bases of DNA from each parent, equating to some 6 billion prise therefore that this length of DNA must be packed very
bases in a diploid cell. The Human Genome Project started accurately and densely into a typical cell of around 8 μm. The
in 1990 and 99.3% of the sequence was completed by 2003. first order of packaging is the nucleosome, in which two loops
Indeed, the genome has now been fully sequenced for sev- of DNA are wrapped round a histone protein. As we shall see
eral individuals, and this has risen substantially over the last later, this structure is critical not only for efficient mechan-
decade. Large-scale sequencing has revealed a number of sur- ical packaging, but also for controlling gene regulation. The
prises about the human genome. The first of these is that second-order structure is coiling of nucleosomes into a rope-
the gene content is much lower than was initially estimated. like structure, but further order packaging beyond this is cur-
Current estimates suggest a gene content of around 25 000, rently unknown.
whereas early proposals placed this nearer 100 000. In addi-
tion, only 2% of the genome actually codes for protein. The
Epigenetic regulation adds a new
additional 98% has a complex regulatory function and its
dimension of complexity
importance is still largely unknown. During the early days
of molecular biology, these regions were rather arrogantly Additional complexities to understanding the transcriptional
termed “junk DNA,” but they have since turned out to be regulation of the DNA code are being identified on a regular
extremely important in factors such as determining specia- basis. It is now clear that protein-induced modifications of
tion and regulation of gene expression. histone structure play an important role in determining the
Different individuals show considerable variation within transcriptional regulation of individual genes. The two major
the genome. This natural polymorphism is typically seen processes involved are methylation of cytosine bases at CpG
as a base change of around 1 in every 1300 bases, equating dinucleotides and acetylation of histone tails. A wide range
to over 4 million differences between individuals. These of methylases, demethylases, acetylases, and deacetylases are
variants are known as single-nucleotide polymorphisms involved in dynamic modification of histone structures, and
(SNPs) and, as well as their intrinsic value in understanding this process is now being targeted in clinical practice. Around
issues such as natural selection and evolutionary change, 75% of all CpG dinucleotides are methylated and unmethy-
they are also very valuable in determining the genetic linkage lated forms are often grouped at the 5′ end of genes, in areas
of genes in a variety of human diseases. Indeed, SNP-based known as CpG islands.
disease association studies are currently the most popular In general, methylation serves to downregulate transcrip-
approach for determining the polygenic basis of disease, and tion from a gene and demethylating agents are used in the
around 50% of these variants are in non-coding regions of management of diseases such as myelodysplasia or acute
the genome. Interestingly, most of these variants are quite myeloid leukemia. In contrast, acetylation encourages tran-
common in the human population and there is less variation scriptional activity and deacetylation inhibitors are therefore
between humans than there is between other great apes. also used in a range of hematopoietic disorders. Given the
358 Molecular Hematology

widespread modifications of methylation and acetylation in disposable cartridge methodology for extraction of nucleic
the human genome, it is perhaps surprising that the non- acids. These are rapid, often cost-effective, and generally
specific activity of these pharmacological agents can prove of much easier to use.
value without engendering significant side effects. However,
current practice suggests that they may be relatively well
Restriction enzymes and expression cloning
tolerated. Epigenetic regulation also represents one example
in which somatic tissues may influence transcription of Having isolated DNA in an aqueous solution, there is not a
the germline. Gender, in particular, can have an important great deal that you can usefully do with it in its native state.
impact on the transcriptional activity of individual genes. If it is run on an electrophoresis gel, it will be of such high
This process of imprinting can be broadly summarized as molecular weight that it will not migrate into the gel to any
revealing that males tend to encourage fetal growth, whereas extent and is therefore not suitable for downstream analy-
the maternal influence is to restrict fetal developmental. sis. A major advance in molecular biology was the devel-
Aberrations of epigenetic imprinting result in disorders such opment and utilization of bacterial restriction endonucleases.
as Prader–Willi or Angelman syndrome. These are a large family of enzymes that are present in bac-
teria and have evolved to digest double-stranded DNA. They
are important in protecting bacteria from invading bacterio-
Experimental techniques and phages and thus they “restrict” the vulnerability of the cell
molecular biology to infection. The critical finding about restriction endonu-
cleases was that they cut DNA at specific DNA sequence
Here I review some of the milestones that have led to the motifs. For instance, the enzyme EcoRI will only cut DNA
development of the experimental discipline known as molec- at positions that have the sequence 5′ -GAATTC. Note that
ular biology. This is a field which is moving perhaps more this sequence is a palindrome when double stranded and
rapidly than any other within biology and, while it offers it is in this form that the DNA is cleaved, after the G on
unparalleled opportunities for clinical practice, it is challeng- both strands. If sufficient enzyme is used in a digestion
ing for clinicians to maintain their knowledge base within reaction, every individual site within the target DNA will
this area. be cut.
These enzymes opened up the field of modern molecular
biology, because they led to the development of DNA cloning.
The extraction of nucleic acids
In this procedure, the DNA of interest is digested with a par-
A good place to begin is the extraction of nucleic acids from ticular endonuclease and then a vector DNA sequence, usu-
cells. Nucleic acids are soluble in water, but can be precipi- ally in the form of a bacterial plasmid or a virus, is also
tated by the addition of ethanol. In one method the cell or digested with the same enzyme. Providing that the vector has
tissue of interest is lysed to destroy cell membranes and the only two of these restriction sites, a portion of the vector will
protein component degraded by proteinase K or extraction be excised and then, by combining the digests of the target
with phenol and chloroform. The latter approach generates DNA and vector, the two populations can be ligated such that
an aqueous phase containing RNA and an organic phase that single copies of DNA are cloned into the vector. Typically
retains protein and lipid, with DNA being trapped at the these vectors have strong promoters that drive expression of
interface. the cloned gene, as well as genes coding for antibiotics, which
The next step is to purify and concentrate the nucleic acid, allow selection for vectors containing an insert. In this way a
which can be done by ethanol precipitation. Because DNA library of the original DNA is made.
is charged, it is highly soluble in water, but the addition of This library can then be introduced into cells for propaga-
non-polar ethanol to at least 66% of the final volume draws tion and functional analysis. Vectors can be transformed into
it out of solution. The precipitate of nucleic acids can then a bacterial host that can be multiplied using culture medium
be centrifuged, the ethanol drawn off, and the nucleic acid to a potentially unlimited population size. Lysis of the bacte-
resuspended in water. Genomic DNA is quite stable in this rial colony, followed by recovery of the plasmid or phage, can
form, perhaps not surprising given its natural physiologi- lead to isolation and analysis of large amounts of individual
cal role. However, RNA is highly unstable and susceptible target DNA sequences. This is a core technique in molecular
to RNases, which are ubiquitous enzymes within the envi- biology and has led to most of the subsequent development
ronment. Inhibitors of RNases and rapid cooling of RNA within the field. Libraries can also be introduced into eukary-
solutions are needed for downstream analysis of ribonucleic otic cells through the process of transfection, which typically
acids. Although these chemical steps are effective, and have involves incubation with cold cations such as calcium or the
been in use for many decades, many laboratories now use use of liposomes.
History and development of molecular biology 359

Electrophoresis and blotting Southern blot technology has been adopted for use in other
of nucleic acids ways and a memorable nomenclature has built up, albeit
based on a mischievous interpretation of the derivation of
Once DNA has been digested with endonucleases, it is bro-
“Southern.” The Northern blot refers to a technique in which
ken up into smaller pieces, the exact size and pattern of which
RNA is run on an agarose gel and blotted in the same way
will depend on the endonuclease that has been used. Edwin
as the DNA in a Southern blot. This approach was widely
Southern published a technique whereby this DNA was ini-
used to examine differential expression of DNA between tis-
tially subjected to electrophoresis on an agarose gel, thus sep-
sues, but has largely been replaced by microarray analysis.
arating individual pieces on the basis of their size and charge.
Western blotting refers to the technique of separating protein
If a nitrocellulose filter is then placed on top of the agarose
molecules by electrophoresis prior to blotting onto a mem-
gel and moderate pressure applied, the DNA will migrate
brane. This membrane is then probed with an antibody for
into the nitrocellulose, where it can be fixed using heat or a
detection of the presence, size, and amount of individual pro-
cross-linking agent. If the DNA filter is then hybridized to a
tein species.
labeled nucleic acid probe, the probe will bind only to that
section of the DNA containing a complementary sequence.
When genomic DNA from different individuals is digested
The polymerase chain reaction
with the same nuclease, all the resulting DNA fragments will
be of comparable size, notwithstanding the natural variation The development of the polymerase chain reaction (PCR) in
found within the human genome. Thus, the hybridization the 1980s led to the award of a Nobel Prize to Kary Mullis in
band should be similar in different individuals. This tech- 1993, and its value in medical practice is perhaps reflected by
nique became known as the Southern blot and was widely the relative swiftness of the Nobel Committee in rewarding
applied to the detection of chromosomal abnormalities such the inventor. The idea is, like most powerful innovations,
as deletions, linkage studies, and gene rearrangement iden- fairly straightforward and involves repetitive cycles of DNA
tification. It is somewhat less widely used today due to the synthesis and dissociation (Figure 25.3). DNA polymerases
advent of more modern technologies such as SNP mapping are used to copy a specific DNA segment, and this step is
and DNA sequencing. followed by an increase in reaction temperature to dissociate

3ʹ 5ʹ
DNA template
5ʹ 3ʹ

Primer
Newly synthesized
Annealing of 5ʹ
DNA strand
onucleotide Newly synthesized
primers 5ʹ DNA strands

DNA polymerase
copies
from the primer

Double strands are


separated by high
temperature

Temperature is
reduced to allow 5ʹ
annealing of primers

Fig. 25.3 The polymerase chain reaction. Typical


temperatures for annealing, extension, and DNA polymerase

extends the primers
denaturation are 50–60, 72, and 96 °C, respectively.
360 Molecular Hematology

DNA double strands and provide further templates for the biology and has played a large role in driving the discipline
amplification process. forward over the last 20 years.
Mullis performed his initial work using a hot water bath
and had to combine DNA polymerase with free nucleotides
for the initial synthesis, heat the reaction to “melt” the dou- Microarray analysis
ble strands, and allow the mixture to cool for reannealing Although sequence information of germline DNA provides
of the original primer mix, before then adding further poly- highly valuable information, in many situations it is more
merase enzyme, as the original aliquot had been killed by the valuable to know how this information is utilized within indi-
high temperatures used in the melting process. Since then, vidual cells or tissues. One approach to this is to use systems
the technique has been dramatically simplified through the which determine the quantitative profile of mRNA expression
use of thermostable DNA polymerases isolated from bacteria within tissues. Microarrays achieve this by a technique which
that live at high temperatures. Solid-phase technology, which relies on the synthesis of a huge number of oligonucleotides
allows very rapid shifts in temperature, has led to a dramatic that are complementary to each of the approximately 25 000
reduction in incubation and extension times. The technique human genes. These oligonucleotides are placed individually
utilizes short oligonucleotide primers that anneal to the tar- on microarray slides and are then hybridized to sample
get gene of interest. The DNA polymerase extends the DNA tissue. mRNA from the sample is reverse transcribed to
sequence beyond the primary sequence and, as the primers cDNA with the use of fluorescent primers, and then this
are directed toward one another on complementary strands material is hybridized to the oligonucleotides; after washing,
of DNA, the result is a complete double-stranded copy of
the target gene. Typical PCR reactions might allow ampli-
fication of between 500 and 2000 bp of DNA, but innova-
tions do exist that can extend these values. Once a gene has
been amplified in this way it can be sequenced, subjected DNA sample
to enzymatic digestion, or used in a range of downstream
applications.
Fragmentation of
DNA
DNA sequencing
Although the physical structure of DNA was defined in the P B P B Attachment of linkers
1960s, it would be many years before it became possible with biotin (B) group
to sequence the bases in order to reveal the genetic code.
Interestingly, protein sequencing had been reported many Single DNA fragment is
B
attached to a streptavidin-
years earlier, but the technology of DNA sequencing has now S
coated bead
advanced so much that its capability far exceeds current abil-
ities to sequence amino acids.
The two original methodologies were developed by Fred DNA is amplified on
Sanger and Walter Gilbert, who shared the Nobel Prize the bead
for Chemistry in 1980 (Figure 25.1d). The techniques
involved the use of DNA polymerases to sequence DNA
from an oligonucleotide primer. However, within the mix
of nucleotides needed for the building blocks of the new
Beads are dispersed
chains, Sanger’s approach incorporated a small quantity into individual wells
of nucleotides that have dideoxy modifications of their
sequence such that they cannot be extended. Four separate
reactions were used for each DNA sequence, incorporating
The four nucleotides
dideoxy reagents for the A, T, C, or G bases, respectively. 1 T are sequentially passed
If radioactivity or fluorescence is used to label the newly A
over the beads. A
2 primer complementary to
extended chains, these can then be run on a high-resolution
3 C the linker is incorporated
acrylamide gel and from this a ladder of chains is derived,
which shows the stop positions that occur when each base luminescence
C When a nucleotide
detector
terminator is used. These bands can then be read off and can be added it
a DNA sequence can be deduced. This technology was of B CTAG P emits a light signal
tremendous value in initial efforts to understand molecular Fig. 25.4 The principles of 454 next-generation sequencing.
History and development of molecular biology 361

Darwin proposes 1859 Mendel defines the


theory of natural 1866 patterns of dominant
selection as mechanism and of recessive inheritence
for evolution

William Bateson first


uses the term “genetics” 1906
Fisher develops the
Levene elucidates the 1918 concept of polygenic
1919 basis for phenotype
three components if DNA and
terms the units “nucleotide”

Oswald Avery demonstrates


that DNA carries genetic 1944
information Watson, Crick, Wilkins, and
1953 Franklyn describe the structure
Meselson and Stahl show of the DNA double helix
that DNA replication is 1958
semi-conservative The genetic code is
1960–1966
elucidated
Ed Southern describes the
Arbor, Smith and 1975
1978 “Southern Blot”
Nathans win the Nobel Prize
for discovery of restriction enzymes The development of the
1980 1983 polymerase chain reaction
Sanger and Gilbert won Nobel
Prize for developing DNA sequencing 1990 1990s Microarray technology
The Human Genome Project 2003 “Next-generation” sequencing
2005+
reads the DNA code
Fig. 25.5 Some of the key developments in molecular biology.

the amount of hybridized material is assayed using laser those developed for the Hubble telescope and have very high
detection. This approach is a highly accurate and repro- sensitivity (Figure 25.4).
ducible means of measuring the distribution of mRNA These technologies have led to a step change in the abil-
species within cells or tissues. Of course, there is no simple ity to rapidly sequence DNA and they produce almost fright-
correlation between mRNA levels and protein expression, ening levels of information. A single machine run can gen-
and proteomic analysis allows additional understanding erate 500 million base pairs of sequence, and most of the
of cellular differentiation control. Microarray systems are challenges relate to the bioinformatic analysis of the infor-
used widely in diagnostic and investigational approaches to mation or simple storage of data. The 454 system sequenced
hematopoietic malignancy. the entire genome of James Watson in two months, and in
time it is likely that many of our patients, and indeed per-
haps ourselves, will undergo full genome sequencing. Our
Next-generation sequencing
current capabilities are probably only limited by our imagi-
Over the last couple of years, new technologies have been nation and they allow experiments such as comparative anal-
developed that have dramatically altered the capability ysis of the complete sequence of germline DNA in compari-
of DNA sequencing. Two broad platforms are currently son with tumor cells, or study of the evolution of infectious
available and are known as 454 pyrosequencing and the organisms in a human host almost in real time.
Illumina/Solexa systems. Both technologies commence with Ultra-deep sequencing refers to the ability to sequence
fragmentation of DNA and then small DNA fragments are many different copies of an individual sequence in one run,
captured individually onto beads prior to an amplification also known as in parallel. As such, it can be very powerful for
step. In the 454 system, these beads are then centrifuged detecting rare mutations in tissue samples or for following
into small wells such that each well contains only one bead, genetic variation within populations of viruses.
and these are sequenced individually. The 454 system is Transcriptome sequencing is the sequence analysis of
based on luminescence, each base generating a different the mRNA population in a cell or tissue. There is the
color; the cameras used for color detection are based on possibility that this approach could replace some of the
362 Molecular Hematology

uses of microarray analysis, as it simultaneously provides


information on the number of mRNA species in the sample
Further reading
as well as determining their complete nucleotide sequence.
Ansorge, W.J. (2009). Next-generation DNA sequencing techniques.
Nat. Biotechnol. 25: 195–203.
Crick F, Watson J. (1962). On the genetic code. http://nobelprize.org/
Conclusion nobel_prizes/medicine/laureates/1962/crick-lecture.html (accessed
May 2019).
The dramatic developments in molecular biology over the Reichard, P. (2002). Osvald T. Avery and the Nobel Prize in Medicine. J.
last 50 years represent one of the most exciting stories of sci- Biol. Chem. 277: 13355–13362.
entific development (Figure 25.5). Refreshingly, they have Sanger F. (1980). Determination of the nucleotide sequences in
also been applied quickly and effectively to human health, DNA. http://nobelprize.org/nobel_prizes/chemistry/laureates/1980/
and are now integral to the clinical practice of hematology. A sanger-lecture.html (accessed May 2019).
more detailed analysis of the molecular basis of a wide range Siomi, H. and Siomi, M.C. (2009). On the road to reading the RNA-
interference code. Nature 457: 396–404.
of conditions is discussed more fully in the other chapters of
this book.
Chapter 26 Cancer stem cells
Sara Ali & Dominique Bonnet
Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, UK

The cancer stem cell concept, 363 CSC-targeted therapies, 365


The cell of origin, 364 Conclusion, 369
Pre-leukemic stem cells, 364 Further reading, 370

a small proportion of leukemic cells (0.2–100/106 leukemic


The cancer stem cell concept
blasts) and to reside in the same compartment as normal
HSCs (lineage negative, CD34+CD38− subset), leading
It was more than a century ago when the term stem cell
to the conclusion that AML likely arises from neoplastic
(Stammzelle) was coined by Haeckel to refer to the unicellular
transformation occurring in primitive stem cells rather than
ancestor of multicellular organisms that stands at the bottom
their committed progeny. These findings sparked interest
of a genealogical tree. Pappenheim later adopted the term
in exploring CSCs in various malignancies, and CSCs have
and applied it in its current meaning to describe the precur-
since been reported in solid tumors as well, providing further
sor cell, which he postulated could give rise to all blood cells.
credence to the CSC concept and extending its applicability
However, it was decades later that experimental proof for the
beyond hematological malignancies.
existence of hematopoietic stem cells (HSCs) was provided.
Although the CSC paradigm appears to hold true for dif-
While studying the effects of radiation on bone marrow func-
ferent cancers, it certainly is not universal. One major caveat
tion, Till and McCulloch made the seminal observation of
that became apparent with the emergence of more permis-
hematopoietic colonies resembling nodules in the spleens of
sive xenotransplantation models is the underestimated fre-
lethally irradiated mice that were transplanted with limiting
quency of tumor-initiating cells (TICs) reported in earlier
numbers of murine bone marrow cells. Even more striking at
studies which employed mouse strains that still had signif-
the time was the observation that upon retransplantation of
icant residual immune function. A typical example illustrat-
these colonies into secondary recipients, some were able to
ing this limitation is melanoma, which was thought, based
generate new multilineage colonies, indicating the ability of
on earlier findings, to follow the CSC model. However, this
these cells to self-renew.
was called into question when it was subsequently reported
Akin to HSCs, the cancer stem cell (CSC) hypothesis
that the frequency of cells with tumorigenic potential in
posits that tumors assume a hierarchical organization in
melanoma could be as high as 25%, arguing against there
which a subpopulation of cells with stem-like properties
being a hierarchical organization. One possible explanation
reside at the apex, and have the capacity to self-renew and
for this observation is provided by the stochastic model,
regenerate tumors that recapitulate the parental tumor from
which proposes that all cells are biologically equivalent with
which they originate. The hierarchical model in cancer was
equal clonogenic potential to facilitate tumor initiation and
first exemplified by the pioneering work of Professor J.E.
growth, provided they receive the appropriate cues, which
Dick’s team, which led to the identification of leukemic stem
may either be intrinsic or extrinsic. It must be noted, how-
cells (LSCs) in acute myeloid leukemia (AML). Leukemia-
ever, that high frequency of TIC does not, in itself, indicate a
initiating cells (LICs), functionally defined by their ability
stochastic pattern of tumor development. What distinguishes
to repopulate non-obese diabetic/severe combined immun-
the CSC paradigm from the stochastic model is that accord-
odeficient (NOD/SCID) mice, were found to represent only
ing to the former framework, only a unique subset of cells
is endowed with long-term tumorigenic potential, and they
can, based on their intrinsic properties, be separated from
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. non-TIC cells. However, this is not possible in cancers that
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. follow a stochastic model, as all cells are equally permissive to

363
364 Molecular Hematology

transformation, and therefore TICs can be potentially found Collectively, these data have led to a revised model which
in any cell fraction. unifies the CSC and stochastic models of tumor heterogene-
Nevertheless, the two models are by no means mutually ity. According to this hypothesis, CSCs in the early stages
exclusive, and can operate simultaneously within a tumor. of tumor development are likely to follow the CSC model,
Furthermore, several lines of evidence, based on findings in where CSCs account for a small proportion of the tumor
normal and malignant tissues, point toward stemness being population, with the bulk comprising non-TIC. However,
a dynamic trait whereby cells can transition between a stem as the disease progresses, these CSCs acquire advantageous
cell and a differentiated cell state and vice versa. This notion, mutations which enhance their self-renewal ability, leading to
known as bidirectional interconversion, provides an alterna- further expansion of the CSC pool, while impairing their dif-
tive explanation that can reconcile these two models. ferentiation capacity. Consequently, as the disease advances,
the hierarchy becomes flatter and the tumor becomes more
homogenous, which would explain the high frequency of
The cell of origin CSCs reported in malignancies such as melanoma.

Although often used interchangeably, the CSC is not nec-


essarily the same as the cell of origin. The cell of origin Pre-leukemic stem cells
refers to the cell that sustains the first hit required for tumor
initiation, but does not necessarily have stem cell properties The earliest evidence supporting the concept of a pre-
to allow it to sustain malignant growth. Equally important to leukemic state came from clonality studies utilizing
note is that CSCs need not be derived from their normal stem X-chromosome inactivation patterns in AML patients het-
cell counterparts, but are rather defined as cells with stem erozygous for glucose-6-phosphate dehydrogenase (G6PD)
cell properties and tumor-propagating capacity, regardless deficiency. In each of these patients, the leukemic blasts
of whether they originated from a bona fide stem cell or not. expressed the same G6PD type, indicating that they arose
Indeed, there is significant evidence indicating that CSCs from a single cell, and in approximately 25% of the patients,
can arise from downstream progenitor cells as they undergo the abnormal clone at diagnosis persisted even after attain-
oncogenic transformation. In a study by Taussig et al., sorted ment of complete morphological remission. Similar findings
leukemic cells from patients with AML with the progenitor were reported in AML patients harboring the translocation
phenotype CD34+ CD38+ were able to induce leukemia in t(8;21), who were found to have detectable levels of the
various immunodeficient mouse strains, indicating that LSC fusion transcript in peripheral blood, despite being in long-
activity is not restricted to the CD34+ CD38− compartment term remission. In recent years, corroborative evidence from
as previously hypothesized. Furthermore, lentivirally trans- sequencing data has been provided by a number of groups.
duced myeloid progenitors expressing the mixed-lineage Work from the Majeti lab demonstrated that in newly diag-
leukemia (MLL) fusion genes MLL-AF9 and MLL-ENL have nosed patients with AML, mutations in epigenetic regulators
been shown to be able to initiate leukemia. The results of such as DNMT3a and ASXL1, were frequently found in
global gene expression profiling of LSC populations in AML the purified HSC population, whereas mutations in genes
have also challenged the prevailing notion that LSCs in AML involved in activated signaling and proliferation such as FLT3
largely arise from stem cells, and suggest instead that LSCs and KRAS/NRAS were not detectable, indicating that the
in AML resemble lymphoid-myeloid multipotent progen- latter mutations were late events. Similarly, Shlush et al. per-
itors (LMPPs) and granulocyte-macrophage progenitors formed targeted sequencing of normal hematopoietic stem,
(GMP) more closely than HSCs. progenitor, and mature cell fractions in AML patients at diag-
Although CSCs can originate from a differentiated pro- nosis, and found that, in contrast to genetic aberrations such
genitor, the cell of origin may still be a stem cell. For example, as NPM1 mutation which was only detected in the leukemic
while it is widely accepted that chronic myeloid leukemia blasts, mutations in DNMT3a were found in both the
(CML) results from neoplastic transformation of an HSC by leukemic and non-leukemic compartments, indicating the
the BCR-ABL fusion gene, progression to blast crisis has been presence of a pre-leukemic clone ancestral to the dominant
shown to be driven by progenitor cells that have acquired AML clone.
stem-like properties as a result of a catalogue of events, Indeed, genomic studies of large cohorts of individuals
including BCR-ABL amplification and B-catenin activation. with no history of hematological malignancy and normal
Similarly, in AML bearing the translocation t(8;21), resulting blood counts have revealed that somatically acquired muta-
in the AML1/ETO fusion gene, although the translocation tions in hematopoietic cells are a frequent event in the aged
is found in HSC it is not sufficient to induce leukemia, population, occurring in at least 10% of individuals above
and the disease is instead driven by cells with a progenitor the age of 70. Intriguingly, most of the mutations impli-
phenotype. cated in this phenomenon, termed clonal hematopoiesis of
Cancer stem cells 365

CHIP LSC Leukemia at diagnosis

Secondary Clonal
mutations expansion
(e.g. DNMT3a (e.g. FLT3-ITD,
TET2, ASXLI) NPMI)

Leukemia at
diagnosis Remission Dominant clone

Treatment Relapse
Minor clone

LSC
Normal HSC
Pre-leukemic HSC
LSC
Dominant clone Preleukemic HSC
Minor clone

Fig. 26.1 Model of clonal evolution from clonal hematopoiesis to disease development, and the different patterns of relapse. CHIP, clonal
hematopoiesis of indeterminate potential; HSC, hematopoietic stem cells; LSC, leukemic stem cells.

indeterminate potential (CHIP), occur in epigenetic mod- diagnostic clone or, alternatively, the NPM1 mutation could
ifying genes that are frequently mutated in myeloid malig- have been acquired independently within a pre-leukemic
nancies such as DNMT3a, TET2, and ASXL1. In addition to clone. Although there has so far been no solid proof of
an increased propensity for developing leukemia, individuals relapse emerging from a pre-LSC, this route to relapse
with CHIP have an increased overall mortality. remains plausible. From a clinical perspective, the persis-
As demonstrated in several studies, pre-leukemic clones tence of pre-LSCs in remission raises the question of whether
can evade chemotherapeutic interventions and persist or testing for minimal residual disease should include moni-
even expand in remission, and it has been shown that this toring for pre-leukemic mutations and not only the “driver”
is more likely to occur among older patients, providing an mutations present in leukemic blasts and LSCs, and whether
additional explanation for the poor outcome observed in this progressive increase in pre-leukemic burden during remis-
group of patients. Additionally, persistence of pre-leukemic sion would warrant early treatment prior to the emergence
mutations in remission in those who did not receive of frank leukemia (see Figure 26.1).
allogeneic transplant was found to be associated with an
increased risk for relapse. In a study by Ding and colleagues,
whole-genome sequencing of paired samples from diagnosis CSC-targeted therapies
and relapse in patients with AML revealed two main patterns
of clonal evolution: in some patients the relapse clone
descended from the dominant clone, which acquired addi-
Targeting cell surface antigens
tional mutations, whereas in others the dominant clone was Since HSCs and LSCs share the cell surface phenotype
eradicated by treatment, but relapse arose from a subclone CD34+ /CD38− , several groups have attempted to identify
that was present at diagnosis. Furthermore, in the study by markers that can accurately distinguish LSCs from HSCs, in
Shlush et al., analysis of early and late remission samples from order to enable preferential targeting of LSCs while sparing
a patient who was still in remission at three years revealed normal HSCs. Among the earliest identified LSC antigenic
a progressive increase of DNMT3a allele frequency in most candidates is interleukin-3 alpha receptor (CD123), which is
of the progenitor and mature cells fractions over time. A strongly expressed in CD34+38− cells of patients with AML,
small proportion of the myeloid cells in the late remission but virtually absent in the normal HSC counterpart. The LSC
sample was also found to harbor both DNMT3a and NPM1 functional capacity of this population has been verified in
mutations, which could be explained by reexpansion of the vivo, where they were able to engraft and induce leukemia
366 Molecular Hematology

in NOD/SCID mice. Other putative LSC markers include recognized that LSCs can be phenotypically heterogeneous
CD33, CLL-1, Tim-3, and CD47, which are currently being and therefore an LSC-targeted therapy aimed at a single anti-
pursued in clinical trials. gen is unlikely to be able to eliminate all LSC clones. The fact
Various therapeutic approaches aimed at targeting surface that LSCs undergo clonal evolution as the disease progresses,
markers on LSCs have been explored, and one strategy has and in response to selective pressure exerted by treatment,
been the use of antibody-drug conjugates (ADCs), consisting adds yet another layer of complexity to the picture.
of a monoclonal antibody linked to a therapeutic moiety,
which allows selective delivery of cytotoxic drugs to neoplas-
Targeting CSC self-renewal
tic cells bearing the targeted antigen, with minimal impact
on normal tissues. A promising example of this class of drugs Aberrant activation of the Hedgehog (Hh) signaling path-
and the first ADC to gain approval for clinical use in cancer way has been reported in CSCs of various cancers, including
is Gemtuzumab ozogamicin (GO), comprising a humanized hematological malignancies. In a mouse model of CML, con-
anti-CD33 antibody conjugated to the antitumor antibiotic stitutive activation of Smoothend, a key component of the Hh
calicheamicin. After being initially withdrawn due to lack of pathway, resulted in an increase in CML LSC frequency lead-
clinical benefit and safety concerns, GO was reapproved by ing to disease acceleration, while inhibition of Hh signaling
the US Food and Drug Administration (FDA) when subse- by cyclopamine caused reduction in CML stem cells, and this
quent clinical trials using a lower dosing schedule showed effect was maintained even in imatinib-resistant CML-LSC.
that the addition of GO to standard chemotherapy for The Wnt pathway effector β-catenin has also been impli-
treatment of patients with newly diagnosed AML resulted in cated in the regulation of LSC. In murine AML models driven
improved outcomes in terms of an event-free survival which by MLL-ENL or co-expression of the oncogenes HOXA9
was nearly twice as long (median event-free survival: 17.3 and MEIS1, β-catenin was required for transformation to
months vs. 9.5 months). Improvement in obtaining complete AML. Furthermore, functional dependence of LSC on the
remission was also observed when used in the relapse canonical Wnt signaling has been demonstrated in T-cell
setting as well as a small, but significant, increase in overall acute lymphoblastic leukemia (T-ALL). In B-ALL, suppres-
survival when used as a single agent in older patients who sion of CBP/catenin using a small molecule inhibitor abro-
are ineligible for intensive chemotherapy. It should be noted, gated self-renewal of pre-B ALL cells, and promoted their
however, that CD33 is not an exclusive marker of LSC, but is differentiation, thereby sensitizing them to chemotherapy.
also expressed in leukemic blasts and myeloid progenitors, Similarly, LSCs in MLL-driven AML that have acquired resis-
and thus far it has not been shown whether the improved tance to GSK3 inhibitors could be resensitized by inhibiting
outcome observed with GO relates specifically to its effect on β-catenin. Of note is that both Hh and β-catenin appear dis-
the LSC compartment or merely the disease bulk. pensable for adult HSC function, rendering them attractive
Other approaches employing the immunophenotypic candidates for selective inhibition of LSCs.
properties of LSCs include bispecific antibodies which link The canonical Notch signaling pathway is another pivotal
the target pathogenic cell to a cytotoxic effector cell, such as player in maintaining self-renewal of LSC. There has been
a T cell or natural killer cell, by simultaneously engaging a a particular interest in targeting Notch in T-ALL due to the
tumor-specific antigen and a receptor expressed on the cyto- high prevalence of Notch1 mutations, which are estimated to
toxic effector cell, thereby inducing cell death. Adoptive cell occur in more than 50% of patients. Treatment with a gamma
therapy using chimeric antigen receptor (CAR)-engineered secretase inhibitor, which blocks Notch, has been shown to
T cells has also been pursued as an LSC-targeting avenue. abolish the leukemic-initiating stem cell activity in T-ALL
Albeit promising, targeting LSCs by utilizing their aber- in vivo, regardless of mutational status. Other strategies to
rant immunophenotype has potential limitations, which may target Notch include the use of monoclonal antibodies and
hinder its clinical utility. One major concern relates to the stapled peptides.
specificity of LSC marker, as it is crucial that an antigen used
to identify LSC is not expressed on normal HSC in order
Targeting CSC survival
to minimize toxicity. For instance, while CD33 has been
thought not to be expressed in normal HSC, Taussig et al. One of the earliest pieces of evidence implicating the nuclear
reported CD33 expression in normal Lin− CD34+ CD38− factor-kB (NF-kB) in CSC came from AML, where NF-kB
fractions isolated from cord blood and healthy adult bone was found to be constitutively activated in the CD34+ frac-
marrow, which were able to repopulate NOD/SCID mice and tion of AML cells but not in normal CD34+ HSCs, indicating
had self-renewal ability, as determined in serial transplanta- that reliance on NF-kB is an exclusive feature of LSCs. Acti-
tion assays. Indeed, CD33 has been recently reported to iden- vation of NF-kB can occur through multiple routes, includ-
tify a subset of cells with serial repopulating capacity within ing chromosomal/molecular aberrations, cytokine signaling,
the most primitive HSC fraction. Furthermore, it is well or increased proteasomal activity. Inhibition of NF-kB by a
Cancer stem cells 367

proteasome inhibitor was shown to induce apoptosis in AML HSCs. As with HSCs, niche-derived signals are vital for main-
cells in both the bulk and CD34+ fraction. taining LSCs (see Figure 26.2). Evidence from several murine
Additionally, inhibitors against the PI3/AKT/mtor path- models even indicates that alterations in components of the
way have been employed in AML. Constitutive activation of bone marrow microenvironment may be sufficient to disrupt
phosphoinositide 3-kinase (PI3K) is necessary for the sur- normal hematopoiesis and induce neoplastic transformation.
vival of AML, and reduction in AML LSC has been reported In an elegant study by the Scadden group, selective deletion
in murine models treated with PI3K inhibitors. Intriguingly, of the miRNA processing endonuclease Dicer 1 in osteopro-
patients with constitutive PI3K have a favorable prognosis, genitors, resulting in impaired obsteoblastic differentiation,
which is proposed to be due to PI3K driving LSCs into the S induced profound hematopoietic defects mimicking
phase of the cell cycle, thus making them more sensitive to myelodysplasia, with subsequent transformation to myeloid
chemotherapy. sarcoma in some mice. Intriguingly, the same phenotype
was observed when wild-type hematopoietic cells were
transplanted into mice with mutant niche, while the effect
Targeting LSC dormancy
was reversed upon transplantation of hematopoietic cells
Chemoresistance is often attributed to CSCs, which due from mutant mice into a wild-type environment. This lends
to their largely quiescent nature can evade the effects further support to the concept of niche-driven oncogenesis,
of chemotherapy agents that preferentially target actively whereby microenvironmental aberrations constitute the
cycling cells. Breaking the dormancy of LSCs therefore repre- primary pathogenic event that drives tumorigenesis, which
sents a rational approach to eliminate the LSC population. In consequently facilitates the acquisition of oncogenic hits in
murine models, AML cells residing in the endosteal region, the hematopoietic compartment. Similarly, constitutive acti-
where quiescent, chemo-resistant LSCs preferentially home vation of β-catenin in murine osteoblasts has been shown to
to, have been shown to enter the cell cycle upon treatment alter the differentiation potential of lymphoid and myeloid
with the mobilizing agent granulocyte colony-stimulating progenitors, leading to a differentiation block and the emer-
factor (GCSF). Furthermore, addition of GCSF to cytara- gence of AML through upregulation of the Notch-ligand
bine resulted in enhanced cell death and elimination of LSCs, Jagged 1 in osteoblasts and subsequent increase of Notch
as assayed by limiting dilution analysis. These findings are signaling in leukemia-initiating HSCs. However, it remains
supported by clinical data from newly diagnosed patients to be determined whether this mechanism of niche-driven
with AML who received GCSF in conjunction with induc- oncogenesis occurs in human hematological malignancies.
tion chemotherapy, showing improved outcomes in terms of The relationship between the niche and leukemic cells
disease-free survival, although this benefit did not extend to is clearly not a one-way street and, just as the niche can
those with poor-risk disease, and no overall survival advan- dictate tumor behavior, so too can leukemic cells reshape
tage was noted. From a mechanistic point of view, it is unclear and manipulate their microenvironment in their favor at the
whether the enhanced chemosensitivity observed with GCSF expense of normal hematopoiesis. For example, in a mouse
relates solely to its effect on cell cycle progression, or whether model of BCR-ABL CML, reduced expression of CXCL12
there are other mechanisms involved, such as displacement was observed in the bone marrow stroma cells secondary
of LSCs from the niche and potential synergistic interaction to secreted factors by leukemic cells, leading to reduced
with chemotherapy. engraftment of normal HSCs, but not of BCR-ABL-positive
The use of arsenic trioxide, which is licensed for the treat- HSCs. Similarly, in a murine model of myeloproliferative
ment of PML/RARa acute promyelocytic leukemia, has also neoplasm (MPN), neoplastic myeloid cells were shown to
been suggested as an alternative intervention to enhance the remodel the endosteal niche by promoting the differentiation
cycling of LSCs and, consequently, their elimination. In a of mesenchymal stroma cells toward the osteoblastic lineage.
CML mouse model, loss of PML led to LSC exhaustion and Notably, the expanded osteoblasts were functionally altered,
diminished repopulating ability. Furthermore, PML inhibi- with compromised ability to support normal HSCs but not
tion with arsenic trioxide in both murine and human CML- LSCs. AML-induced alterations in endothelial cells resulting
LSC resulted in cell cycle induction which was more pro- in increased vascular permeability and sympathetic neuropa-
nounced compared to normal HSC, and was associated with thy of the niche in MPN are among other reported examples
enhanced chemosensitivity. of neoplasia-induced remodeling of the bone marrow niche.
Given the important role of the niche in sustaining tumor
development, targeting the niche has been explored as a
Targeting the CSC niche
potential therapeutic strategy. One approach has been to dis-
The bone marrow niche represents a complex and dynamic rupt homing of LSCs by targeting the CXCL12/CXC4 axis in
microenvironment which provides a host of signaling cues leukemia. Pretreatment of AML cells with CXCR4 antibodies
that are crucial for determining the fate decision of normal has been shown to impair their homing into the bone marrow
368 Molecular Hematology

mesenchymal
stroma cells
Sympathetic neurons

Osteoclast

?
? Endothelial
cells
Bone

Osteoblast
? Blood vessel

LSC ?
?

Endosteal niche Vascular niche


Adipocvtes
Fig. 26.2 Schematic representation of the interplay between the niche and leukemic cells. The bone marrow (BM) niche consists of
several cellular components including mesenchymal stroma cells, osteoblasts, endothelial cells, adipocytes, and sympathetic neurons. The
leukemia/microenvironment interaction is a dynamic, bidirectional process whereby leukemic cells receive vital cues from the niche that influence
their behavior. Conversely, leukemic cells hijack and remodel the BM niche into a malignant environment that is more permissive to the disease at
the expense of normal hematopoiesis. LSC, leukemic stem cells.

and spleen of transplanted mice. Conversely, mice already marrow stromal and endothelial cells, and inhibiting
engrafted with AML and treated with CXCR4-neutralizing the VLA-4/VCAM-1 interaction induces mobilization of
antibodies had marked reduction in their AML burden, but hematopoietic/stem progenitor cells. In AML, interaction
no significant difference in the level of engraftment was between VLA-4 on leukemic cells and fibronectin has been
observed in mice engrafted with cord blood mononuclear shown to enhance their survival and reduce their chemosen-
cells, indicating that AML cells are more CXCR4 dependent. sitivity through activation of PI-3k/AKT/BCL2 signaling.
Moreover, using a different small molecule CXCL12/CXCR4 Combination therapy with the anti-VLA4 monoclonal
inhibitor, CXCR4 inhibition abrogated the protection con- antibody Natalizumab (NZM) and cytarabine resulted in
ferred by the stroma, resulting in increased sensitivity to increased AML apoptosis, and mice treated with single-agent
chemotherapy. Of note is that higher expression of CXCR4 NZM had a significant reduction in their AML burden.
has been reported in patients with AML, and CXCR4 expres- Alternative therapeutic approaches include targeting the
sion level appeared to be an independent prognostic indi- adhesion molecule CD44. A monoclonal antibody directed
cator across a heterogeneous group of AML patients with against CD44 resulted in significant reduction of leukemic
varying mutational backgrounds. Given the role of CXCR4 in burden in mice transplanted with AML. This effect was medi-
homing, it is not unreasonable to speculate that the negative ated not only by impeding LSC homing, but also by altering
impact on outcome may, at least in part, be due to enhanced the fate of LSCs. A major drawback, however, is that CD44
CXCR4-mediated retention of LSCs. is also expressed on normal HSCs, and ligation of CD44 has
Disrupting the LSC–niche interaction can also be achieved been shown to affect the migratory behavior of normal HSCs.
by targeting the integrin very late antigen 4 (VLA4), which
is known for its important role in homing and retention
of HSCs in the niche. VLA-4 binds to the fibronectin Targeting epigenetic modifiers
component of the extracellular matrix as well as the vascular The use of epigenetic modulating therapies as a means to
cell adhesion molecular-1 (VCAM-1), expressed by bone target CSCs in hematological malignancies has been mainly
Cancer stem cells 369

explored in AML, in which mutations in epigenetic modifiers inhibition when treated with combination of a tyrosine
are a frequent event, particularly in MLL-rearranged AML, kinase inhibitor and tigecycline, an antibiotic that inhibits
which is primarily driven by epigenetic dysregulation. mitochondrial protein translation. Reliance on oxida-
The DNA methyltransferase inhibitors, azacytidine and tive phosphorylation rather than glycolysis has also been
decitabine, are currently the only two approved epigenetic reported in AML, where LSCs have been shown to have lower
targeted therapies in AML. The effect of azacitidine on AML levels of endogenous reactive oxygen species (ROS). Fur-
LSCs has been investigated by Craddock and colleagues, who thermore, BCL-2 was upregulated in ROS-low LSCs, which
found a substantial reduction in the LSC pool size, although when inhibited resulted in elimination of LSC both in vitro
it was never eradicated, even in patients who achieved com- and in vivo. Similarly, in AML with isocitrate dehydrogenase
plete morphological remission. (IDH) mutations, an RNA interference screen revealed syn-
Histone methyltransferase inhibitors are another class of thetic lethality between IDH1 and BCL-2. Treatment with
epigenetic therapies that have shown demonstrable effect BCL-2 inhibitor induced apoptosis in AML cells and affected
against LSCs. Key among these is 3-Deazaneplanocin A the survival of LSCs in xenotransplants through a mecha-
(DZNep), which targets several components of the Polycomb nism involving 2-hydroxybutarate-mediated inhibition of
repressor complex 2, including EZH2. In a study by Zhou and cytochrome c oxidase activity in the mitochondrial electron
colleagues, DZNep demonstrated significant anti-leukemic transport chain.
effect, including in LSC-enriched populations, with no The role of hypoxia in maintaining normal HSC func-
observed impact on normal HSCs in colony-forming assays. tions, especially self-renewal, is well recognized. Similarly,
However, more recently, loss of EZH2 has been reported hypoxia is important for the maintenance of LSCs. Wang
to correlate with poor prognosis and chemoresistance in et al. reported that in patients with AML, HIF1a and its
AML, and treatment of AML cells with DZNeP was found target GLUT1 were selectively overexpressed in the LSC
to induce resistance to cytarabine. In contrast to AML, fraction. Furthermore, inhibition of HIF-1α by echinomycin
overexpression of EZH2 has been reported in CML LSCs, induced apoptosis, which was more pronounced in the LSC
and inactivation of EZH2, in a conditional mouse model and fraction compared to the AML bulk population. The selec-
by clustered regularly interspaced short palindromic repeats tive elimination of the LSC population by HIF inhibition was
(CRISPR)/Cas9-mediated gene editing, has been shown to also demonstrated in vivo by serial transplantation exper-
block disease initiation and impair CML LSC function. Sim- iments, where mice injected with residual leukemic cells
ilar findings were reported by Scott et al., who showed a syn- from echinomycin-treated mice failed to develop leukemia,
ergistic impact on the CML LSC compartment when EZH2 indicating an absence of LSCs in the residual leukemic
inhibitors were used in conjunction with tyrosine kinase population. Similar effects of HIF-1α inhibition have been
inhibitors. reported in CML LSCs. It is worthy of note, however, that the
In addition to EZH2, the histone 3 lysine 79 (H3K79) inhibition of HIF by echinomycin is not restricted to HIF-1α,
methyltransferase Dot1l has emerged as a critical regulator and that the effect of echinomycin may, at least in part, be
of LSC activity. In MLL-AF9 induced leukemia, Dot1l has mediated by suppression of HIF-2α, which has been demon-
been shown to play an important role in promoting LSC strated to protect both normal HSC and AML cells from
self-renewal, and DOT1l inhibitors are currently being endoplasmic stress-induced apoptosis.
investigated in a number of clinical trials. Other histone
modifiers with the potential to modulate LSC activity in AML
include the histone demethylases KDM2b/JHDM1b and Conclusion
LSD1/KMD1A.
Furthermore, anti-leukemic effect against AML LSC has The mainstay treatment in patients with hematological
been shown with the histone deacetylase inhibitor AR-42, malignancies remains traditional therapies that target the
which causes inactivation of NF-κB, as well as inhibitors tumor bulk but leave CSCs largely untouched, potentially
against the chromatin reader Bromodomain-containing pro- contributing to chemoresistance and disease relapse. In
tein 4 (BRD4). recent years, tremendous advances have been made in our
understanding of CSC biology, which paved the way for the
development of a number of therapeutic agents that tar-
Other CSC-targeted therapies
get CSCs. While promising, several considerations should
LSCs can be distinguished from normal HSCs by virtue of be taken into account before CSC-targeted therapies can be
their distinct metabolic traits. In a study of chronic-phase incorporated into routine clinical practice.
CML, the stem cell–enriched population exhibited an The timing of administration of a CSC-directed therapy is
increase in oxidative metabolism compared to normal HSCs, likely to influence its efficacy. Ideally, a CSC-targeted therapy
and was more susceptible to oxidative phosphorylation should be given before standard chemotherapy when CSCs
370 Molecular Hematology

are still in a naı̈ve state, as the selective pressure exerted by Singh, S.K., Hawkins, C., Clarke, I.D. et al. (2004). Identification of
chemotherapy can inadvertently lead to the acquisition of human brain tumour initiating cells. Nature 432: 396–401.
further mutations in CSCs, making them more complex Visvader, J.E. and Lindeman, G.J. (2008). Cancer stem cells in solid
and potentially drug resistant. However, this may not be tumours: accumulating evidence and unresolved questions. Nat. Rev.
pragmatic in highly proliferative malignancies such as AML, Cancer 8: 755–768.
and the concurrent use of CSC-targeted therapies and “de-
bulking” chemotherapy would therefore be more appropri- The cell of origin
ate. It has also been proposed to use CSC-targeted treatment
in the post-remission setting, either as part of the consolida- Cozzio, A., Passegué, E., Ayton, P.M. et al. (2003). Similar MLL-
associated leukemias arising from self-renewing stem cells and short-
tion regimen or as long-term maintenance strategy in order
lived myeloid progenitors. Genes Dev. 17: 3029–3035.
to eradicate residual CSCs.
Goardon, N., Marchi, E., Atzberger, A. et al. (2011). Coexistence of
Additionally, caution should be exercised in applying LMPP-like and GMP-like leukemia stem cells in acute myeloid
the same clinical endpoints used to measure the efficacy of leukemia. Cancer Cell 19: 138–152.
standard cancer therapies, as they could potentially result in Jamieson, C.H.M., Ailles, L.E., Dylla, S.J. et al. (2004). Granulocyte–
significant underestimates of ultimate clinical benefit, lead- macrophage progenitors as candidate leukemic stem cells in blast-
ing to erroneous conclusions about their efficacy. To make an crisis CML. N. Engl. J. Med. 351: 657–667.
accurate assessment of response rates, long follow-up periods Kreso, A. and Dick, J.E. (2014). Evolution of the cancer stem cell model.
are required; however, in the short term, clinical endpoints Cell Stem Cell 14: 275–291.
such as progression-free and overall survival may be more Krivtsov, A.V., Twomey, D., Feng, Z. et al. (2006). Transformation from
meaningful. committed progenitor to leukaemia stem cell initiated by MLL-AF9.
Nature 442: 818–822.
Validation of CSC-targeted therapies in the in vivo setting
Miyamoto, T., Weissman, I.L., and Akashi, K. (2000). AML1/ETO-
is also paramount, as stemness is a functional definition. This expressing nonleukemic stem cells in acute myelogenous leukemia
would require having reliable methods to isolate CSCs, which with 8;21 chromosomal translocation. Proc. Natl. Acad. Sci. U.S.A.
pose an additional challenge, as CSCs can be phenotypically 97: 7521–7526.
unstable. Finally, a rational selection of combination thera- Taussig, D.C., Vargaftig, J., Miraki-Moud, F. et al. (2010). Leukemia-
pies is required in order to maximize efficacy and reduce the initiating cells from some acute myeloid leukemia patients with
emergence of resistance. mutated nucleophosmin reside in the CD34- fraction. Blood 115:
1976–1984.

Further reading Pre-leukemic stem cells

The cancer stem cell concept Corces-Zimmerman, M.R., Hong, W.-J., Weissman, I.L. et al. (2014).
Preleukemic mutations in human acute myeloid leukemia affect epi-
Ailles, L.E. and Weissman, I.L. (2007). Cancer stem cells in solid tumors. genetic regulators and persist in remission. Proc. Natl. Acad. Sci.
Curr. Opin. Biotechnol. 18: 460–466. U.S.A. 111: 2548–2553.
Al-Hajj, M., Wicha, M.S., Benito-Hernandez, A. et al. (2003). Prospec- Ding, L., Ley, T.J., Larson, D.E. et al. (2012). Clonal evolution in relapsed
tive identification of tumorigenic breast cancer cells. Proc. Natl. Acad. acute myeloid leukaemia revealed by whole-genome sequencing.
Sci. U.S.A. 100: 3983–3988. Nature 481: 506–510.
Baccelli, I. and Trumpp, A. (2012). The evolving concept of cancer and Fialkow, P.J., Janssen, J.W., and Bartram, C.R. (1991). Clonal remissions
metastasis stem cells. J. Cell Biol. 198: 281–293. in acute nonlymphocytic leukemia: evidence for a multistep patho-
Bonnet, D. and Dick, J.E. (1997). Human acute myeloid leukemia is genesis of the malignancy. Blood 77: 1415–1417.
organized as a hierarchy that originates from a primitive hematopoi- Fialkow, P.J., Singer, J.W., Raskind, W.H. et al. (1987). Clonal develop-
etic cell. Nat. Med. 3: 730–737. ment, stem-cell differentiation, and clinical remissions in acute non-
Maehle, A.H. (2011). Ambiguous cells: the emergence of the stem cell lymphocytic leukemia. N. Engl. J. Med. 317: 468–473.
concept in the nineteenth and twentieth centuries. Notes Rec. R. Soc. Genovese, G., Kähler, A.K., Handsaker, R.E. et al. (2014). Clonal
65: 359–378. hematopoiesis and blood-cancer risk inferred from blood DNA
Marjanovic, N.D., Weinberg, R.A., and Chaffer, C.L. (2013). Cell plas- sequence. N. Engl. J. Med. 371: 2477–2487.
ticity and heterogeneity in cancer. Clin. Chem. 59: 168–179. Jaiswal, S., Fontanillas, P., Flannick, J. et al. (2014). Age-related clonal
Quintana, E., Shackleton, M., Sabel, M.S. et al. (2008). Efficient tumour hematopoiesis associated with adverse outcomes. N. Engl. J. Med. 371:
formation by single human melanoma cells. Nature 456: 593–598. 2488–2498.
Schatton, T., Murphy, G.F., Frank, N.Y. et al. (2008). Identification of Jan, M., Snyder, T.M., Corces-Zimmerman, M.R. et al. (2012). Clonal
cells initiating human melanomas. Nature 451: 345–349. evolution of preleukemic hematopoietic stem cells precedes human
Shackleton, M., Quintana, E., Fearon, E.R., and Morrison, S.J. (2009). acute myeloid leukemia. Sci. Transl. Med. 4: 149ra118.
Heterogeneity in cancer: cancer stem cells versus clonal evolution. Miyamoto, T., Nagafuji, K., Akashi, K. et al. (1996). Persistence of mul-
Cell 138: 822–829. tipotent progenitors expressing AML1/ETO transcripts in long-term
Cancer stem cells 371

remission patients with t(8;21) acute myelogenous leukemia. Blood Giambra, V., Jenkins, C.E., Lam, S.H. et al. (2015). Leukemia stem cells
87: 4789–4796. in T-ALL require active Hif1α and Wnt signaling. Blood 125: 3917–
Rothenberg-Thurley, M., Amler, S., Goerlich, D. et al. (2018). Persis- 3927.
tence of pre-leukemic clones during first remission and risk of relapse Tatarek, J., Cullion, K., Ashworth, T. et al. (2011). Notch1 inhibition
in acute myeloid leukemia. Leukemia 32: 1–11. targets the leukemia-initiating cells in a Tal1/Lmo2 mouse model of
Shlush, L.I., Zandi, S., Mitchell, A. et al. (2014). Identification of pre- T-ALL. Blood 118: 1579–1590.
leukaemic haematopoietic stem cells in acute leukaemia. Nature 506: Wang, Y., Krivtsov, A.V., Sinha, A.U. et al. (2010). The Wnt/beta-catenin
328–333. pathway is required for the development of leukemia stem cells in
Wong, T.N., Miller, C.A., Klco, J.M. et al. (2016). Rapid expansion of pre- AML. Science (New York, NY) 327: 1650–1653.
existing nonleukemic hematopoietic clones frequently follows induc- Yeung, J., Esposito, M.T., Gandillet, A. et al. (2010). β-Catenin mediates
tion therapy for de novo AML. Blood 127: 893–897. the establishment and drug resistance of MLL leukemic stem cells.
Xie, M., Lu, C., Wang, J. et al. (2014). Age-related mutations associated Cancer Cell 18: 606–618.
with clonal hematopoietic expansion and malignancies. Nat. Med. 20:
1472–1478.
Targeting CSC survival
Guzman, M.L., Neering, S.J., Upchurch, D. et al. (2001). Nuclear factor-
Targeting cell surface antigens
kappaB is constitutively activated in primitive human acute myeloge-
Hoseini, S.S. and Cheung, N.K. (2017). Acute myeloid leukemia targets nous leukemia cells. Blood 98: 2301–2307.
for bispecific antibodies. Blood Cancer J. 7: e522. Park, S., Chapuis, N., Tamburini, J. et al. (2010). Role of the PI3K/AKT
Jen, E.Y., Ko, C.-W., Lee, J.E. et al. (2018). FDA approval: gemtuzumab and mTOR signaling pathways in acute myeloid leukemia. Haemato-
ozogamicin for the treatment of adults with newly-diagnosed CD33- logica 95: 819–828.
positive acute myeloid leukemia. Clin. Cancer Res. 24 (14): 3242–
3246.
Targeting LSC dormancy
Jin, L., Lee, E.M., Ramshaw, H.S. et al. (2009). Monoclonal antibody-
mediated targeting of CD123, IL-3 receptor α chain, eliminates Ito, K., Bernardi, R., Morotti, A. et al. (2008). PML targeting eradicates
human acute myeloid leukemic stem cells. Cell Stem Cell 5: quiescent leukaemia-initiating cells. Nature 453: 1072–1078.
31–42. Löwenberg, B., van Putten, W., Theobald, M. et al. (2003). Effect of
Kikushige, Y., Shima, T., Takayanagi, S.I. et al. (2010). TIM-3 is a promis- priming with granulocyte colony-stimulating factor on the outcome
ing target to selectively kill acute myeloid leukemia stem cells. Cell of chemotherapy for acute myeloid leukemia. N. Engl. J. Med. 349:
Stem Cell 7: 708–717. 743–752.
Knapp, D.J.H.F., Hammond, C.A., Hui, T. et al. (2018). Single-cell anal- Saito, Y., Uchida, N., Tanaka, S. et al. (2010). Induction of cell cycle entry
ysis identifies a CD33+ subset of human cord blood cells with high eliminates human leukemia stem cells in a mouse model of AML. Nat.
regenerative potential. Nat. Cell Biol. 20: 710–720. Biotechnol. 28: 275–280.
Liu, K., Zhu, M., Huang, Y. et al. (2015). CD123 and its potential clinical
application in leukemias. Life Sci. 122: 59–64.
Majeti, R., Chao, M.P., Alizadeh, A.A. et al. (2009). CD47 is an adverse Targeting the CSC niche
prognostic factor and therapeutic antibody target on human acute Arranz, L., Sánchez-Aguilera, A., Martı́n-Pérez, D. et al. (2014). Neu-
myeloid leukemia stem cells. Cell 138: 286–299. ropathy of haematopoietic stem cell niche is essential for myelopro-
Mardiros, A., Forman, S.J., and Budde, L.E. (2015). T cells expressing liferative neoplasms. Nature 512: 78–81.
CD123 chimeric antigen receptors for treatment of acute myeloid Hsieh, Y., Jiang, E., Pham, J., et al. (2013). VLA4 blockade in acute
leukemia. Curr. Opin. Hematol. 22: 484–488. myeloid leukemia. In 55th ASH Annual Meeting and Exposition,
Taussig, D.C., Pearce, D.J., Simpson, C. et al. (2005). Hematopoietic stem 122, 7–13 December 2013. New Orleans, LA: American Society of
cells express multiple myeloid markers: implications for the origin Hematology.
and targeted therapy of acute myeloid leukemia. Blood 106: 4086– Jin, L., Hope, K.J., Zhai, Q. et al. (2006). Targeting of CD44 eradi-
4092. cates human acute myeloid leukemic stem cells. Nat. Med. 12: 1167–
Van Rhenen, A., Van Dongen, G.A.M.S., Kelder, A.L. et al. (2007). The 1174.
novel AML stem cell-associated antigen CLL-1 aids in discrimination Kode, A., Manavalan, J.S., Mosialou, I. et al. (2014). Leukaemogenesis
between normal and leukemic stem cells. Blood 110: 2659–2666. induced by an activating β-catenin mutation in osteoblasts. Nature
506: 240–244.
Targeting CSC self-renewal Matsunaga, T., Takemoto, N., Sato, T. et al. (2003). Interaction between
leukemic-cell VLA-4 and stromal fibronectin is a decisive factor for
Dierks, C., Beigi, R., Guo, G.-R. et al. (2008). Expansion of Bcr-Abl- minimal residual disease of acute myelogenous leukemia. Nat. Med.
positive leukemic stem cells is dependent on hedgehog pathway acti- 9: 1158–1165.
vation. Cancer Cell 14: 238–249. Passaro, D., Di Tullio, A., Abarrategi, A. et al. (2017). Increased vascular
Gang, E.J., Hsieh, Y.T., Pham, J. et al. (2014). Small-molecule inhibition permeability in the bone marrow microenvironment contributes to
of CBP/catenin interactions eliminates drug-resistant clones in acute disease progression and drug response in acute myeloid leukemia.
lymphoblastic leukemia. Oncogene 33: 2169–2178. Cancer Cell 32: 324–341.e6.
372 Molecular Hematology

Raaijmakers, M.H.G.P., Mukherjee, S., Guo, S. et al. (2010). Bone Herrmann, H., Blatt, K., Shi, J. et al. (2013). Small-molecule inhibition of
progenitor dysfunction induces myelodysplasia and secondary BRD4 as a new potent approach to eliminate leukemic stem- and pro-
leukaemia. Nature 464: 852–857. genitor cells in acute myeloid leukemia (AML). Oncotarget 3: 1588–
Schepers, K., Pietras, E.M., Reynaud, D. et al. (2013). Myeloprolifera- 1599.
tive neoplasia remodels the endosteal bone marrow niche into a self- Nguyen, A.T., Taranova, O., He, J., and Zhang, Y. (2011). DOT1L,
reinforcing leukemic niche. Cell Stem Cell 13: 285–299. the H3K79 methyltransferase, is required for MLL-AF9 - mediated
Spoo, A.C., Lübbert, M., Wierda, W.G., and Burger, J.A. (2007). CXCR4 leukemogenesis. Blood 117: 6912–6922.
is a prognostic marker in acute myelogenous leukemia. Blood 109: Scott, M.T., Korfi, K., Saffrey, P. et al. (2016). Epigenetic reprogramming
786–791. sensitizes CML stem cells to combined EZH2 and tyrosine kinase
Tavor, S., Petit, I., Porozov, S. et al. (2004). CXCR4 regulates migration inhibition. Cancer Discov. 6: 1248–1257.
and development of human acute myelogenous leukemia stem cells Xie, H., Peng, C., Huang, J. et al. (2016). Chronic myelogenous
in transplanted NOD/SCID mice. Cancer Res. 64: 2817–2824. leukemia-initiating cells require polycomb group protein EZH2.
Zeng, Z., Shi, Y.X., Samudio, I.J. et al. (2009). Targeting the leukemia Cancer Discov. 6: 1237–1247.
microenvironment by CXCR4 inhibition overcomes resistance to Zhou, J., Bi, C., Cheong, L.L. et al. (2011). The histone methyltransferase
kinase inhibitors and chemotherapy in AML. Blood 113: 6215–6224. inhibitor, DZNep, up-regulates TXNIP, increases ROS production,
Zhang, B., Ho, Y.W., Huang, Q. et al. (2012). Altered microenvironmen- and targets leukemia cells in AML. Blood 118: 2830–2839.
tal regulation of leukemic and normal stem cells in chronic myeloge-
nous leukemia. Cancer Cell 21: 577–592.
Other CSC-targeted therapies
Zöller, M. (2015). CD44, hyaluronan, the hematopoietic stem cell, and
leukemia-initiating cells. Front. Immunol. 6: 235. Chan, S.M., Thomas, D., Corces-Zimmerman, M.R. et al. (2015). Isoci-
trate dehydrogenase 1 and 2 mutations induce BCL-2 dependence in
Targeting epigenetic modifiers acute myeloid leukemia. Nat. Med. 21: 178–184.
Kuntz, E.M., Baquero, P., Michie, A.M. et al. (2017). Targeting mito-
Craddock, C., Quek, L., Goardon, N. et al. (2013). Azacitidine fails to chondrial oxidative phosphorylation eradicates therapy-resistant
eradicate leukemic stem/progenitor cell populations in patients with chronic myeloid leukemia stem cells. Nat. Med. 23: 1234–1240.
acute myeloid leukemia and myelodysplasia. Leukemia 27: 1028– Lagadinou, E.D., Sach, A., Callahan, K. et al. (2013). BCL-2 inhibition
1036. targets oxidative phosphorylation and selectively eradicates quiescent
Göllner, S., Oellerich, T., Agrawal-Singh, S. et al. (2017). Loss of the his- human leukemia stem cells. Cell Stem Cell 12: 329–341.
tone methyltransferase EZH2 induces resistance to multiple drugs in Rouault-Pierre, K., Lopez-Onieva, L., Foster, K. et al. (2013). HIF-2a
acute myeloid leukemia. Nat. Med. 23: 69–78. protects human hematopoietic stem/progenitors and acute myeloid
Guzman, M.L., Yang, N., Sharma, K.K. et al. (2014). Selective activity of leukemic cells from apoptosis induced by endoplasmic reticulum
the histone deacetylase inhibitor AR-42 against leukemia stem cells: a stress. Cell Stem Cell: 549–563.
novel potential strategy in acute myelogenous leukemia. Mol. Cancer Wang, Y., Liu, Y., Malek, S.N. et al. (2011). Targeting HIF1α eliminates
Ther. 13: 1979–1990. cancer stem cells in hematological malignancies. Cell Stem Cell 8:
Harris, W.J., Huang, X., Lynch, J.T. et al. (2012). The histone demethylase 399–411.
KDM1A sustains the oncogenic potential of MLL-AF9 leukemia stem
cells. Cancer Cell 21: 473–487.
Conclusion
He, J., Nguyen, A.T., and Zhang, Y. (2011). KDM2b/JHDM1b, an
H3K36me2-specific demethylase, is required for initiation and main- Pollyea, D.A. and Jordan, C.T. (2017). Therapeutic targeting of acute
tenance of acute myeloid leukemia. Blood 117: 3869–3880. myeloid leukemia stem cells. Blood 129: 1627–1635.
Chapter 27 Molecular basis of transplantation
Francesco Dazzi1,2 & Antonio Galleu1,2
1 School of Cancer & Pharmaceutical Sciences, King’s College London, London, UK
2
King’s Health Partners Cancer Research UK Centre, London, UK

Introduction, 373 Graft-versus-host and graft-versus-Leukemia, 379


Principles and clinical indications of HSCT, 373 Graft-versus-host disease, 381
Non-immunological factors regulating HSCT, 374 Conclusions, 386
Exploiting HSCT for resetting the immune system, 375 Further reading, 387
Basic concepts in the immunology of allogeneic HSCT, 376

own HSCs, harvested before, to restore the otherwise per-


Introduction manently ablated hemopoietic system. Although autologous
HSCT is useful in some solid tumors, its efficacy in the treat-
Transplantation is a successful therapeutic modality for a
ment of hemopoietic malignancies is limited by the contam-
variety of diseases of different etiology and pathogenesis.
ination of the harvested stem cells by the original tumor
Liver, heart, and kidney failures are common indications
and/or by the insufficient activity of the chemotherapy in
which would be even more widely pursued should the donor
eliminating the tumor itself. However, such an approach
availability not be so limited. Although the basic general
maintains some efficacy, because normal HSCs have a tempo-
principle underlying transplantation is the replacement of
rary growth advantage at repopulating the recipient as com-
a malfunctioning tissue with a healthy one, in the case of
pared to neoplastic stem cells.
hemopoietic stem cell transplantation (HSCT) the proce-
A more recent application of autologous HSCT has been
dure is associated with a number of other beneficial effects
the treatment of severe autoimmune diseases, whereby the
which can be exploited not only to restore hemopoietic
repopulation of the immune system with primitive HSCs
failure itself, but also for treating cancer and autoimmune
is believed to re-educate the ill immune system. Phase I/II
diseases.
trials have reported high response rates in systemic lupus
erythematosus, systemic sclerosis, rheumatoid arthritis, and
multiple sclerosis. Randomized studies are ongoing to com-
Principles and clinical indications of pare these achievements to conventional immunosuppressive
HSCT therapies.
When a compatible donor is available, the use of allo-
Hematopoietic stem cells (HSCs) have the capacity to self- geneic HSCT has profoundly modified the outcome of several
renew and give rise to all formed elements in the blood. hematological malignancies. The conditioning regimen con-
Because of this property, they can be used to rescue the tributes to the eradication of the abnormal cells and ensures
hemopoietic system from the intensification of anti-cancer sustained engraftment of the healthy allogeneic stem cells.
cytotoxic therapies. HSCs can be autologous when donor and However, the efficacy of this approach cannot simply be
recipient are the same individual, or allogeneic if another ascribed to the chemo-radiotherapy nor to the administra-
individual is selected as the HSC donor. In autologous tion of healthy HSCs, but is greatly dependent on the immune
HSCT, patients can be subjected to lethal doses of chemo- recognition of the tumor by the lymphocytes contained in the
radiotherapy to eradicate the tumor and then receive their donor stem cell preparation.
From these preliminary considerations, it is clear how sev-
eral mechanisms contribute to the outcome of HSCT and
these involve HSC engraftment, expansion, and differentia-
Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben. tion as well as, in the case of allogeneic HSCT, the interplay
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd. between donor and recipient immune responses.

373
374 Molecular Hematology

early progenitors has been proposed based on the selec-


Non-immunological factors regulating tive expression of signaling lymphocyte activation molecule
HSCT (SLAM) family receptors. The highly enriched bone marrow
HSC population is marked by the expression of CD150 and
HSCs normally face four different outcomes: self-renewal,
the absence of CD224 and CD48, whereas multipotent pro-
differentiation, mobilization, or programmed cell death. All
genitors and restricted B-cell progenitors selectively express
these processes are tightly regulated and play a fundamen-
only CD224 and CD48, respectively. A similar expression
tal role during transplantation. Since HSCs are the source
pattern of SLAM receptors has been described in fetal
of every blood cell, their self-renewal and lifespan must be
hematopoiesis.
securely controlled. For this reason, a proportion of HSCs
The number of HSCs infused after the conditioning reg-
remains quiescent, but available to contribute to blood home-
imen remains a major prognostic factor to predict the
ostatic renewal. These HSCs reside in specialized areas of the
outcome of clinical HSCT. The larger the number of
bone marrow (BM) in which they receive signals required for
HSCs administered, the faster the recovery of recipient
their homeostatic quiescent and stationary state. These HSC
hemopoiesis and the lower the transplant-related mortality.
niches are constituted by endosteal bone-lining osteoblasts
Therefore, much effort is being spent with a view to expand-
and other stromal cells which are responsible for generating
ing the number of HSCs ex vivo with a variety of cytokine
the signals. The niche saves stem cells from depletion, while
cocktails, although very limited success has been achieved
protecting the host from unnecessary stem cell proliferation.
thus far, because expansion is almost invariably associated
with differentiation. However, an aryl hydrocarbon receptor
antagonist (StemRegenin-1, SR-1) that expands CD34+ cells
The hemopoietic stem cell
has been shown to produce a 330-fold increase, with a dra-
Probably the best definition of HSC is functional and refers to matic impact on patient engraftment.
a cell capable of providing lifelong reconstitution of all blood
cell lineages after serial transplantation into lethally irradi-
The hemopoietic niche
ated recipients.
In mice, the phenotype of HSCs is characterized by the In the postnatal marrow, stromal cells form a three-
surface expression of Sca-1 (stem cell antigen-1), c-Kit dimensional network investing marrow sinusoids. Four main
(receptor for stem cell factor), with associated low levels cell types of the marrow stromal tissue are known to take
of Thy-1 (CD90) expression, and the absence of other part in supporting hemopoiesis: one is of hemopoietic origin
lineage-specific markers: Lin− Thy1.1+lo c-Kit+ Sca1+ . The (macrophages), whereas the others derive from mesenchy-
biological activity of the candidate mouse HSC population mal progenitors (reticular cells, adipocytes, and osteoblasts).
has been enumerated by in vitro assays and in vivo competi- Among the various players, osteoblasts expressing N-
tive repopulation assays. These functional assays have shown cadherin are the most important component of the niche.
that extensive self-renewal potential and proliferation ability Osteoblasts control of HSC proliferation within the niche
are gradually lost during commitment and have defined a is regulated by both cell contact mechanisms and soluble
hierarchy in the hematopoiesis. Long-term hematopoietic factors, which mediate their effects via transcription factors,
stem cells (LT-HSCs) give rise to short-term hematopoietic cell cycle regulators, adhesion molecules, and chromoso-
stem cells (ST-HSCs) that self-renew for six to eight weeks mal modifiers. These include a series of molecules which
only, then progress to multipotent progenitors (MPPs), regulate the quiescent state of HSCs and their population
which have very limited self-renewal ability and are unable size, including Notch1/Jagged1, the Wnt/β-catenin signaling
to durably engraft in lethally irradiated mice. pathways, Tie-2/Ang-1, osteopontin, and tenascin C. Ani-
In humans, HSCs have been characterized by the expres- mals deficient in these molecules have a reduced number
sion of the cell surface antigens CD34, c-Kit, Thy-1 (CD90), and function of HSCs. A wide variety of cytokines produced
and the absence of CD38 and other lineage-specific markers, by stromal cells has also been described, they maintain HSCs
and are therefore named Lin−c-Kit+Thy-1+CD38−CD34+. in quiescence or promote their self-renewal rather than
The phenotype is the results of functional studies conducted differentiation. Among these are stem cell factor, leukemia
not only in vitro, but also using strains of immunodeficient inhibitory factor, the SDF-1/CXCL12 chemokine axis, bone
mice suitable for human HSC engraftment. morphogenetic protein-4 (BMP-4), the cytokine fms-like
Further studies have identified the heterogeneity of the tyrosine kinase-3 (Flt-3), and transforming growth factor-β
HSC compartment and identified new markers, like CD133, (TGF-β). Stroma also produces a variety of interleukins
which is unique to a rare CD34– subset with LT-HSC func- and, under certain circumstances, also cytokines, which act
tion in vivo, and can similarly identify stem cells of other tis- on more mature hemopoietic progenitors, such as gran-
sue origin. Very recently a new classification of HSCs and ulocyte macrophage colony-stimulating factor (GM-CSF)
Molecular basis of transplantation 375

and granulocyte colony-stimulating factor (G-CSF). Lastly, clinical trials have demonstrated its efficacy even in patients
adhesion molecules also play an important role in the or donors refractory to G-CSF.
control of HSC proliferation, because the engagement of β-1
integrins prevents the progression of CD34+ cells from the The concept of BM space and
G1 to S phase of the cell cycle, thus inhibiting progenitor cell competition
proliferation.
A large bulk of studies in both animals and humans have
Although osteoblasts are ultimately the stromal cell mak-
shown that recipients of HSCs must receive some form
ing contact with and regulating the HSC compartment, bone
of myeloablation for the HSCs to engraft, irrespective of
is continuously subject to bone remodeling, a process gen-
whether there is an underlying malignancy to eradicate or
erated by the cooperation of osteoblasts, the bone-forming
an allogeneic immune system to suppress. This has intro-
cells, and osteoclasts, the bone-resorbing cells. As part of
duced the notion that the quantitative reduction of host cel-
this tightly regulated process, osteoblasts regulate osteoclast
lular components provides “space” for the incoming HSCs
maturation and proliferation, and there is emerging evidence
to expand and compete more favorably with recipient cells,
that the hemopoietic niche is the result of this process. In
reduced in numbers by the conditioning regimen. Further-
fact, pharmacological agents with the ability to modulate the
more, there is also evidence that such homeostatic expan-
number and/or functions of these cells have an impact on the
sion might also affect some of the immunological mecha-
number of HSCs and their ability to engraft in myeloablated
nisms involved in transplantation tolerance that we will dis-
recipients.
cuss later.
However, “opening space” is not the only mechanism at
Mobilization and homing of HSCs the basis of engraftment. High levels of long-term marrow
engraftment are obtained with the infusion of high levels of
The function of the niche is not confined to the control of
marrow cells in untreated mice. This indicates that syngeneic
HSC size, but also to their mobilization. The ability to migrate
engraftment is also determined by stem cell competition in
is a fundamental property of HSCs throughout their devel-
the hemopoietic niche.
opment. During ontogeny, blood formation occurs in dis-
tinct extra-embryonic and embryonic sites which involve in
sequence the yolk sac, aorto-gonadomesonephros (AGM), Exploiting HSCT for resetting the
placenta, and fetal liver, before hemopoiesis finally takes immune system
place only in the bone marrow. Migration from one site to
another is critical, as demonstrated by the fact that mice made While the long-term efficacy of autologous HSCT in malig-
genetically deficient in CXCL12, a chemokine responsible nancies is confined to a minority of tumors, the fact that
for the homing of HSCs to the bone marrow, or its receptor HSCs are also the precursors of immune cells has prompted
CXCR4, have normal production of fetal liver HSCs, but fail investigators to test the ability of autologous HSCT to treat
to transition to hematopoiesis in the marrow space. conditions characterized by abnormal immune responses. In
The migratory nature of HSCs is maintained in adulthood, support of this initiative is the evidence that the maturation
during which HSCs are found in the blood even under home- of new T cells in the thymus continues, albeit at a decreased
ostatic conditions. Following the administration of cytokines, rate, also in adult life.
including G-CSF, GM-CSF, and interleukin-8 (IL-8), the Autologous HSCT is currently being explored with
number of HSCs in the peripheral blood changes dramat- remarkable success in severe forms of autoimmune diseases,
ically, with different kinetics depending on the mechanism including multiple sclerosis, systemic lupus erythematosus,
involved. For example, G-CSF can modify niche cells by systemic sclerosis, rheumatoid arthritis, and Crohn’s disease.
reducing the production of CXCL12 by osteoblasts, despite The main rationale for applying HSCT to autoimmune
the absence of G-CSF receptors on these cells. Of particular diseases has been the idea that intensive immune deple-
interest is the observation that mice with altered sympathetic tion could eliminate the pathogenic repertoire, and that
nervous system function lack the ability to mobilize stem cells reconstitution of a new immune system from hematopoietic
from the bone marrow in response to G-CSF, thus indicating precursors could restore immune tolerance, halting ongoing
the possibility that stem cells might be able to “sense” changes inflammatory activity and preventing relapses.
such as those occurring distantly at the site of an injury. Recent studies have confirmed the notion that HSCT
Further developments in the area of HSC mobilization induces alterations of the immune system which are
have recently identified the use of chemokine antagonists beyond the effects of a dose-escalating immunosuppressive
as an alternative and probably more effective strategy than approach. HSCT has been shown not only to affect the B-cell
G-CSG. AMD3100, a specific antagonist of the chemokine populations associated with the production of autoantibod-
receptor CXCR4, is currently in clinical development, and ies, but also to profoundly perturb the T-cell compartment,
376 Molecular Hematology

as illustrated by the normalization of the deregulated T-cell HLA-E, HLA-F, and HLA-G, with functions which have not
receptor (TCR) repertoire in multiple sclerosis. Furthermore, yet been fully elucidated.
it appears that following myeloablation, a subset of T cells The role of MHC molecules is to present peptide antigens
(regulatory T cells) with the specific function of controlling to T cells. Class I molecules present endogenous peptides,
immunity to self antigen selectively expands and could con- like those that are virus or tumor encoded, which are
tribute to the control of the underlying autoimmune disease. generated in the cytosol, transported to the endoplasmic
On the contrary, the immune reconstitution following reticulum (ER), and finally presented to the cell surface. HLA
allogeneic HSCT remains incomplete for several months class II molecules are assembled in the ER, then transported
or years, depending on the histocompatibility differences through the Golgi to endosomal compartments, where they
between donor and recipient. This is one of the several prob- load peptides that have entered the cell via endocytosis or
lems associated with the various immune responses gener- receptor-mediated internalization. The selection of peptides
ated in an allogeneic setting. by MHC molecules is dictated by the sequence of the MHC
antigen-binding groove. Although MHC molecules have
limited polymorphism as compared to TCRs, they exhibit
Basic concepts in the immunology of different avidity for different peptides, thus accounting for
allogeneic HSCT individual variability to respond to the same antigen and
against different moieties.
Allogeneic HSCT triggers a network of immune responses
which fundamentally affect the outcome of the procedure,
Mechanisms of allorecognition
in terms of both complications and therapeutic success.
Whereas recipient anti-donor immune responses (host ver- Allorecognition is a particular form of antigen presenta-
sus graft, HvG) are important in solid organ transplan- tion, occurring only after transplantation of tissues between
tation, in allogeneic stem cell transplantation (SCT) they genetically disparate individuals of the same species (or after
are profoundly inhibited by the conditioning regimen. The in vitro simulation of such). HvG reactions refer to the
immunologically competent cells, present in the HSC prepa- immune response of the host to disparate antigens expressed
ration, play a more important role because they mediate a on donor cells, which results in graft rejection. The current
reaction against the host which targets recipient normal tis- understanding of HvG responses derives mainly from stud-
sues (graft-versus-host, GvH), but also mediate the effect at ies of solid organ transplantation, in which the host does
the basis of the eradication of residual neoplastic cells (graft- not receive any conditioning prior to the transplant and thus
versus-leukemia, GvL). maintains the ability to reject it.
Three pathways of allorecognition have been described:
direct, indirect, and the most recently described semi-direct
The major histocompatibility complex
allorecognition (Figure 27.1). The term direct allorecogni-
The major histocompatibility complex (MHC) defines a tion was initially used to describe the recognition by host
genetic region which includes genes encoding class I and T cells of intact donor MHC-peptide complex directly on
class II membrane-bound cell surface glycoproteins. The the surface of donor antigen-presenting cells (APCs), but
function of MHC proteins is to present peptide antigens to it has also later been extended to include the recognition
T cells, a vital part of initiating an antigen-specific immune of other donor-derived transplantation antigens (minor his-
response. MHC proteins are also involved in the recognition tocompatibility antigens) presented on donor APCs by an
of virally infected cells, or those cells in which genetic anoma- MHC molecule that is shared between donor and recipient.
lies arise, by natural killer (NK) cells. The frequency of alloreactive T cells using this pathway has
There are two major classes of genes within the MHC been estimated at between 0.1% and 10%, as compared to
region, namely class I and class II MHC genes. In addition approximately 10−5 for nominal peptide antigens. An expla-
to these, the MHC class III region encodes other proteins of nation for such a high frequency is that direct allorecognition
the immune system, like certain complement and cytokine arises as a consequence of the cross-reactivity of self MHC-
genes. In humans, the MHC region is found on the short arm restricted T cells.
of chromosome 6 and encodes for the human leukocyte anti- Indirect allorecognition is the recognition of donor-
gens (HLAs). Different loci are designated by a letter, thus the derived antigens which have been processed and are
major class I loci are HLA-A, HLA-B, and HLA-C. HLA class presented on the cell surface of host APCs in the context
II genes are collectively designated HLA-D, and individual of self MHC. Therefore, the distinction between direct and
loci identified by a second letter, HLA-DR, HLA-DP, HLA- indirect presentation is thus the source of the APCs on
DQ. In addition to the “classical” class I and class II MHC which alloantigens are presented. The mechanism of indirect
genes, there exists a number of non-classical MHC genes, like allorecognition is indistinguishable from the physiological
Molecular basis of transplantation 377

Direct presentation Indirect presentation Semi-direct presentation

MHC TCR

Fig. 27.1 Mechanisms of allo-recognition. MHC,


major histocompatibility complex; TCR, T-cell Donor
receptor. Recipient T cells

processing and presentation of pathogen-derived peptide transplantation. The contribution of this pathway to the
antigens. Donor MHC molecules can be processed, and rejection of the transplanted organ is yet to be elucidated.
peptide fragments presented to T cells, by host APCs.
Although only approximately 5–10% of alloreactive T cells
Transplantation tolerance: clonal deletion
are specific for indirectly presented donor antigens, they
are still thought to play a major role in chronic allograft The establishment of tolerance to antigens expressed by
rejection, which occurs at a time when donor APCs are no donor tissues is a major goal of allogeneic transplantation.
longer thought to be present. The mechanisms by which There are three main mechanisms which contribute to trans-
indirectly primed T cells mediate graft rejection are unclear. plantation tolerance: clonal deletion, anergy, and peripheral
Following organ transplantation, the donor endothelial regulation (Figure 27.2).
layer is repopulated with recipient cells. One theory is that During T-cell ontogeny, intrathymic clonal deletion of self-
donor antigen is presented to direct-pathway T cells by recip- reactive T cells is the primary process for the selection of the
ient MHC I by the recipient endothelium. An alternative mature T-cell repertoire. The transplantation of donor HSCs
hypothesis is that graft destruction is the result of bystander into sublethally or lethally myeloablated recipients gives rise
killing following re-encounter of antigen on the surface of to an immune system which is immunologically tolerant to
graft-infiltrating APCs. donor antigens. The chimerism is established not only at the
More recently, a predominant role of CD4+ T cells level of the BM, but also in the thymus, where recipient T
has been advocated, whereby they orchestrate both the cells learn to recognize the donor as “self” after interaction
production of donor-specific antibodies and the initiation with donor-derived professional APCs, which orchestrate the
of inflammatory signals through myeloid cells. Following continuous depletion of alloreactive T and B cells. Since it is
the presentation of donor-derived antigens, host B cells are mainly based on clonal deletion, such tolerance is long term
induced to produce specific antibodies by CD4+ T cells. Allo- and does not require immunosuppression except for the first
graft injury is ultimately mediated through opsonization and few months after the transplant.
activation of host NK and macrophages. Host macrophages There is now plenty of anecdotal evidence that patients
can also interact with CD4+ T cells as host APCs. undergoing allogeneic HSCT can subsequently receive an
A further mechanism of allorecognition has been organ from the original HSCT donor without the need for
described as the semi-direct presentation of donor antigens immunosuppression. The induction of allograft tolerance
and the underlying mechanisms identified very recently. through HSCT in both HLA-matched and mismatched
The original hypothesis assumed that intact donor-derived kidney transplants has been actively pursued in three major
MHC complexes were transferred to and presented by North American centers (Stanford, Northwestern University,
host-derived APCs to activate alloantigen-specific CD4+ and Massachusetts General Hospital). By 2017, more than
T cells through the indirect pathway and specific CD8+ T 80 patients had undergone this approach, and the results
cells through the direct pathway. It was recently found that of these clinical trials seem very encouraging, with the vast
high numbers of recipient APCs cross-dressed with donor majority of patients (50–76%) being off immunosuppression,
MHC are present in the lymph nodes draining the graft. with improved quality of life and very low transplant-related
Donor MHCs are derived from allogeneic exosomes, which mortality rates (0–5%). Also negligible is the occur-
can induce pro-inflammatory allo-responses even without rence of graft-versus-host disease (GvHD) and infectious
378 Molecular Hematology

Recipient thymus

Natural Treg
Clonal deletion
Donor stem cells

Selection of
mature T cell
repertoire Induced Treg

Recipient
bone marrow

Naïve and
memory T cells

Fig. 27.2 Mechanisms of transplant tolerance.


Peripheral anergy Treg, T-regulatory cells.

complications. Graft rejection is less than 25% at long-term or if anergic T cells are cultured in the absence of their
follow-ups. Notably, although in most cases hematopoietic cognate antigen. The blockade of costimulatory pathways
chimerism was achieved during the induction phase of the has been widely exploited as a mechanism to induce tol-
treatment, it was often not durable, thus questioning its erance in solid organ transplantation and HSCT. In the
requirement in the maintenance of the allograft tolerance context of experimental models of allogeneic HSCT, the
in the long term. A better understanding of the underlying administration of anti-CD154 mAb was shown to induce
mechanisms of tolerance induction after HSCT and the mixed lympho-hematopoietic chimerism and subsequent
improvement of the safety of the protocols used will cer- permanent skin graft acceptance if used as a single agent, in
tainly extend the success of this approach to allografts from combination with reduced-intensity HSCT, or in combina-
non-living donors. tion with the antagonists of CTLA-4. However, there is little
Clonal deletion can also occur at extra-thymic sites evidence in support of a role for anergy in the induction of
and can account for the removal of mature alloreactive T transplantation tolerance.
cells, thus leading to the establishment and maintenance of
donor-specific tolerance in experimental models of mixed
Transplantation tolerance: peripheral
chimerism induction following MHC-mismatched HSCT. T
regulation
cells are deleted in the periphery by either activation-induced
cell death (AICD) or passive cell death, both leading to Since the demonstration that tolerance to allogeneic graft can
apoptosis. be transferred to naı̈ve recipients by T cells derived from a
T-cell anergy describes a persistent state of unrespon- tolerant host, induction and maintenance of transplantation
siveness of T cells to their cognate antigen. This functional tolerance have largely been ascribed to immunoregulation.
inactivation occurs as a consequence of TCR engagement The initial problems in the identification of a suppressor
in the absence of full costimulatory signals. T cells receive T cell in the 1970s have been partially overcome more
costimulatory signals via ligation of surface CD28 with B7 recently with the discovery of a distinct CD4+ T cell subset
molecules expressed on APCs. Other costimulatory path- constitutively expressing CD25. Besides CD25, regulatory
ways include CD40/CD40 ligand (CD154). Furthermore, T cells (Treg) are characterized by the expression of the
signaling through CTLA-4 (CD152) has also been impli- transcription factor Foxp3, crucially involved in suppressing
cated in the generation of hyporesponsive T cells. Anergic overactive immune responses, as demonstrated by the
T cells fail to produce sufficient interleukin (IL)-2 and to high incidence of self-reactive lymphocytes in immune
proliferate in response to antigen, but the unresponsive dysregulation, polyendocrinopathy, enteropathy, X-linked
state can be overcome by the exogenous addition of IL-2 (IPEX) syndrome in humans and scurfy in mice. Further
Molecular basis of transplantation 379

markers to identify Treg cells include the expression of The immunological targets of GvHD
CTLA-4, glucocorticoid-induced tumor necrosis factor and GvL
receptor (GITR), folate receptor-4 (FR4), and the absence
We have previously discussed how differences between
of CD127. CD4+CD25+ naturally occurring Treg cells are
donor and recipient at the MHC level produce vigorous
thymic derived and have been shown to be important for
immune responses. Since genetic differences are the major
maintaining self-tolerance, regulating the homeostasis of the
factors influencing the outcome of allogeneic HSCT, the
peripheral T-cell pool, and contributing to tolerance induc-
selection of the donor is crucial, and where possible it
tion in various models of solid organ transplantation, as well
should be an MHC-identical sibling or an MHC-matched
as in allogeneic HSCT. In vivo depletion of Treg cells in ani-
unrelated donor. However, MHC matching is not sufficient
mal models increases the incidence of autoimmune diseases
for long-term graft survival and/or to prevent GvH reactions
and immune responses to tumors. It has also been observed
without the use of potent immunosuppressive regimens. The
that in a condition of full or mixed donor chimerism after
existence of additional histocompatibility loci, first indicated
HSCT, there is a selective advantage in the homeostatic
in inbred mice, was then clearly demonstrated in humans
expansion of Treg cells as compared to effector T cells. The
in HSCT, whereby they were associated with severe GvH. In
presence of donor antigens during this phase skews the Treg
this genetic situation, immune responses are directed against
repertoire toward the preferential expansion of those recog-
alloantigens encoded by histocompatibility (H) loci outside
nizing the donor antigens, thus contributing to the induction
the MHC, the so-called minor H loci. Minor H antigens
of transplantation tolerance. The use of conditioning regi-
are polymorphic self-derived peptides expressed on the cell
mens aimed at expanding Treg cells in vivo or the infusion of
surface in association with MHC class I and II molecules.
Treg cells expanded in vitro and co-transplanted with HSCs
The fact that they are recognized, as are viruses, by MHC-
have been proven efficacious in prolonging the survival of the
restricted T cells with specificity for peptides brought to
allograft.
the cell surface during biosynthesis of MHC class I and II
The mechanisms underlying naturally occurring Treg cell
molecules makes their identification more difficult. Unlike
immunosuppression require cell-to-cell contact. However,
in vitro T-cell responses to MHC antigens, which can be
adaptive CD4+CD25+ regulatory cells with a similar phe-
measured readily by proliferation in mixed lymphocyte reac-
notype have been described, they derive from mature effec-
tions (MLRs) without previous exposure to antigen, those
tor T cells and exert their suppressive activity via inhibitory
against minor H antigens need prior in vivo immunization.
cytokines such as TGF-β or IL-10. It is likely that a combi-
Although there is a large number of polymorphic pro-
nation of naturally occurring and adaptive regulatory cells
teins, not all give rise to peptides recognized as minor H
is involved in transplantation tolerance, depending on the
antigens. Furthermore, there are several factors that appear
time and the conditioning regimens. Although a proportion
in practice to severely limit the number of minor H antigens
of Treg cells specifically recognizes alloantigens, their over-
eliciting an in vivo response in a particular donor/recipient
all suppressive activity is not limited to cells expressing their
combination. In fact, it is clear that there is a hierarchy of
cognate antigens, thus implicating the possibility of unde-
responsiveness. In a genetic situation where there are many
sirable effects on virus- or tumor-specific immunity. There-
minor H disparities, one or a few are immunodominant,
fore, alloantigen-specific Treg cells are the most promising
with clones of CD8+ T cells responding to such an antigen
approach for their therapeutic application in the context of
expanding selectively, while T cells against others appear
transplantation.
transiently early in the response, or not at all. In most
models, immunodominance results from competition for
APC resources among responding CD8+ T cells, because it
Graft-versus-host and
disappears when competing epitopes are presented on dif-
graft-versus-Leukemia
ferent APCs or when APCs are present in large excess. There
is strong statistical and genetic evidence that this also occurs
A unique and prominent feature of allogeneic HSCT as com-
in human immune responses to multiple minor H antigens,
pared to solid organ transplantation is the presence, in the
thus in principle making it possible to predict, measure, and
graft, of immunologically competent cells which have the
manipulate the immune response following allografting.
ability to recognize normal and malignant recipient tissues.
As a consequence, donor lymphocytes mediate a reaction
Graft-versus-leukemia
against normal tissues which is defined as GvHD, but also
mediate the fundamental therapeutic effect at the basis of the Clinical studies and experimental animal models have
eradication of residual neoplastic cells, the GvL. It is not sur- shown that the efficacy of allogeneic HSCT in hematological
prising, therefore, that the molecular targets of GvH and GvL malignancies is related not only to the intensive chemo-
largely overlap. radiotherapy, but also to an immunological anti-tumor effect
380 Molecular Hematology

exerted by the graft itself. This effect, referred to as GvL, documented by studies on solid cancers, several mechanisms
has been initially recognized because patients who received implicated in the tumor evasion from immune surveillance
T-cell-depleted HSC preparations with the intention of can explain the resistance to graft-versus-tumor responses.
reducing GvHD had a much higher incidence of leukemia An important component for the induction of GvL
recurrence after the transplant. Further lines of evidence reactions is a fully functional antigen presentation. There
support this concept. For example, the risk of relapse is is emerging evidence that host dendritic cells (DCs) of
higher if donor and recipient are identical twins, and some leukemic origin may be harnessed in this process by induc-
reports have indicated that remission can be re-established ing the generation of anti-leukemic cytotoxic T cells in in
by the withdrawal of post-transplant immunosuppressive vitro and in vivo preclinical models. Early efforts to trans-
treatment and/or by the recurrence of GvHD. The proof late this approach into the clinical setting seem encourag-
of principle of the GvL effect came from the evidence that ing. The principle has been adopted by exploiting the abil-
the infusion of lymphocytes from the original stem cell ity of cytarabine and azacytidine to induce the maturation of
donor could restore complete remission in patients with leukemic blasts into DCs, and the consequent provision of
chronic myeloid leukemia (CML) relapsed after allogeneic antigenic and accessory stimulation of donor naı̈ve T cells.
SCT. These data support the requirement of two main There are reports of complete hematological remissions in
components for GvL to occur: the presence of T cells in the AML patients relapsed after HSCT by using a combination
donor preparation, and the existence of antigenic differences of low-dose cytarabine, G-CSF-mobilized donor blood cells
between donor and recipients. and GM-CSF post-transplant, or a combination of azacyti-
Although the GvL effect of donor lymphocyte infusions dine and DLI. The efficacy of such a strategy has been associ-
(DLIs) is extremely successful in CML (the response rate ated with the differentiation of central memory T cells and
is more than 90%), the experience in other malignancies the induction of a selective, GvHD-sparing, anti-leukemia
is not as good and, in some cases, rather disappointing. effect.
However, the introduction of reduced-intensity condition- Another cell subset involved in the elimination of residual
ing has extended the indications for allogeneic HSCT, and leukemic cells is NK cells. NK are fundamental players
thus promoted the use of DLI in several diseases. The rate of innate immunity. Their killing activity is tightly tuned
of response after reduced-intensity allograft in patients with by the balance of activating and inhibitory signals trans-
high-risk acute myeloid leukemia (AML) and myelodysplas- mitted by the interaction of different receptors on the
tic syndrome (MDS) has been reported to be comparable surface of NK cells, such as killer-cell immunoglobulin-like
with the results obtained using myeloablative regimens. In receptors (KIRs), NK Group 2 member D (NKG2D), or
these diseases, DLI can produce better outcomes if admin- NKG2A/CD94, and the corresponding ligands on the target
istered prophylactically or pre-emptively, guided by reduc- cell membrane. NK cells lyse target cells which lack MHC
tion of donor chimerism. Durable responses, which exceed class I molecules specific to the inhibitory receptors KIRs
the response durations seen both after conventional trans- (“missing-self hypothesis”). Therefore, NK cytotoxicity can
plantation and following previous chemotherapy cycles, were be particularly important in HLA-mismatched or haploiden-
reported in a small cohort of patients with Hodgkin lym- tical donor–recipient pairs. Importantly, since NK cells are
phoma and in low-grade non-Hodgkin lymphomas. Initial the first immune population to recover after HSCT, they are
enthusiasm with multiple myeloma has been subsequently also the first cell subset to recognize normal and malignant
disappointing, since it has become apparent that responses allogeneic targets. The involvement of “alloreactive” NK
can be achieved only in association with severe GvHD. It cells in GvL, suggested in preclinical models, has been
is possible that higher doses of donor lymphocyte are nec- corroborated in clinical studies, whereby the transplantation
essary, but then they would be incompatible with the side of haploidentical NK-“alloreactive” donors in high-risk
effects. AML patients has been shown to prevent disease relapse
Exploitation of the beneficial effect of allogeneic immune and improve survival. Accordingly, a slow NK recovery after
responses in non-hematological malignancies has indicated HSCT has been correlated to a higher risk of relapse.
that renal cell carcinoma (RCC) may be susceptible to the
GvH/tumor effect. However, no further evidence was pro-
Are tumor-specific T cells generated
vided, thus suggesting it had a minor impact.
after allografting?
The reason for the different sensitivity to DLI among var-
ious malignancies remains unclear. A possible explanation There is evidence that the immune system can mount a
for low susceptibility to DLI of acute lymphoblastic leukemia response against a tumor by recognizing antigens which
(ALL) has been attributed to the limited duration and mag- are either specific or associated to the tumor. Tumor-
nitude of leukemia-specific T-cell response generated in vivo, specific antigens (TSAs) include antigenic peptides gener-
which has been attributed to clonal exhaustion. As widely ated by genetic mutations specific to an individual cancer.
Molecular basis of transplantation 381

Tumor-associated antigens (TAAs) come from normal non- of diseases in terms of symptoms, organ involvement, and
polymorphic proteins overexpressed or aberrantly expressed pathological changes, there is not necessarily a temporal
in the tumor. correlation, with several cases presenting as overlapping
Almost all patients with CML harbor a specific molec- syndromes. The uncertainty in the clinical classification is
ular abnormality, the reciprocal chromosome translocation the result of the poor understanding of the pathogenesis of
t(9,22)(q34;q11), which juxtaposes BCR and ABL genes. the disease and the very limited access to its monitoring.
The fusion gene encodes for a chimeric protein, namely
p210BCR-ABL . Peptides derived from p210BCR-ABL can be pre-
Acute GvHD
sented by both MHC class I and class II molecules on CML
cells in vitro and p210BCR-ABL -specific CD8+ T cells have Acute GvHD is a potentially lethal disease which usually
been detected in the peripheral blood of CML patients. How- targets skin, liver, and the gastrointestinal tract. The typi-
ever, it is unclear whether these T cells have any purpose cal pathological finding in the skin is epithelial apoptotic
in the control of the leukemia. Early-phase clinical trials damage, with characteristic nuclear alterations and a small
using p210BCR-ABL vaccination have showed some encourag- inflammatory infiltrate. In the liver, the first injury involves
ing results. In patients with CML receiving a peptide-based vascular endothelial cells, and later the biliary epithelium is
vaccination, the development of bcr-abl peptide-specific destroyed as a result of portal lymphocytic infiltration. In the
immune responses was associated with clinical responses gastrointestinal tract there is destruction of intestinal crypts
and/or further reduction of residual disease. with acute or chronic inflammatory infiltrates and flattening
Other non-polymorphic proteins, such as proteinase-3 of the villous architecture. The clinical score is graded at four
(Pr3) and Wilm’s tumor 1 (WT1), have also been investigated levels, with grade 4 being very often lethal.
as targets for T cells in CML because of their high expression The cellular effectors of alloreactivity after HSCT are
levels in this leukemia. Pr3- or WT1-specific T cells at very cytotoxic T cells and NK cells, depending on the histo-
low frequency have been identified in patients with CML in incompatibility of the donor–recipient pair. Cytotoxic T cells
remission after allogeneic HSCT, but the findings have never are activated CD8+ T cells that recognize their specific anti-
been confirmed and their significance is not supported by gen bound to MHC class I molecules on the target cells. They
proper clinical trials. exert their functions through two main contact-dependent
Several other tumor-associated and TSAs, such as cytolytic pathways: the Fas/Fas-ligand (Fas-FasL, CD95-
preferentially expressed antigen in melanoma (PRAME), CD178)-mediated apoptosis and the release of cytotoxic
melanoma family antigen (MAGE), receptor for hyaluron- granules containing perforin/granzyme. Studies performed
mediated motility (RHAMM), New York esophageal in mice genetically deficient for these molecules showed that
squamous cell carcinoma-1 cancer-testis antigen (NY3ESO), a prominent but not exclusive role for the perforin/granzyme
or nucleophosmin (NPM1), are often expressed in leukemic pathway in GvHD-induced organ damage. In contrast, the
blasts. Cytotoxic T cells specific for the cognate antigen same pathway appears to be critical for the GvL effect, thus
have been isolated from the bone marrow of healthy donors suggesting the possibility of selectively impairing the alter-
as well as from non-transplanted AML, ALL, and CML native pathways to inhibit GvHD without compromising
patients, indicating that the alloreactivity post-transplant is GvL. In fact, during experimental and clinical GvHD, FasL
more prominent than leukemia-reactive immune responses. expression on donor T cells is increased and elevated serum
The presence of higher titers of PRAME-specific CD8+ levels of soluble FasL and Fas are correlated with severity of
T cells after HSCT has been correlated with a higher GvHD, and the inhibition of the Fas-FasL pathway markedly
probability of longer remission. However, most immune reduces GvHD of the liver in animal models.
reactivities against any of these antigens traced after DLI NK cells mediate direct cytotoxicity with the same
are weak, and the results so far obtained with the use of pathways of cytotoxic lymphocytes, albeit recognizing
TAA-specific T cells as immunotherapeutic agents have been different targets. Experimental studies have shown that NK
disappointing. cells are neither necessary nor sufficient to induce GvHD,
but in the setting wherein GvHD is already underway,
NK cells contribute to the overall outcome. The effect of
Graft-versus-host disease donor NK-mediated cytotoxicity in vivo, in the absence of
alloreactive T cells, is shown to be confined to recipient
GvHD is the main drawback of allogeneic SCT and is largely lympho-hematopoietic cells. Although the mechanism
responsible for non-relapse mortality. In relation to the responsible for such selectivity is unclear, it has been hypoth-
time of onset, GvHD can be classified as acute or chronic, esized that donor “alloreactive” NK cells could prevent
depending on whether it develops within 100 days from the GvHD manifestations by killing off host DCs, thus avoiding
transplant. However, although there are clearly two types alloantigen presentation to donor T cells.
382 Molecular Hematology

Chronic GvHD antibiotic associations active against anaerobic bacteria, such


as imipenem-cilastatin and piperacillin-tazobactam, seems
Chronic GvHD is a syndrome characterized by multiorgan
to correlate with higher GvHD grades and mortality. Con-
involvement and is clinically similar to an autoimmune dis-
versely, the use of antibiotics with less perturbative effects on
order. It usually involves skin, liver, gastrointestinal, and res-
the gut microbiota (such as rifaximin) was associated with
piratory mucosa, but several immunological functions are
lower incidence of gut GvHD and GvHD-related mortality.
impaired. The incidence of chronic GvHD ranges from 6%
The studies of microbiota composition point out that its
to 80%, mainly depending on the degree of disparity in the
role in GvHD development is not merely related to the
major histocompatibility antigen and the previous appear-
release of bacterial particles. In fact, microbiota seems to
ance of acute GvHD. Pathological findings are not typical
actively participate in the generation of a specific environ-
and are characterized by epithelial atrophy, increased hyaline
ment which can be pro- or anti-GvHD through the pro-
deposits, fibrosis, and chronic inflammatory infiltrates. His-
duction of metabolites. Short-chain fatty acids, for exam-
torically, chronic GvHD was classified as limited or extensive
ple, stimulate the extrathymic maturation of Treg cells and
on the basis of the results of a small retrospective study, but
one of these acids (butyric acid) can reduce the production
that has been shown neither to be reproducible nor predic-
of pro-inflammatory cytokines while inducing the produc-
tive of the clinical outcome, and a new scoring system is being
tion of indoleamine 2,3-dioxygenase (IDO), which can ame-
proposed.
liorate GvHD. Based on these findings, several approaches
have been tested in clinical trials with the aim of restor-
ing the diversity of the microbiota by the use of probiotics
The pathogenesis of GvHD or specific combinations of antibiotics for decontamination
of the gut at the HSCT. Recently, the use of fecal micro-
Acute GvHD and the “Cytokine Storm”
biota transplantation has also been investigated for the man-
Donor T cells recognizing major and/or minor H antigen dis- agement of steroid-resistant gut GvHD. Although the ratio-
parities on recipient tissues are certainly critical in the induc- nale for these approaches is very intriguing, clinical results
tion of acute GvHD. However, other factors need to be taken are still controversial and they have not yet entered clinical
into account. In particular, the incidence of acute GvHD has practice.
been reported to be much higher if allogeneic donor lym- The second step in the cytokine storm involves the
phocytes are administered in concomitance with the condi- activation of DCs and the presentation of alloantigens. The
tioning regimen than if they are infused a few months later inflammatory changes promoted in recipient tissues by con-
in the form of DLI. The reason for this is that the injuries ditioning regimens can initiate DC maturation and license
produced by the cytotoxic agents and the infections at time them to potently induce T-cell responses. While host APCs
of transplant activate the innate immune system and induce are necessary and sufficient for GvHD development, the role
pro-inflammatory changes in endothelial and epithelial cells. of donor APCs appears confined to the intensification of
This process, described as the “cytokine storm,” consists of ongoing disease. The differential role of donor and recipient
three main phases: (i) the destruction of natural barriers by APCs can be accounted for by the fact that endogenously
the conditioning regimens (first phase); (ii) the activation of synthesized minor H antigen-derived peptides are presented
DC and presentation of alloantigens (second phase); and (iii) to CD8+ T cells on host MHC class I (induction phase).
the recruitment and proliferation of activated effector cells Subsequently, the same antigens can be processed by donor
(third phase). APCs through the exogenous route of antigen processing and
In the first phase, the destruction of natural barriers by presented to the T cells, thus accounting for augmenting the
the conditioning regimens and the release of inflammatory GvHD initiated by the host APCs (maintenance phase). This
cytokines, like tumor necrosis factor (TNF)-α and IL-1, two-step process is similar to that described for the rejection
allows bacterial products to permeate in the tissues. The T of solid organ transplants (direct and indirect alloresponses).
cells exposed to bacterial products like lipopolysaccharide For its full development, GvHD finally requires cellular
exhibit enhanced migration and survival. In accord with this effectors to be recruited. Although activated donor T and
notion, the use of low-intensity preparatory regimens sub- NK cells effect the killing of host cells by contact-dependent
stantially reduces the incidence of acute GvHD. There is cytotoxicity, the release of inflammatory cytokines also plays
increasing evidence that lower diversity of the microbiota in an important role. In accordance, target organ injury can be
the gut at the time of the HSCT is associated with higher partially prevented by the neutralization of TNF-α and IL-1
incidence of GvHD rates and mortality. Studies in humans in animal models, and the blockade of TNF-α is of some
and mice indicate that active GvHD is associated with an efficacy in pilot clinical studies. The importance of multiple
imbalance in favor of specific bacterium phyla, with a pre- inflammatory effectors in GvHD suggests that more than
dominance of Enterococci. Furthermore, the use of specific one pro-inflammatory cytokine should be simultaneously
Molecular basis of transplantation 383

inhibited to effectively antagonize systemic GvHD. The contrast to acute GvHD, the development of the chronic form
case of IL-6 blockade can well explain the complexity is dependent on the APCs of donor rather than recipient ori-
of the matter. IL-6 is a cytokine usually associated with gin, thus justifying the possibility of a broader repertoire of
pro-inflammatory functions, but it also plays an important antigens and more widespread organ involvement.
role in controlling local and systemic inflammation. This However, one of the most remarkable features of chronic
dual activity depends on the fact that IL-6 can transmit GvHD is the clinical and pathological similarities with
signals through alternative pathways that can target different systemic autoimmune disease. Elevated levels of serum
subsets of CD4+ cells. While the small proportion of CD4+ autoantibodies (i.e. antinuclear, anti-dsDNA, anti-smooth-
cells expressing IL-6R are subject to a suppressive signal, muscle antibodies) in up to 70% of chronic GvHD patients
the vast majority respond to IL-6 linked to a soluble form of support the hypothesis that functionally relevant autoreac-
IL-6R present in the serum, and that drives an inflammatory tive T and B cells can be generated during chronic GvHD. It
response. Blockade of IL-6 signaling has been successfully has been suggested that antibodies to ubiquitously expressed
used in murine models of GvHD, but its efficacy in patients minor H antigens may be analogous to pathogenic autoan-
seems to be confined to its prophylactic use. tibodies in systemic autoimmune diseases, thus justifying
An obvious candidate to inhibit the expansion of allore- the similarities of tissue pathology. The importance of B-cell
active GvHD T cells is IL-2, but the efficacy of anti-CD25 responses in chronic GvHD was documented by clinical
antibodies has not been sufficiently supported by the clini- improvement following B-cell depletion with anti-CD20
cal experience. In contrast, the use of low-dose IL-2 has been monoclonal antibody.
demonstrated to effectively increase the number of Treg cells Studies in animal models have been carried out with
and to be effective at treating the chronic form of GvHD. The the aim of investigating the autoimmune manifestations of
therapeutic efficacy of IL-2 could also be enhanced by com- chronic GvHD. Using an HSCT model in which donor and
bination with rapamycin, which synergizes in increasing the recipient differed for multiple minor H antigens, it was pos-
proliferation and expansion of Treg cells. sible to isolate from mice developing chronic GvHD autore-
active CD4+ T cells with a specificity for an MHC class
II-encoded determinant common to donor and recipient.
Acute GvHD and T-Cell Trafficking
Moreover, donor CD4+ T cells isolated from recipients with
Although essentially all tissues express transplantation anti- chronic GvHD cause autoimmune disease if transferred to
gens, clinical manifestations of acute GvHD display remark- new syngeneic recipients. The development of donor T cells
able tissue tropism involving primarily gut, liver, and skin. that recognize antigens shared by donor and host has also
Recent studies have suggested that the skewed organ involve- been attributed to impaired mechanisms of deletion or reg-
ment is related to the homing properties of activated allo- ulation during chronic GvHD. It has been hypothesized that
geneic T cells and the presence of local tissue inflammation. autoreactive T cells may be the consequence of a damaged
Naı̈ve T cells injected in lethally irradiated allogeneic recipient thymus, unable to select the new T-cell repertoire
recipients are initially retained within secondary lymphoid following the pre-transplant therapy or acute GvHD. How-
tissues where, activated by recipient-derived APCs, they ever, it seems that also thymectomized animals can develop
undergo a rapid burst of proliferation, enter the peripheral GvHD.
circulation, and subsequently accumulate in the gut, liver, Not dissimilarly from the acute disease, cytokine dysreg-
and skin. However, local inflammation also appears crucial ulation has a causative role in chronic GvHD. High levels
in permitting the entry of activated T cells to target tissues, of IL-1β, IL-6, interferon (IFN)-γ, TNF-α, and low levels
because the infusion of alloreactive T cells produces GvHD of the inhibitory cytokine IL-10, are associated with more
in irradiated recipients, but not in recipients in which mixed severe forms. Multiple cytokines produced by activated T
hemopoietic chimerism has already been established, despite cells, such as INF-γ and TGF-β, were shown to promote
similar levels of alloreactive T cells in the periphery. To con- increased collagen deposition in preclinical models. Other
firm the role of local inflammation, GvHD can be induced soluble mediators, such as the CCL2 and CCL3 chemokines,
also in mixed chimeric mice if inflammatory stimuli are seem to modulate collagen turnover and deposition by
administered together with the donor T-cell transfer. sending signals to fibroblast via macrophages or indirectly
through stimulation of TGF-β.
Chronic GvHD

The pathogenesis of chronic GvHD largely remains to be elu- Can GvHD and GvL be dissected?
cidated. Mature donor alloreactive T cells infused with the Currently, the major aim in allogeneic HSCT is to identify
graft are supposed to play a key role in chronic GvHD. Sim- a way to the separate the beneficial GvL from GvHD (sum-
ilarly, APCs have been shown to be important although, in marized in Figure 27.3). Several attempts have been made
384 Molecular Hematology

1. Reduce 2. Deplete CD8*/


the dose Enrich CD4*

3. Remove alloreactive T cells


(suicide-inducible gene)
4. Induce anergy to alloantigens
(costimulation, Treg cells

5. Th1–Th2 shift Hemopoietic-restricted


minor Ags

6. Selection of
Leukemia-Associated
7. Block cytokines leukemia-specific
Ags
clones

TNFα, IFNγ
IL-2, IL-6 Cytotoxic pathways
JAK Inhibitors (FAS vs. perforin)

8. NK cells infusion

+ IL-15/IL-2
+ BiKEs
Leukemic APCs

9. DLI in association
with differentiating
factors
+ Cytarabine
+ Azacytidine
Leukemic blasts

DLI cells
Fig. 27.3 Strategies to separate GvHD from GvL. Ags, antigens; APCs, antigen-presenting cells; BiKEs, bispecific killer engagers; DLI, donor
lymphocyte infusion; IFN, interferon; IL, interleukin; JAK, Janus kinase; NK, natural killer; Th1, T helper type 1; Th2, T helper type 2; TNF, tumor
necrosis factor; Treg, T-regulatory cells.

during the last 15 years, either by selectively controlling Such a strategy allows donor T cells to be safely exploited for
GvHD or selectively inducing GvL. immune reconstitution and the GvL effect, with the option
of switching them off in case of GvHD. The positive experi-
ence with the gene encoding herpes simplex virus thymidine
Control of GvHD
kinase (HSV-TK) is being confirmed by the use of caspase 9
The simpler and more pragmatic approach is to escalate transgene. While the first relies on the administration of the
the dose of donor lymphocytes until a clinical response is antiviral drug ganciclovir to induce necrosis in transduced
achieved. This permits the incidence of GvHD to be reduced donor T cells, the second is based on the drug-induced acti-
to less than 20% of cases and has proved more useful than vation of the intrinsic apoptotic pathway. The results of new
depleting T-cell subsets like CD8+. Alternatively, the route of clinical trials are keenly awaited.
selectively depleting in vitro recipient-reactive donor T cells The possibility of interfering with the non-specific effects
before their infusion is valuable and its application has been of the cytokine storm with neutralizing monoclonal antibod-
successfully tested in haploidentical transplantation. ies is currently under scrutiny and the topic has been dis-
Alternative approaches to diminishing GvHD have been cussed before. Based on the observation that some of the
based on the use of donor T cells genetically engineered cytokines involved in GvHD development act through the
with suicide genes that can be activated in case of GvHD. STA3/JAK2, JAK1, or JAK3 pathways, it is not surprising that
Molecular basis of transplantation 385

Treg cells
MSCs
MDSCs

Fig. 27.4 Cell-based immunosuppression in


hemopoietic stem cell transplantation. GvHD, ↓ Tissue damage
graft-versus-host disease; GvL, graft-versus- ↓ GvHD severity
leukemia; MDSCs, myeloid-derived suppressor ↓ GvHD mortality
No effect on GvL
cells; MSCs, Mesenchymal stromal cells; Treg,
T-regulatory cells.

the availability of Janus kinase (JAK) inhibitors has paved the recent commercial study has claimed the final evidence in
way to testing these reagents in the management of GvHD. a large phase III clinical trial, but the results have not yet
The results are encouraging, but not yet sufficient to support been made publicly available. The heterogeneous clinical
their use on a vast scale. results are likely to result from the poor knowledge of the
Active immune regulation of donor/recipient reactive cells mechanisms underlying immunosuppression and the lack
is emerging as a key mechanism for inducing and maintain- of criteria to stratify patients for treatment with MSCs.
ing tolerance to alloantigens (summarized in Figure 27.4). Recent studies have shown that MSCs must undergo apop-
Treg cells are probably the main player in this process and tosis to be effective, and that this apoptosis is induced
a few studies in animal models have documented that their by the recipient cytotoxic cells responsible for GvHD.
adoptive transfer can also prevent and partially treat GvHD. This observation can provide a new perspective to stratify
There are also suggestions that Treg cells may preserve the patients for treatment and refine the use of this therapeutic
GvL activity, but data obtained in patients after HSCT have tool.
failed to show a significant correlation with GvHD and have Finally, preclinical studies have tested the efficacy of
found that leukemia relapses are associated with an incre- myeloid-derived suppressor cells (MDSCs), which are a
ment in the number of Treg cells in the peripheral blood of heterogenous myeloid population with immunosuppressive
patients. Similarly, the amount of Treg infiltration seems to activity. The adoptive transfer of MDSCs in animal models
correlate with a bad prognosis in ovarian cancer. of GvHD profoundly inhibits the development of GvHD.
Employing immunosuppressive networks represents a fur-
ther approach to controlling GvHD. Treg cells isolated from
Induction of Selective GvL
umbilical cord blood and expanded in vitro or freshly isolated
from adult peripheral blood have been used for GvHD pro- Cell dose and the time of administration are probably the
phylaxis in two different studies. In both cases, there was a most important factors affecting the efficacy and toxicity
reduction in the development of GvHD in comparison with of DLI. Clinical responses are largely dependent on disease
historical controls and the treatment was well tolerated. How- stage, but the cell dose plays an important role, because the
ever, Treg-treated patients exhibited a higher frequency of number of responders increases with increasing the cell dose
opportunistic infections than controls. and the effective cell dose correlates with disease stage.
Mesenchymal stromal cells (MSCs) have received center- Surprisingly, the incidence of GvHD is not affected by the
stage attention as potent modulators of immune responses, cell dose, but rather influenced by the time at which DLI is
especially because of the virtual absence of any side effects. administered. The longer the interval between the transplant
These cells exhibit a potent immunosuppressive activity in and DLI, the lower the incidence of acute and chronic GvHD.
vitro and in vivo, which targets virtually any cells of the Although these observations would strongly advise delaying
immune system. Their effect is not cognate dependent, thus the administration of DLI, the fast kinetics of disease relapse
not requiring MHC identity between MSCs and immune in diseases like AML and ALL make DLI completely ineffec-
cells. There is extensive experience with the use of third- tive if given at the time of full-blown relapse. Furthermore,
party MSCs infused into patients with steroid-resistant acute leukemia with high proliferative capacity are more prone to
GvHD. All studies have shown a significant improvement escape T-cell donor immune control. Leukemic clones which
of overall survival in responding patients, although no ran- had become resistant to donor lymphocytes after losing mis-
domized trial has yet formally proven their efficacy. A very matched HLA haplotypes by acquired uniparental disomy
386 Molecular Hematology

have been described in relapsed AML patients treated with and a low rate of GvHD. Responding patients exhibited a
repeated DLI infusions. significant downregulation of exhaustion-related genes in
Developing a DLI with a more selective antileukemia activ- their bone marrow CD8+ cells. These approaches could also
ity has been the holy grail of hematological immunotherapy. benefit from the association with drugs able to block the
It is generally true that minor H antigens are the targets of immunological checkpoints, thus further acting against the
both GvHD and GvL, thus in principle making the separa- immunosuppressive environment of the tumor.
tion of the two processes impossible. However, there are two An alternative is to exploit the phenomenon of immun-
strategies to pursue selectivity. One is based on the fact that odominance that we described before. T cells directed against
not all minor H antigens are equally expressed on the various a broadly expressed minor H antigen, but specific for the
tissues and some of them are preferentially, if not exclusively, immunodominant epitope, appear to be able to selectively
expressed on hemopoietic tissues. In the case of leukemia, for exert a powerful GvL without GvHD. The mechanisms for
example, the generation of T cells with a specificity against this tumor-specific effect largely remain unknown.
hemopoietic polymorphisms may limit the collateral dam- Long-term responses can be achieved with more aggres-
age by confining the GvH activity to that tissue and sparing sive strategies based on the use of pre-emptive or prophy-
the others. Further selectivity could be achieved by concen- lactic DLI in association with differentiating factors able to
trating on lineage-specific polymorphisms, as demonstrated induce the maturation of DCs from leukemic blasts and the
for B-cell malignancies (Figure 27.5). A promising approach stimulation of naı̈ve donor lymphocytes. Promising also is
could be the production of effector CD4+ T cells specific the combination of DLI with other therapeutic agents like
for minor H antigens presented in the context of HLA class lenalidomide and sorafenib, although the mechanistic basis
II molecules, which are mainly expressed on cells of the of their interaction is still poorly understood.
hematopoietic system. However, limitations to this approach The evidence that NK cells can produce a potent
are represented by the observation that non-hematopoietic antileukemic effect with minimal risk of GvHD makes such
precursors are induced to express HLA class II peptides in the an approach ideal. However, significant limitations are rep-
presence of inflammation. Furthermore, quiescent leukemic resented by the relatively small number of NK cells obtained
stem cells do not express HLA class II epitopes. with leukapheresis and the poor knowledge of how to main-
The selective infusion of “CD4-enriched DLI” would also tain NK therapeutic activity after their ex vivo expansion. The
induce the reversal of host antileukemic CD8+ cells with administration of recombinant IL-2 and IL-15 has been used
exhausted phenotype by “awakening” dormant endogenous in patients after NK infusions with a view to augmenting their
GvL effector cells. The use of CD4+-selected DLI has been number and activation in vivo.
associated with a high incidence of durable responses The use of bispecific killer engagers (BiKEs) has been
recently tested in clinic. BiKEs are small molecules made by
two antibody regions of different specificity, the first specific
Hemopoietic
GVHD GVL for a TAA/TSA and the second CD16a specific, thus cross-
failure
linking NK cells to a specific target cell. Such approaches have
been tested for the treatment of MDS and AML using a com-
Normal Normal
Leukemia bination of CD16a and CD33.
tissues hemopoietic tissues
Much research is still needed to improve the therapeutic
efficacy of these procedures and to further extend their use
in clinical practice. It is conceivable that in the near future
a combination of strategies will become the most successful
approach to maximizing the GvL effect after HSCT.

Conclusions
Alloreactive donor T cell
HSCT is a potent clinical methodology which, by leveraging
on both recipient and donor immune systems, gener-
Polymorphisms expressed on all normal and malignant tissues ates long-term immunological tolerance and antitumor
immunity. Despite the limitations and toxicities of the
Hemopoietic-restricted polymorphisms
procedure, the many successes of HSCT in malignant and
Leukemia-restricted polymorphisms non-malignant conditions have provided a unique platform
Fig. 27.5 Minor histocompatibility antigens in graft-versus-host for the development of new cellular therapies that are
disease (GvHD) and graft-versus-leukemia (GvL). currently being tested in other ailments.
Molecular basis of transplantation 387

Fozza, C. and Dazzi, F. (2012). Regulatory T cells in stem cell transplan-


Further reading tation: main characters or walk-on actors? Crit. Rev. Oncol. Hematol.
84 (1): 18–25.
Clinical use of HSCT Kitagawa, Y. and Sakaguchi, S. (2017). Molecular control of regulatory
T cell development and function. Curr. Opin. Immunol. 49: 64–70.
Jethava, Y.S., Sica, S., Savani, B. et al. (2017). Conditioning regimens
Marino, J., Babiker-Mohamed, M.H., Crosby-Bertorini, P. et al. (2016).
for allogeneic hematopoietic stem cell transplants in acute myeloid
Donor exosomes rather than passenger leukocytes initiate allore-
leukemia. Bone Marrow Transplant. 52 (11): 1504–1511.
active T cell responses after transplantation. Sci. Immunol. 1
Lindsay, J.O., Allez, M., Clark, M. et al., ASTIC trial group; European
(1):aaf8759.
Society for Blood and Marrow Transplantation Autoimmune Disease
Waldmann, H., Chen, T.C., Graca, L. et al. (2006). Regulatory T cells in
Working Party; European Crohn’s and Colitis Organisation (2017).
transplantation. Semin. Immunol. 18 (2): 111–119.
Autologous stem-cell transplantation in treatment-refractory Crohn’s
disease: an analysis of pooled data from the ASTIC trial. Lancet Gas-
troenterol. Hepatol. 2 (6): 399–406. GvHD and GvL
Muraro, P.A., Pasquini, M., Atkins, H.L. et al. (2017). Multiple Sclerosis–
Brunstein, C.G., Miller, J.S., McKenna, D.H. et al. (2016). Umbilical cord
Autologous Hematopoietic Stem Cell Transplantation (MS-AHSCT)
blood-derived T regulatory cells to prevent GVHD: kinetics, toxicity
long-term outcomes study group. JAMA Neurol. 74 (4): 459–469.
profile, and clinical effect. Blood 127 (8): 1044–1051.
Takami A. Hematopoietic stem cell transplantation for acute
Galleu, A., Riffo-Vasquez, Y., Trento, C. et al. (2017). Apoptosis
myeloid leukemia. Int. J. Hematol. 2018; 107(5): 513–518. doi:
in mesenchymal stromal cells induces in vivo recipient-mediated
https://doi.org/10.1007/s12185-018-2412-8
immunomodulation. Sci. Transl. Med. 9 (416). pii: eaam7828.
van Laar, J.M., Farge, D., Sont, J.K. et al., EBMT/EULAR Scleroderma
Martin, P.J., Rizzo, J.D., Wingard, J.R. et al. (2012). First- and second-line
Study Group (2014). Autologous hematopoietic stem cell transplan-
systemic treatment of acute graft-versus-host disease: recommenda-
tation vs intravenous pulse cyclophosphamide in diffuse cutaneous
tions of the American Society of Blood and Marrow Transplantation.
systemic sclerosis: a randomized clinical trial. JAMA 311 (24): 2490–
Biol. Blood Marrow Transplant. 18 (8): 1150–1163.
2498.
McLornan, D., Potter, V., and Dazzi, F. (2000). Donor lymphocyte infu-
sion: rationale, benefits, and limitations. In: Hematopoietic Cell Trans-
The hemopoietic niche plants: Concepts, Controversies and Future Directions (eds. H. Lazarus,
R. Gale, A. Keating, et al.), 223–231. Cambridge: Cambridge Univer-
Crane, G.M., Jeffery, E., and Morrison, S.J. (2017). Adult haematopoietic sity Press https://doi.org/10.1017/9781316335727.026.
stem cell niches. Nat. Rev. Immunol. 17 (9): 573–590. https://doi.org/ Schroeder MA, Choi J, Satser K, Di Persio JF 2018. The role of
10.1038/nri.2017.53. janus kinase signaling in graft-versus-host disease and graft versus
Korn, C. and Méndez-Ferrer, S. (2017). Myeloid malignancies and leukemia. Biol. Blood Marrow Transplant. 24(6): 1125–1134.
the microenvironment. Blood 129 (7): 811–822. https://doi.org/ Shlomchik, W.D. (2007). Graft-versus-host disease. Nat. Rev. Immunol.
10.1182/blood-2016-09-670224. 7 (5): 340–352.
Trento, C., Marigo, I., Pievani, A. et al. (2017). Bone marrow mesenchy- Simonetta, F., Alvarez, M., and Negrin, R.S. (2017). Natural killer cells
mal stromal cells induce nitric oxide synthase-dependent differenti- in graft-versus-host-disease after allogeneic hematopoietic cell trans-
ation of CD11b+ cells that expedite hematopoietic recovery. Haema- plantation. Front. Immunol. 8: 465.
tologica 102 (5): 818–825. https://doi.org/10.3324/haematol.2016. Staffas, A., Burgos da Silva, M., and van den Brink, M.R. (2017). The
intestinal microbiota in allogeneic hematopoietic cell transplant and
Transplantation tolerance graft-versus-host disease. Blood 129 (8): 927–933.
von Dalowski, F., Kramer, M., Wermke, M. et al. (2016). Mesenchy-
Chhabra, A.Y., Leventhal, J., Merchak, A.R., and Ildstad, S. (2017). mal stromal cells for treatment of acute steroid-refractory graft versus
HSCT-based approaches for tolerance induction in renal transplant. host disease: clinical responses and long-term outcome. Stem Cells 34
Transplantation 101 (11): 2682–2690. (2): 357–366.
Index

5q− syndrome 53 mixed-lineage leukemia gene 64 adhesion


11q23, acute myeloid leukemia 39 molecular methodologies 59–60 blood group antigens 291–293
pharmacogenomics 342–348 malaria and host cells 200–202
AAT see α1-antitrypsin Philadelphia chromosome 63–64 adhesion-related platelet disorders 253–256
AAV see adeno-associated virus Philadelphia-like 68 ADR see adverse drug reactions
Abelson gene (ABL) 63 relapsing 68 adult hemoglobin, structure 2
ABL see Abelson gene T cell 65–68 adverse drug reactions (ADR)
accelerated phase chronic myeloid leukemia acute myeloid leukemia (AML) single nucleotide polymorphisms 344
(CML-AP) 83 activated signaling 41–42 tyrosine kinase inhibitors 81
aceruloplasminemia 170 chromatin modification 45 AE1 see anion-exchanger protein 1
acquired bone marrow failure syndromes chromosomal abnormalities 39–40 African siderosis 170
131–137 cohesin complex 44 AGM see aorta-gonad-mesonephrous region
idiopathic aplastic anemia 131–134 core-binding factor 38 agonist receptor defects, platelets 257
inherited glycosylphosphatidylinositol core concepts 50 AKT 88–89
deficiency 136–137 cytogenic abnormalities 53–54 ALCL see anaplastic large cell lymphoma
paroxysmal nocturnal hemoglobinurua donor lymphocyte infusions 380 ALG see anti-lymphocyte globulin
134–136 driver gene mutations 40–41 ALK see anaplastic lymphoma kinases
activated factor V (FVa) 209–212 genetics 37–47 alkylating agent-related chromosomal
activated factor VII (FVIIa) 207–209 cytogenic abnormalities 37–40 aberrations 54
activated factor VIII (FVIIIa) 210 monitoring and therapeutics 45–46 allogeneic stem cell transplantation 373–387
activated factor IX (FIXa) 207 next generation sequencing 40–45 chronic myeloid leukemia 82–83
activated factor X (FXa) 207–210, 212 recurrent mutations 56–57 clonal deletion 377–378
activated protein C (APC) 210–213 International Prognostic Scoring System Diamond–Blackfan anemia 144
activated signaling Revised 54 dyskeratosis congenita 142
acute myeloid leukemia 41–42 KMT2A rearrangements 39 Fanconi anemia 140
myelodysplastic syndromes 56 methylation 44–45 graft-versus-host disease 377–386
ACTs see artemisinin-based combination recurrent mutations 56–57 graft-versus-leukemia 379, 383–384
therapies retinoic acid receptor 38–39 homing and mobilization 375
acute graft-versus-host disease 381–383 secondary 54 idiopathic aplastic anemia 133
acute lymphoblastic leukemia (ALL) 59–70 spliceosome 43–44 immune system reset 375–376
biological consequences 60 therapy-related chromosomal aberrations immunology 376–379
B-precursor 62–65 54 indications 373
chromosomal translocations 63–66 transcription factors 42–43 microenvironment 374–375
cooperating genetic events 60–62 tumor suppressors 43 non-immunological factors 374–375
cytogenic subgroups 60–61 acute promyelocytic leukemia 38–39 principles 373
development 62 adaptive immunity 300–303 sickling disorders 183–184
donor lymphocyte infusions 380 additional sex combs like-1 (ASXL1) space and competition 375
hyperdiploidy 64–65 45 tumor-specific antigens 380–381
hypodiploidy 65 adeno-associated virus (AAV) vectors 322, allorecognition mechanisms 376–377
minimal residual disease evaluation 330–331 allostery, hemoglobin 173–175
69–70 adenoviral vectors 322, 329–330 all-trans-retinoic acid (ATRA) 38–39

Molecular Hematology, 4th Edition. Edited by Drew Provan and John G. Gribben.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.

389
390 Index

α1-antitrypsin (AAT) deficiency 330–331 human studies 306–307 glycophorin C/D 290–291
α globins 2, 9–12 idiopathic aplastic anemia 131–133 Rh antigens 289–290
α-granule defects 258 infections 306, 313–314 BMF see bone marrow failure
α2β1 integrins 255 mouse models 305–306 BMPR see bone morphogenetic protein
alpha-ketoglutarate 55–56 non-infectious triggers 306–307 receptor
α0 thalassemia 9–10 pathophysiology 308 BMT see bone marrow transplantation
α+ thalassemia 10–11 T cell selection 301–303 bone marrow, multiple myeloma 127–128
α thalassemias 9–12, 15–16 thrombocytopenia 297, 305–306, 308–311 bone marrow failure (BMF) syndromes
AML see acute myeloid leukemia tissue damage 308 131–153
anaplastic large cell lymphoma (ALCL) 107 treatment 311–315 acquired 131–137
anaplastic lymphoma kinases (ALK) 107 autosomal dominant siderosis 170 anemia of chronic disease 156
anemia Diamond–Blackfan anemia 142–144
of chronic disease 155–159 BAFF see B cell activating factor dyskeratosis congenita 140–142
diagnosis 155–156 B cell activating factor (BAFF) 127 Fanconi anemia 138–140
pathogenesis 156–158 B cell precursor acute lymphoblastic leukemia glycosylphosphatidylinositol deficiency
treatment 158–159 (B-precursor ALL) 62–65 136–137
fetal 276 B-cell receptor (BCR) signaling pathway idiopathic aplastic anemia 131–134
malarial 202–204 115–116 inherited 138–144
sickle cell 175–187 BCL-2 103–104, 106, 108, 116 paroxysmal nocturnal hemoglobinurua
anemia of inflammation see anemia, of BCL11A 184 134–136
chronic disease Bcl-xL 89 red cell enzymopathies 144–151
anion-exchanger protein 1 (AE1) 285–289 BCR see B-cell receptor, breakpoint cluster bone marrow space 375
ankyrin 286 region bone marrow transplantation (BMT)
antenatal diagnosis of hemophilia 229–230 BCR-ABL1 51, 73–74 sickling disorders 183–184
anti-D/anti-K prophylaxis 276–277 Bernard–Soulier syndrome (BSS) 253–254 see also allogeneic stem cell transplantation
antigen-presenting cells (APCs) 297–298, β-catenin 366, 367 bone morphogenetic protein receptor (BMPR)
302–303 β globins 2, 7–9 166
anti-lymphocyte globulin (ALG) 131, 133 β thalassemias 7–9, 13–15 bosutinib 74–75, 81
antiplatelet antibodies 310 with hemoglobin E 187 2,3-BPG see 2,3-diphosphoglycerate
antithrombin (AT) 209 with sickle cell anemia 184 B-precursor ALL see B cell precursor acute
deficiencies 213–214 Beth Israel hemoglobin 189 lymphoblastic leukemia
pharmacogenomics 348–350 BL see Burkitt lymphoma BRC-ABL1 51
aorta-gonad-mesonephrous (AGM) region blastic phase chronic myeloid leukemia breakpoint cluster region (BCR) signaling
29 (CML-BP) 83 pathway 108
APC see activated protein C bleeding disorders see hemophilia, platelet brush border ferrireductases 162
APCs see antigen-presenting cells disorders, thrombophilia, von Bruton’s tyrosine kinase (BTK) 107, 108,
APECED see autoimmune polyglandular Willebrand disease 115–116
endocrinopathy with candidiasis and blood cell alloantigens 267–283 BSS see Bernard–Soulier syndrome
ectodermal dysplasia class I 271–272 BTK see Bruton’s tyrosine kinase
apical membrane antigen 1 (AMA-1) 199 class II genes 272 Burkitt lymphoma (BL) 103–105
apoptosis suppression 103–104 genetics 268–271 Bushwick hemoglobin 188
artemisinin-based combination therapies hemolytic disease of the fetus and newborn busulfan (1,4-dimethane-sulfonyl-oxybutane)
(ACTs) 193 273–277 74, 82
AT see antithrombin molecular typing 268–271
ATA boxes 4 neonatal alloimmune neutropenia C4b-binding protein (C4BP) 211, 215
ATR-16 syndrome 11 280–282 CAFCs see cobblestone area-forming cells
ATRA see all-trans-retinoic acid neonatal alloimmune thrombocytopenia CALM-AF10 see PICALM-MLLT 10
atransferrinemia 170 277–280 CALR 93–94
ATR-X syndrome 11–12 see also blood group antigens Campath-1H 311–312
autoimmune polyglandular endocrinopathy blood coagulation 207–211 cancer stem cells (CSCs) 363–372
with candidiasis and ectodermal protein C system 208–211 cell of origin 364
dysplasia (APECED) 304 regulation 209 dormancy 367
autoimmunity 297–317 see also thrombophilia epigenetics 368–369
benefits of 303–304 blood group antigens 285–296 microenvironment 367–368
epitope spread 307 adhesion molecules 291–293 pre-leukemic 364–365
genetics 304–305 anion-exchanger protein 1 285–289 surface antigens 365–366
hormonal influences 307 diversity of functions 292 targeted therapies 365–369
Index 391

CAP structures 5 chromatin modification frontline 75–78


carrier testing, hemophilia 228–229 acute myeloid leukemia 45 hydroxyurea 74, 82
CARs see chimeric antigen receptors see also epigenetics interferon alpha 81–82
CBF see core-binding factor chromosomal abnormalities monitoring 78–79
CBFB 38 acute myeloid leukemia 39–40 Philadelphia chromosome-negative 83
CBFβ–MYH11 fusion protein 38 chronic lymphocytic leukemia 113 secondary 79
CCAAT boxes 4 multiple myeloma 122–123 side effects 81
CCAAT/enhancer-binding protein α chromosomal translocations tyrosine kinase inhibitors 74–81
(CEBPA) 42–43, 57 acute lymphoblastic leukemia 60, 63–66 treatment failure 78–79
CD34+ cells 30 chronic myeloid leukemia 73–74 chronic neutrophilic leukemia (CNL) 95
CD44 291–292 myeloid malignancies 51–52 chronic refractory immune thrombocytopenia
CD123 see interleukin-3 alpha receptor Philadelphia chromosome 63–64 311, 313–314
CD177 281 chromosome 1, multiple myeloma 126 CIDRs see cysteine-rich interdomain regions
CDAE1 see N-terminal cytoplasmic domain chromosome 7, acute myeloid leukemia circumsporozoite protein (CSP) 195
of AE 1 39–40 c-kit 25, 38, 42
CDKIs see cyclin-dependent kinase chromosome 9, Philadelphia chromosome classical Hodgkin lymphoma (cHL) 108
inhibitors 63–64 classifications
CDKN2A 66 chromosome 13, multiple myeloma 125 multiple myeloma 124
CEBPA see CCAAT/enhancer-binding chromosome 17, multiple myeloma 126 von Willebrand disease 236–238
protein α chromosome 22 clinical factors
cell-cycle-active drugs 31 B-precursor ALL 63–64 chronic myeloid leukemia 71–72
cell cycle control chronic myeloid leukemia 51 dyskeratosis congenita 140
hematopoiesis 24 see also Philadelphia chromosome Fanconi anemia 138, 140
lymphomas 106 chronic eosinophilic leukemia 95 glucose-6-phosphate dehydrogenase
multiple myeloma 125 chronic graft-versus-host disease 382, 383 deficiency 148
cell-extrinsic regulators, hematopoiesis chronic lymphocytic leukemia (CLL) hemophilia 221–223
24–29 B-cell receptor signaling 115–116 idiopathic aplastic anemia 133
cell-intrinsic regulators, hematopoiesis cell of origin 112–113 immune thrombocytopenia 310
24 chromosomal alterations 113 JAK2V617F myeloproliferative neoplasms
cell of origin (COO) environmental factors 112 89–91
cancer stem cells 364 epidemiology 111–112 neonatal alloimmune thrombocytopenia
chronic lymphocytic leukemia 112–113 epigenetics 115 279–280
cellular compartment model 21–23 genetics 107–108 paroxysmal nocturnal hemoglobinurua
central dogma 356 impaired immunity 116–117 136
central tolerance 302–303, 377–378 microenvironment 116 red cell enzymopathies 144
ceruloplasmin 170 micro RNA alterations 114–115 sickle cell anemia 177–180
CFCs see colony-forming cells molecular biology 111–120 von Willebrand disease 236–238
CFU-S see colony-forming units, spleen prognostic factors 117–118 see also diagnosis
CGH see comparative genomic Richter transformation 118–119 clinical features
hybridization somatic mutations 113–114 chronic myeloid leukemia 71–72
Chédiak–Higashi syndrome 259 stereotype receptors 113 JAK2V617F myeloproliferative neoplasms
chemokine receptors, hematopoiesis 26–29, chronic myeloid leukemia (CML) 71–85 89–91
30 BCR-ABL1 73–74 CLL see chronic lymphocytic leukemia
childhood acute lymphoblastic leukemia chromosome 22 51 clonal deletion 302–303, 377–378
342–348 clinical presentation 71–72 clonal hematopoiesis 49, 133–134
Chile hemoglobin 190 donor lymphocyte infusions 380 clonal hematopoiesis of indeterminate
chimeric antigen receptors (CARs) epidemiology 71 potential (CHIP) 49
331–334 prognostic models 73 clopidogrel, pharmacogenomics 350
CHIP see clonal hematopoiesis of SRC family of kinases 74 CLP see common lymphoid progenitor
indeterminate potential therapy 74–83 clustered regularly interspersed short
ChIP assays see chromatin accelerated/blastic phases 83 palindromic repeats (CRISPR)-Cas9
immunoprecipitation assays allogeneic stem cell transplantation system 103, 184, 320
cHL see classical Hodgkin lymphoma 82–83 CML-AP see accelerated phase chronic
chorionic villus sampling 229 busulfan 74, 82 myeloid leukemia
Christmas disease 226 discontinuation 77 CML-BP see blastic phase chronic myeloid
chromatin immunoprecipitation (ChIP) drug dose schedules 79–81 leukemia
assays 103 endpoints 77–78 c-MYC 66–68, 103–105, 125–127
392 Index

CNL see chronic neutrophilic leukemia CYP2C9 genetic analysis techniques 102–103
CNV see copy number variants clopidogrel efficacy 350 glucose-6-phosphate dehydrogenase
coagulation 207–211 warfarin efficacy 349 deficiency 148, 149
protein C system 208–211 CYP2D6 and codeine 341 hemophilia 229–230
regulation 209 cysteine-rich interdomain regions (CIDRs) immune thrombocytopenia 310
see also hemophilia, platelet disorders, 201 multiple myeloma 124–125
thrombophilia, von Willebrand disease cytarabine 367, 369 myelodysplastic syndromes 52–53
cobblestone area-forming cells (CAFCs) cytogenic abnormalities myeloid malignancies 50–51
22 acute lymphoblastic leukemia 60–61 myeloproliferative neoplasms 95–96
codeine, pharmacogenomics 341 acute myeloid leukemias 37–40 neonatal alloimmune neutropenia 282
codons 5, 356 myelodysplastic syndromes 52–53 neonatal alloimmune thrombocytopenia
cohesin complex cytokine receptor-like factor 2 (CRLF2) 279–280
acute myeloid leukemia 44 deregulation 65 pharmacogenomics 339–352
myelodysplastic syndromes 56 cytokines platelet disorders 252–253
collagen receptors, platelet disorders anemia of chronic disease 158 pyruvate kinase deficiency 147
255–256 autoimmunity 305 red cell enzymopathies 144
colony-forming cells (CFCs) 22–23, 30 graft-versus-host disease 382–385 von Willebrand disease 238
colony-forming units, spleen (CFU-S) 22 hematopoiesis regulation 24–25 see also clinical factors
colony-stimulating factor 3 receptor (CSF3R) immunity 300–301 Diamond–Blackfan anemia (DBA)
mutations 95 JAK2 signaling 87–89 142–144
commitment, hematopoiesis 24 MPL mutations 92 Dicer 1 367
common lymphoid progenitor (CLP) cytoskeletal platelet disorders 260 differentiation 21
populations 23 diffuse large B-cell lymphoma (DLBCL) 104,
comparative genomic hybridization (CGH) dasatinib 74–77, 79–81 105, 118–119
102 side effects 81 1,4-dimethane-sulfonyl-oxybutane see
competition, stem cell transplantation DBA see Diamond–Blackfan anemia busulfan
375 DBP see Duffy binding proteins 2,3-diphosphoglycerate (2,3-BPG) 3
complement system 135, 298–299 DC see dyskeratosis congenita discontinuation, chronic myeloid leukemia
congenital cyanosis 17 DCYTB see duodenal cytochrome b treatment 77
conventional metaphase cytogenic analysis 3-deazaneplanocin A (DZNep) 369 divalent metal transporter 1 (DMT1) 162
50–51 deficiencies DKC1 140–142
COO see cell of origin α1-antitrypsin 330–331 DLBCL see diffuse large B-cell lymphoma
copy number variants (CNV) and antithrombin 213–214 DLIs see donor lymphocyte infusions
pharmacogenomics 341 glucose-6-phosphate dehydrogenase 345, DMT1 see divalent metal transporter 1
core-binding factor (CBF) 38 364 DNA
costimulatory blockade 313 protein C 214–215 discovery of 354–357
coumarins, pharmacogenomics 348–350 protein S 215 see also epigenetics, genetics, transcription
counseling DEK-NUP214 39 DNA methyltransferase 3A (DNMT3A) 44,
hemophilia 228–229 deletions, α thalassemias 10–11, 12 55, 57, 94–95
neonatal alloimmune thrombocytopenia δ-granule defects 258 DNA sequencing 103, 360–362. See also
280 δβ thalassemias 12 next-generation sequencing
CRISPR see clustered regularly interspersed dense δ-granule defects 258 DNMT3A see DNA methyltransferase 3A
short palindromic repeats deoxy(T) hemoglobin 2 donor lymphocyte infusions (DLIs) 380,
crizanlizumab 183 desmopressin 244–245 385–386
CRLF2 see cytokine receptor-like factor 2 deterministic theory of hematopoiesis 23 dormancy, leukemic stem cells 367
cryptic splice sites 8 development Dot1l 369
CSCs see cancer stem cells acute lymphoblastic leukemia 62 double-hit lymphomas 104–106
CSF3R see colony-stimulating factor 3 globin expression 3, 6 driver gene mutations 40–41
receptor hematopoietic stem cells 22, 29 drug dose schedules
CSP see circumsporozoite protein multiple myeloma 125–126 chronic myeloid leukemia 79–81
CXCR4, hematopoiesis 30 stem cells 22 see also management, treatment
cyanosis, congenital 17 diagnosis Duffy antigen 196–197
cyclin D1 106, 123–124 anemia of chronic disease 155–156 Duffy binding proteins (DBP) 197–198
cyclin-dependent kinase inhibitors (CDKIs) autoimmunity 310 duodenal cytochrome b (DCYTB) 162
24 Diamond–Blackfan anemia 143 dyskeratosis congenita (DC) 140–142
cyclin-dependent kinases (CDKs) 106 dyskeratosis congenita 142 dyskerin 140
cyclophosphamide, hematopoiesis 31 Fanconi anemia 140 DZNep see 3-deazaneplanocin A
Index 393

eADA see erythrocyte adenosine deaminase erythropoiesis FGFR3 125


early T-cell precursor acute lymphoblastic iron restriction 156–157 fibronectin 30–31
leukemia (ETP ALL) 67 malaria 204–205 FISH see fluorescent in situ hybridization
EBA ligands 197–198 erythropoietin (EPOR) FLT3-ITD 39, 41–42, 57
EBP see erythrocyte-binding proteins JAK2 signaling 87–89 Flt3 receptor
EBV see Epstein–Barr virus resistance and inflammation 158 acute myeloid leukemia 41–42, 57
eculizumab 136 E-selectins, hematopoiesis 30 hematopoiesis 25
egress of hematopoietic stem cells 30–31 essential thrombocythemia (ET) 87–96 fluorescence-activated cell sorting (FACS) 32
electrophoresis 359 CALR mutations 93–94 fluorescent in-situ hybridization (FISH) 102
eltrombopag 133, 314 JAK2 mutations, other 91–92 follicular lymphoma, genetics 106
Embden–Meyerhof pathway 147 JAK2V617F mutation 89–91 frequencies of hemoglobin variants 17–18
endoplasmic reticulum (ER), CALR mutations MPL mutations 92 frontline therapy
93–94 ESXL1 94–95 chronic myeloid leukemia 75–78
endosomes, iron transport 163 ethanol precipitation 358 malaria 193
endothelial protein C receptor (EPCR) 209 ethnic groups, pharmacogenomics 342 functional analysis, hematopoietic stem cells
engraftment 29, 29–30 ETP ALL see early T-cell precursor acute 33–34
environmental factors lymphoblastic leukemia functions of hemoglobin 2–3
autoimmunity 306 ETV6 65 FVa see activated factor V
chronic lymphocytic leukemia 112 expression FVII see factor VII
EPCR see endothelial protein C receptor globins FVIIa see activated factor VII
epidemiology developmental changes 6 FVIII see factor VIII
autoimmunity 306–307 gene action 4–5 FVIIIa see activated factor VIII
chronic lymphocytic leukemia 111–112 genetic control 3–4 FX see factor X
chronic myeloid leukemia 71 regulation 5–6 FXa see activated factor X
FV Leiden 212 expression cloning 358
hemoglobin variants 17–18, 187 extracellular-signal-regulated kinase (ERK) G6PD see glucose-6-phosphate
malaria 193–195 pathway 88–89, 108 dehydrogenase
neonatal alloimmune thrombocytopenia ex-vivo expansion 32–33 gain of function mutations, prothrombin
279 EZH2 55, 94–95, 369 215–217
sickling disorders 176, 184–186 γ chains, hemoglobin 2–3
epigenetics 2, 357–358 F1P1L1-PDGFRA fusion gene 95 ganciclovir 384
acute myeloid leukemias 44–45 FACS see fluorescence-activated cell sorting GATA2 39
cancer stem cells 368–369 factor V (FV) 209–212 GBT440 183
chronic lymphocytic leukemia 115 Leiden 211–215, 217 G-CSF see granulocyte colony-stimulating
Fanconi anemia 139 factor VII (FVII) 207–209 factor
myelodysplastic syndromes 55–56 factor VIII (FVIII) 210 gemtuzumab ozogamicin (GO) 366
myeloproliferative neoplasms 94–95 hemophilia 223–225 gene action, hemoglobin 4–5
pharmacogenomics 342 inhibitory antibodies 226–228 gene expression profiling (GEP) 102
epitope spread, autoimmunity 307 von Willebrand disease 236, 238, 243–245 gene therapy 319–337
EPOR see erythropoietin factor IX (FIX) 207, 209, 225–226 cancer 331–335
Epstein–Barr virus (EBV) 105 gene therapy 330 factor IX 330
ERK see extracellular-signal-regulated inhibitory antibodies 226–228 hemophilia 231–232
kinase factor X (FX) 207–210, 212 lysosomal storage diseases 319, 327
erythrocyte adenosine deaminase (eADA) factor XI (FXI) 207–209 severe combined immunodeficiency
142 factor FXII (FXII) 207 326–327
erythrocyte-binding proteins (EBP) Fanconi anemia 138–140 suicide induction 334–335
197–198, 200–201 FBXW7 66 vectors 321–331
erythrocyte lifespan, inflammation effects FCGR3B 281 adeno-associated 322, 330–331
158 Fcγ receptors 305 adenoviral 322, 329–330
erythrocytes ferritin 155–156, 164 lentiviral 322–323, 327–329
adhesion molecules 291–293 ferroportin 162–165 oncoretroviral 321–327
anion-exchanger protein 1 285–289 hemochromatosis 169 genetic counseling, hemophilia 228–229
antigens 267–296 siderosis 170 genetic immunotherapy 331–335
glycophorin C/D 290–291 fetal anemia 276 genetics
malarial invasion 196–200 fetal hemoglobin 2, 3, 6–7 acute lymphoblastic leukemia 60–62
Rh antigens 289–290 fetal tests for hemophilia 229–230 acute myeloid leukemias 37–47
see also blood, hemoglobin, red blood cells FGFR1 95 analytical techniques 102–103
394 Index

genetics (Continued) GM-CSF receptor see granulocyte homing 29–30, 375


anaplastic large cell lymphoma 107 macrophage colony-stimulating factor isolation 31–34
autoimmunity 304–305 receptor lentiviral vectors 322–323, 327–329
Burkitt lymphoma 103–105 GO see gemtuzumab ozogamicin lineage commitment 23–24
chronic lymphocytic leukemia 107–108, Gower hemoglobin 2 microenvironment 29
111–112 GPC/GPD see glycophorin C/D mobilization 31, 375
Diamond–Blackfan anemia 142–143 GPCRs see G-protein-coupled receptors oncoretroviral vectors 321–327
diffuse large B-cell lymphoma 104, 105 GPI see glycosylphosphatidylinositol primitive cell trafficking 29–31
double-hit lymphomas 104–106 G-protein-coupled receptors (GPCRs) regulation 24–29
dyskeratosis congenita 140–142 26–29 sources 22
Fanconi anemia 139 GPVI 255–256 hematopoietic stem cell transplantation
follicular lymphoma 106 graft-versus-host disease (GvHD) 377–386 (HSCT) 373–387
glucose-6-phosphate dehydrogenase acute 381–383 clonal deletion 377–378
deficiency 148 chronic 382, 383 Diamond–Blackfan anemia 144
hemophilia 223 control 384–385 dyskeratosis congenita 142
HFE-associated hemochromatosis pathogenesis 382–383 Fanconi anemia 140
167–169 separation from graft-versus-leukemia graft-versus-host disease 377–386
HLA genes 268–271 383–386 graft-versus-leukemia 379, 383–386
Hodgkin lymphoma 108 graft-versus-leukemia (GvL) 379, 383–386 homing and mobilization 375
lymphomas 101–110 selective induction 385–386 idiopathic aplastic anemia 133
lymphoplasmacytic lymphoma 104, granulocyte colony-stimulating factor immune system reset 375–376
106–107 (G-CSF) 23–24, 31, 367, 374–375 immunology 376–379
mantle cell lymphoma 106 granulocyte macrophage colony-stimulating indications 373
mucosa-associated lymphoid tissue factor (GM-CSF) receptor 23, 25, microenvironment 374–375
lymphomas 107 374–375 non-immunological factors 374–375
neonatal alloimmune neutropenia Gray platelet syndrome 258–259 peripheral tolerance 378–379
281–282 Griscelli syndrome 259 principles 373
neonatal alloimmune thrombocytopenia GT see Glanzmann thrombasthenia space and competition 375
280 GvHD see graft-versus-host disease tumor-specific antigens 380–381
pyruvate kinase deficiency 146 GvL see graft-versus-leukemia hematopoietic stem/progenitor cells (HSPCs)
Rh antigens 273–276 GWAS see genome-wide association studies 29
sickle cell anemia 175–177 hemochromatosis 167–170
thrombophilia 211–217 Hammersmith hemoglobin 188 ferroportin-associated 169
type 1 diabetes 304 haplotypes and pharmacogenomics 341 HFE-associated 167–169
von Willebrand disease 238–244 HApMAp consortium 341 juvenile 169
von Willebrand factor 236 Hb see hemoglobin TFR2-associated 169
see also pharmacogenomics HDFN see hemolytic disease of the fetus and hemoglobin (Hb) 1–19
genome annotation 1 newborn allostery 173–175
genome-wide association studies (GWAS) 6 Heinz bodies 188 α thalassemias 9–12, 15–16
JAK2V617F myeloproliferative neoplasms Helicobacter pylori eradication 313–314 β thalassemias 7–9, 13–15
90–91 hematopoiesis 21–31 developmental changes 6
genomic microarrays 51 cellular compartment model 21–22 δβ thalassemias 12
genotype-phenotype relationships, lineage commitment 23–24 gene action 4–5, 12–16
thalassemias 12–16 microenvironmental niche 29 Gower 2
geography of malaria 194 regulation 24–29 high oxygen affinity 188–189
GEP see gene expression profiling trafficking 29–31 Lepores 12
giant platelet disorders 260 hematopoietic-specific factors 6 nitric oxide 278–279
Glanzmann thrombasthenia (GT) 256–257 hematopoietic stem cells (HSCs) 21–35 oxygen dissociation curves 2–3
globins adeno-associated virus vectors 330–331 pathologies 6–18
genetic control 3–4 adenoviral vectors 329–330 Portland 2
structural variants 6–7, 16–18 cell-extrinsic regulators 24–29 prevention & treatments 18
synthesis 4–6 cell-intrinsic regulators 24 regulation 3–4, 5–6
glucose-6-phosphate dehydrogenase (G6PD) cellular compartment model 21–23 structural variants 6–7, 16–18, 173–191
deficiency 148–150, 345, 364 egress 30–31 structure and function 2–3, 173–175
glycophorin A 197 engraftment 29–30 synthesis 4–6
glycophorin C/D (GPC/GPD) 290–291 functional analysis 33–34 tetramers 2–3
glycosylphosphatidylinositol (GPI) 134–137 gene therapy 319–334 unstable 9, 17, 188
Index 395

hemoglobinopathies 173–191 Hodgkin lymphoma (HL) myelodysplastic syndromes 55–56


with β thalassemias 184, 187 genetics 108 myeloproliferative neoplasms 94–95
hemoglobin C 185 Richter transformation 118–119 idiopathic aplastic anemia (IAA) 131–134
hemoglobin E 187 Hoechst 33342 31–32 IFN-γ see interferon-γ
hemoglobin SC 185–186 homing, hematopoietic stem cells 29–30, IGD see inherited
high oxygen affinity 188–189 375 glycosylphosphatidylinositol
low oxygen affinity 189 host cell adhesion, malaria 200–202 deficiency
M hemoglobins 189–190 host ligands, malarial invasion 196–197 IgH see immunoglobulin heavy chain
sickle cell disease 16–17, 175–187 Hoyerall–Hreidarsson syndrome 140 IL see interleukin
sickle/β thalassemia 184 HPA see human platelet antigen imatinib 63–64, 74–77, 79–81
unstable 9, 17, 188 HPFH see hereditary persistence of fetal side effects 81
hemojuvelin 169 hemoglobin immune thrombocytopenia (ITP) 297,
hemolytic disease of the fetus and newborn HRD tumors see hyperdiploidy 305–306, 308–311
(HDFN) 273–277 HSCs see hematopoietic stem cells immunity
frequency 276 HSCT see hematopoietic stem cell adaptive 300–303
Kell system 276 transplantation clonal deletion 377–378
Rh antigens 273–276 HSPCs see hematopoietic stem/progenitor complement system 298–299
treatment and prevention 277 cells innate 297–299
hemophilia 221–234 HSV-TK see herpes simplex virus thymidine peripheral tolerance 302, 378–379
antenatal diagnosis 229–230 kinase resetting 375–376
A-type 223–225 Human Genome Project 1, 357 see also autoimmunity
B-type 225–226 human immunodeficiency virus (HIV) 323, immunoglobulin heavy chain (IgH)
carrier testing 228–229 327–329 transcription elements 104
clinical features 221–223 human leukocyte antigen (HLA) genes immunoglobulin heavy chain (IgH)
genetics 223 268–273 translocations, multiple myeloma
inhibitory antibodies 226–228 class I 271–272 122, 123
treatment 230–232 class II 272 immunoglobulins
heparin 209, 218 identification 268–271 hemophilia 226–228
hepcidin 162, 164–166 stem cell transplantation 376–378 neonatal alloimmune neutropenia 282
hephaestin 162 human neutrophil antigens (HNAs) immunoglobulin translocations, multiple
hereditary factors see genetics 280–283 myeloma 122
hereditary persistence of fetal hemoglobin human platelet antigen (HPA) 267, 277–280 immunology
(HPFH) 6–7, 12 human studies, autoimmunity 306–307 graft-versus-host disease 379
Hermansky–Pudlak syndrome 259 hydroxycarbamide (hydroxyurea) 74, 82, stem cell transplantation 376–379
herpes simplex virus thymidine kinase 183 impaired immunity, chronic lymphocytic
(HSV-TK) 384 2-hydroxyglutarate (2-HG) 55–56 leukemia 116–117
herpes virus vectors 322 5-hydroxymethylfurfural (5-HMF) 183 Indian blood group antigens 291–292
HFE 166–169 hyperdiploidy induced pluripotent stem cells (iPSCs) 21,
HFE-associated hemochromatosis acute lymphoblastic leukemia 64–65 142
167–169 multiple myeloma 122–123 induction, graft-versus-leukemia 385–386
2-HG see 2-hydroxyglutarate hypnozoites 196 infections and autoimmunity 306, 313–314
high-grade B-cell lymphomas with MYC and hypodiploidy, acute lymphoblastic leukemia inflammation
BCL2 or BCL6 translocations 65 anemia of chronic disease 155–159
104–106 hypoferremia, anemia of chronic disease 156 erythrocyte lifespan 158
high hyperdiploidy, acute lymphoblastic hypomethylation, acute myeloid leukemia erythropoietin resistance 158
leukemia 64–65 44–45 iron absorption and release 158, 164–166
high oxygen affinity hemoglobins hypoxia inherited bone marrow failure syndromes
188–189 cancer stem cells 369 138–144
high-throughput RNA sequencing (RNA-Seq) iron homeostasis 165 Diamond–Blackfan anemia 142–144
103 dyskeratosis congenita 140–142
histone arginine methyltransferases 89 IAA see idiopathic aplastic anemia Fanconi anemia 138–140
histone modifications ibrutinib 108, 116 inherited glycosylphosphatidylinositol
cancer stem cells 368–369 ICAM see intercellular adhesion molecules deficiency (IGD) 136–137
myelodysplastic syndromes 55 ICLs see intrastrand DNA cross-links inhibitory antibodies, hemophilia 226–228
HL see Hodgkin lymphoma idelalisib 108 initiation codons 5
HLA see human leukocyte antigen IDH1/2 44 innate immunity 297–299
HNAs see human neutrophil antigens acute myeloid leukemia 57 instructive model of hematopoiesis 23
396 Index

integrins JAK2 mutations genetics 101–110


hematopoiesis 30–31 cytokine signaling 87–89 common 103–104
platelet disorders 255 myeloproliferative neoplasms 87–92 techniques 102–103
intercellular adhesion molecules (ICAM) other 91–92 see also individual lymphoma types
201 V617F 87–91 lymphoplasmacytic lymphoma (LPL) 104,
interferon alpha, chronic myeloid leukemia JAK homology domains (JH) 88–89 106–107
81–82 Janus kinase (JAK) inhibitors 385 lysosomal storage diseases (LSDs) 319,
interferon-β (IFN-β), anemia of chronic JH see JAK homology domains 327
diease 158 juvenile hemochromatosis 169
interferon-γ (IFN-γ), anemia of chronic macrophage colony-stimulating factor
diease 158 Kansas hemoglobin 189 (M-CSF) 23, 25
interferon-γ (IFN-γ) receptors, hematopoiesis karyotypes macrophages, iron transport 163–164
25 acute lymphoblastic leukemia 60–61 major histocompatibility complex (MHC)
interleukin-3 alpha receptor (CD123) hemophilia 223 300, 304–305, 376
365–366 International Prognostic Scoring System malaria 193–206
interleukin (IL) receptors 25, 158, 365–366 Revised 54 anemia 202–204
internal tandem duplications (ITDs) 39, multiple myeloma 122–123 epidemiology 193–195
41–42, 57 myeloid malignancies 52–54 erythropoiesis 204–205
International Prognostic Scoring System Kell system 276 host cell adhesion 200–202
Revised (IPSS-R) 54 KMT2A 39, 45, 64 invasion 196–200
intervening sequences (IVSs) 3–4 Köln hemoglobin 188 life cycle 195–196
intestinal iron transport 162 K-RAS 125 malaria resistance 17–18
intracellular signaling, platelet disorders MALT lymphomas see mucosa-associated
257–258 laminins 292 lymphoid tissue lymphomas
intrastrand DNA cross-links (ICLs) 138–139 LCA see Leber congenital amaurosis management
introns see intervening sequences LCR see locus control regions anemia of chronic disease 158–159
in vitro assays, hematopoietic stem cells 33 Leber congenital amaurosis (LCA) glucose-6-phosphate dehydrogenase
in vitro colony-forming cells (CFCs) 22–23. 330 deficiency 149–150
See also colony-forming cells Leiden Factor V allele 211–215, 217 graft-versus-host disease 384–385
in vivo assays, hematopoietic stem cells lentiglobin vectors, sickle cell disease hemoglobin disorders 18
33–34 184 hemophilia 230–231
iPCS see induced pluripotent stem cells lentiviral vectors 322–323, 327–329 pyruvate kinase deficiency 147–148
IPSS-R see International Prognostic Scoring Lepores hemoglobin 12 red cell enzymopathies 146
System Revised leukemia inhibitory factor (LIF) 25 thrombophilia 217–218
iron chelation 143–144 leukemic stem cells (LSC) 364–369 see also treatment
iron metabolism 161–171 dormancy 367 mantle cell lymphoma 106
disorders 166–170 surface antigens 365–366 MAPK see mitogen-activated protein kinase
homeostatic regulation 164–166 life cycle, malaria 195–196 markers, primitive hematopoietic stem cells
storage 164 lineage commitment 23–24 31
transport mechanisms 161–164 lineage determination, cell-intrinsic 24 mature cells 23
iron parameters, anemia of chronic disease linkage disequilibrium 341 MAX 103
155 locus control regions (LCR) 4, 5–6 M-CSF see macrophage colony-stimulating
iron restriction, erythropoiesis 156–157 long-term culture-initiating cells (LTC-ICs) factor
iron storage 164 22, 30 MDS see myelodysplastic syndromes
iron transport 161–164 low oxygen affinity hemoglobins 189 MDSCs see myeloid-derived suppressor cells
general principles 161–162 LPL see lymphoplasmacytic lymphoma MECOM activation 39
intestinal 162 LSC see leukemic stem cells mental retardation, α thalassemias 11–12
transferrin 162–164 L-selectins, hematopoiesis 30 merozoites 195–196
isocitrate dehydrogenases 44, 55–56, 57, LTC-ICs see long-term culture-initiating merozoite surface protein 1 (MSP-1)
94–95, 369 cells 199–200
isolation of hematopoietic stem cells Lutheran blood group antigens 292 mesenchymal stromal cells (MSCs) 385
31–34 lymphocytes messenger RNA (mRNA) 4–6, 356
ITGAL 282 iron transport 162–163 β thalassemias 7–9
ITGAM 282 see also B cell, T cell methylation 44–45, 55–56, 94–95
ITP see immune thrombocytopenia lymphomas MHC see major histocompatibility complex
IVSs see intervening sequences common translocations 103–104 M hemoglobins 189–190
Iwate hemoglobin 189 genetic immunotherapy 331–335 microarray analysis 360–361
Index 397

microenvironmental niches pathogenensis 126–127 NCOA4 see nuclear receptor coactivator 4


cancer stem cells 367–368 post-germinal center B cells 121 neomycin phosphotransferase 324
chronic lymphocytic leukemia 116 prognostic factors 124–125 neonatal alloimmune neutropenia 280–283
hematopoiesis 29 transformative events 125–126 neonatal alloimmune thrombocytopenia
hematopoietic stem cells 374–375 treatment 127–128 (NAITP) 277–280
multiple myeloma 127–128 multiple sclerosis 315 neovascular age-related macular degeneration
micro RNA (miRNA) 103, 114–115, 356 multipotent stem cells 21. See also (NVAMD) 329
minimal residual disease (MRD) monitoring hematopoietic stem cells next-generation sequencing (NGS) 40–45,
acute lymphoblastic leukemia 69–70 MYC oncogene 66–68, 103–105, 125–127 51, 103, 271, 361–362
acute myeloid leukemias 45–46 myelodysplastic syndromes (MDS) 12 NFκB pathway
miRNA see micro RNA cell signaling genes 56 multiple myeloma 126
mitogen-activated protein kinase (MAPK) cohesin complex 56 targeting 366–367
88–89 core concepts 49–50 NGS see next-generation sequencing
mixed-lineage leukemia (MLL) 64, 66. See cytogenic abnormalities 52–53 NHRD tumors see non-hyperdiploid tumors
also KMT2A donor lymphocyte infusions 380 nilotinib 74–77, 79–81
mixed lymphocyte reactions (MLRs) 379 epigenetics 55–56 side effects 76
MLL see mixed-lineage leukemia histone modifications 55 nitric oxide (NO) 287–289
MLLT3 39 idiopathic aplastic anemia 133 non-deletion α thalassemias 11
MLRs see mixed lymphocyte reactions International Prognostic Scoring System non-heme iron absorption 162
MM see multiple myeloma Revised 54 non-Hodgkin lymphoma (NHL) 105
MMLV see Moloney murine leukemia virus methylation 55–56 non-hyperdiploid (NHRD) tumors 122–123
mobilization, hematopoietic stem cells 31, recurrent mutations 54–56 non-infectious triggers of autoimmunity
375 splicing factor mutations 55 306–307
molecular biology transcription factors 56 nonsense-mediated decay 9
chronic lymphocytic leukemia 111–120 treatment selection 52–53 non-transferrin-bound iron (NTBI) 163
history and development 353–362 myeloid-derived suppressor cells (MDSCs) Northern blotting 359
multiple myeloma 121–130 385 NOTCH1 66–68, 366
molecular genetics see genetics myeloid malignancies NPM 1 see nucleophosmin 1
molecular medicine, definition 1 chromosomal translocations 51–52 N-RAS 125
Moloney murine leukemia virus (MMLV) diagnosis 50–51 NT5C2 347
321–327 International Prognostic Scoring System NT5C3 150
monitoring Revised 54 NTBI see non-transferrin-bound iron
acute myeloid leukemias 45–46 karyotypes 52–54 N-terminal cytoplasmic domain of AE1
chronic myeloid leukemia therapy 78–79 recurrent mutations 54–57 (CDAE1) 286–289
monoclonal gammopathy of undetermined risk assessment 51–52 nuclear receptor coactivator 4 (NCOA4) 164
significance (MGUS) 121–130 therapy-related chromosomal aberrations nucleic acid extraction 358
transformations 125–126 54 nucleophosmin 1 (NPM 1) 40–41, 57, 107
see also multiple myeloma treatment selection 52–54 nucleosomes 357
monogenic inheritance 353–354 see also acute lymphoblastic leukemia, acute NUDT15 346–347
mouse models, autoimmunity 305–306 myeloid leukemia, chronic myeloid numerical chromosomal abnormalities
MPL mutations, myeloproliferative neoplasms leukemia, myelodysplastic syndromes acute myeloid leukemia 39–40
92 myeloproliferative neoplasms (MPN) 87–99 multiple myeloma 122–123
MPN see myeloproliferative neoplasms CALR mutations 93–94 NVAMD see neovascular age-related
MRD see minimal residual disease chronic neutrophilic/eosinophilic leukemia macular degeneration
mRNA see messenger RNA 95
MSCs see mesenchymal stromal cells diagnosis 95–96 omacetaxine 74–75, 81
MSP-1 see merozoite surface protein 1 JAK2 mutations oncoretroviral vectors 321–327
mucosa-associated lymphoid tissue (MALT) other 91–92 oxygen dissociation curves 2–3
lymphomas 107 V617F 87–91 oxy(R) hemoglobin 2
multiple myeloma (MM) methylation disorders 94–95
chromosome alterations 122–123 microenvironmental niche 367–368 P2RY8-CRLF2 65
classification 124–125 MPL mutations 92 P2Y receptor 257
immunoglobulin translocations 122 splicesome disorders 95 p53 43, 56
karyotypic complexity 122 splicing factor mutations 55 pancytopenia 131–133
microenvironmental niche 127–128 paroxysmal nocturnal hemoglobinurua
molecular biology 121–130 NAITP see neonatal alloimmune (PNH) 134–136
neoplasm stages 121–122 thrombocytopenia partial tandem duplications (PTDs) 45
398 Index

pathogenesis β thalassemias 9, 13–15 polymerase chain reaction-sequence-specific


anemia of chronic disease 156–158 JAK2V617F myeloproliferative neoplasms primers (PCR-SSP) 270–271
graft-versus-host disease 382–383 89–91 poly(rC)-binding proteins (PCRB) 163
malarial anemia 203–204 phenotypic diversity, β thalassemias 14–15 ponatinib
pathologies, hemoglobin 6–18 Philadelphia chromosome 63–64 chronic myeloid leukemia 74–75, 81,
pathophysiology chronic myeloid leukemia 73–74 83
α thalassemias 15–16 Philadelphia chromosome-negative chronic side effects 81
autoimmunity 308 myeloid leukemia 83 Poole hemoglobin 188
β thalassemias 13–15 Philadelphia-like acute lymphoblastic Portland hemoglobin 2
Diamond–Blackfan anemia 142–143 leukemia 68 positional cloning 1
dyskeratosis congenita 140–141 phosphatidylinositol 3-kinase (PI3K) 88–89 positive selection, T cells 302
glucose-6-phosphate dehydrogenase Ph-pos ALL see Philadelphia chromosome post-germinal center B cells, multiple
deficiency 149 physical isolation, hematopoietic stem cells myeloma 121
hemolytic disease of the fetus and newborn 31 pox virus vectors 322
277 PICALM-MLLT10 (CALM-AF10) 66 precursor cells 23
idiopathic aplastic anemia 131–133 PIGA 133–136 pregnancy, tyrosine kinase inhibitors 82
malaria 193–206 PIGM 137 prekallikrein 207
multiple myeloma 126–127 PK see pyruvate kinase pre-leukemic stem cells 364–365
paroxysmal nocturnal hemoglobinurua plasma cell neoplasm stages 121–122 prevention
134–136 plasmacytoma (PCT) 121–122 hemoglobin disorders 18
platelet disorders 257–261 Plasmodium falciparum erythrocyte hemolytic disease of the fetus and newborn
pyruvate kinase deficiency 146–147 membrane protein 1 (PfEMP1) 277
red cell enzymopathies 144–146 200–202 malaria 200
sickle cell anemia 180–183 Plasmodium falciparum erythrocyte Protein 1 primary myelofibrosis (PMF) 87–96
PCM1-JAK2 95 (PfEMP1) 17–18 CALR mutations 93–94
PCR see polymerase chain reaction Plasmodium Spp. see malaria JAK2 mutations, other 91–92
PCRB see poly(rC)-binding proteins platelet disorders 251–265 JAK2V617F mutation 89–91
PCR-SSOP see polymerase chain adhesion-related 253–256 MPL mutations 92
reaction-sequence-specific agonist receptor defects 257 PRMT5 89
oligonucleotide probes collagen receptor defects 255–256 progenitor cells 23
PCR-SSP see polymerase chain cytoskeletal 260 prognostic factors
reaction-sequence-specific primers diagnosis 252–253 chronic lymphocytic leukemia 117–118
PDGFRA 95 Glanzmann thrombasthenia 256–257 multiple myeloma 124–125
PDGFRB 95 intracellular signaling 257–258 see also risk assessment
PECAM see platelet endothelial cell pathophysiology 257–261 prognostic models, chronic myeloid leukemia
adhesion molecule polymorphisms 260–261 73
perforin-like protein 1 (PfPLP1) 195 procoagulant regulation 259–260 programmed cell death ligands, chronic
peripheral tolerance 302, 378–379 risk assessment 260–261 lymphocytic leukemia 117
permissive model of hematopoiesis storage pool defects 258–259 promoter elements, hemoglobin 4
23–24 transcription factors 260 promyelocytic leukemia (PML), retinoic acid
PfEMP1 see Plasmodium falciparum von Willebrand disease 254–255 receptor fusion 38–39, 52
erythrocyte membrane protein 1 platelet endothelial cell adhesion molecule prophylactic platelet transfusions 273
PfPLP1 see perforin-like protein 1 (PECAM) 201 protein C 208–211
PfSPECT see sporozoite microneme protein platelets, normal function 251–252 activated 210–213
essential for cell traversal pluripotent stem cells 21 anticoagulation system 209–211
pharmacogenomics 339–352 PNH see paroxysmal nocturnal deficiencies 214–215
childhood acute lymphoblastic leukemia hemoglobinurua protein S 211, 215
342–348 polyA 5 protein serine-threonine kinase receptors
clopidogrel 350 polycythemia vera (PV) 87, 94–96 25–26
codeine 341 polygenic inheritance 353–354 proteomics 1–2
coumarins 348–350 polymerase chain reaction (PCR) 102, prothrombin 207, 215–217
oral antithrombin therapy 348–350 359–360 PRPS1 347–348
principles 340–342 HLA genes 268–271 P-selectins 30
rasburicase 345 myeloid malignancies 51 psoriasis 315
thiopurines 345–347 polymerase chain reaction-sequence-specific PTDs see partial tandem duplications
phenotypes oligonucleotide probes (PCR-SSOP) PTEN, acute lymphoblastic leukemia
α thalassemias 9–12, 15–16 268–270 66–67
Index 399

pyrimidine 5ʹ-nuclotidase deficiency ribosome proteins (RB), Diamond–Blackfan pathophysiology 180–183


150–151 anemia 142–143 treatment 183–184
pyruvate kinase (PK) deficiencies 146–148 Richter transformation (RT) 118–119 side effects, tyrosine kinase inhibitors 76, 81
ring sideroblasts (RARS) 55 siderosis 170
Quebec platelet disorder 259 RIPA assay see ristocetin-induced platelet SIL-TAL1 chimeric gene 66
agglutination assay single-nucleotide polymorphisms (SNPs) 2
RARS see ring sideroblasts risk assessment adverse drug reactions 344
rasburicase pharmacogenomics 345 chronic lymphocytic leukemia 117–118 arrays 102–103
RAS pathway, multiple myeloma 125 hemolytic disease of the fetus and newborn JAK2V617F myeloproliferative neoplasms
RB see retinoblastoma, ribosome proteins 277 90–91
RBCs see red blood cells myeloid malignancies 51–52 myeloid malignancies 51
RBM15-MKL1 39 neonatal alloimmune thrombocytopenia pharmacogenomics 340–341, 344
RBP see reticulocyte-binding proteins 279–280 small nuclear RNAs (snRNAs) 356
receptor tyrosine kinases 25 platelet disorders 260–261 SMM see smoldering multiple myeloma
recombinant blood products, hemophilia ristocetin-induced platelet agglutination smoldering multiple myeloma (SMM)
230–231 (RIPA) assay 238 121–122
recurrent mutations rituximab 105, 312–313 S-nitrothiols (SNOs) 287–289
acute myeloid leukemia 56–57 rivipansel 183 SNPs see single-nucleotide polymorphisms
myelodysplastic syndromes 54–56 RNA polymerase 4–5 somatic mutations
red blood cells (RBCs) RNA-Seq see high-throughput RNA chronic lymphocytic leukemia 113–114
adhesion molecules 291–293 sequencing pharmacogenomics 341–342
anion-exchanger protein 1 285–290 romiplostim 314 sources, stem cells 22
glycophorin C/D 290–291 RPS14 39 Southern blotting 1, 359
malarial invasion 196–200 RPS19 142–143 spliceosome 43–44, 95
Rh antigens 289–290 rRNA see ribosomal RNA splice site consensus sequences 8
red cell enzymopathies 144–151 RT see Richter transformation splicing factors 55
glucose-6-phosphate dehydrogenase RUNX1 38, 42, 52, 57, 65 sporozoite microneme protein essential for
148–150 cell traversal (PfSPECT) 195
pyrimidine 5ʹ-nuclotidase 150–151 St Mande hemoglobin 189 SRC family of kinases (SFKs) 74
pyruvate kinase 146–148 SCD see sickle cell disease SRSF2 43–44, 45, 55, 95
regulation SCID see severe combined STAT5 signaling 88–89
blood coagulation 209 immunodeficiency stem cells 21–35
hematopoiesis 24–29 SCL44A2 282 cancerous 363–372
hemoglobin synthesis 3–4, 5–6 SCL 24, 66 cell of origin 364
transcription 356–357 Scott syndrome 259–260 dormancy 367
relapses SCT see stem cell transplantation epigenetics 368–369
acute lymphoblastic leukemia 68 secondary acute myeloid leukemia 54 microenvironment 367–368
post-transplantation chronic myeloid selectins, hematopoiesis 30 pre-leukemic 364–365
leukemia 82–83 selective induction, graft-versus leukemia surface antigens 365–366
reporter genes 324 385–386 targeted therapies 365–369
resistance self-renewal 21, 24, 366 cell of origin 364
malaria 17–18 sequencing-based typing 271 cellular compartment model 21–23
thrombophilia, APC 212–213 sequencing technology development clinical usage 31–34
restriction enzymes 358 359–362 definitions and distinctions 21
retention 29 serum ferritin, anemia of chronic disease lineage commitment 23–24
reticulocyte-binding proteins (RBP) 199 155–156 pre-leukemic 364–365
retinoblastoma (RB) 106, 125 severe combined immunodeficiency (SCID) sources 22
retinoic acid receptor (RARα) 38–39, 52 326–327 see also cancer stem cells, hematopoietic
reverse genetics 1 SF3B1 55, 95 stem cells
Rh-123 see rhodamine- 123 SFKs see SRC family of kinases stem cell transplantation (SCT)
Rh see rhesus antigens sialic acid ligands, malaria 197 chronic myeloid leukemia 82–83
R hemoglobin see oxy hemoglobin sickle cell disease (SCD) 16–17, 175–187 see also hematopoietic stem cell
rhesus (Rh) antigens 273–277, 289–290 clinical presentation 177–180 transplantation
rheumatoid arthritis 315 epidemiology 176, 184–186 stereotype receptors 113
rhodamine-123 (Rh-123) 32 genetics 175–177, 186–187 stochastic theory of hematopoiesis 23
rhoptry proteins 199 hemoglobin C 185 storage of iron 164
ribosomal RNA (rRNA) 356 hemoglobin SC 185–186 storage of platelets 258–259
400 Index

Stormorken syndrome 260 TGF-β see transforming growth factor-β myelodysplastic syndromes 56
structurally abnormal globin chains 6–7, thalassemias 2 platelet disorders 260
16–18, 173–191 α-type 9–12, 15–16 transcriptome sequencing 361–362
β thalassemias 184, 187 β-type 7–9, 13–15, 184, 187 transfection 359
hemoglobin C disease 185 with hemoglobin E 187 transferrin 155, 162–164, 170
hemoglobin SC disease 185–186 with sickle cell anemia 184 transferrin receptors (TFRs) 163–164, 199
high oxygen affinity 188–189 T hemoglobin see deoxy hemoglobin transfer RNA (tRNA) 5, 356
low oxygen affinity 189 therapy see treatment transforming growth factor-β (TGF-β)
M hemoglobins 189–190 therapy-related chromosomal aberrations 54 25–26
sickling 175–187 thiopurine pharmacogenomics 345–347 transfusions
unstable 9, 17, 188 thrombin 210 Diamond–Blackfan anemia 143–144
structure thrombocytopenia hemophilia 230–231
hemoglobin 2–3 immune 297, 305–306, 308–311, 313–314 platelets 273
von Willebrand factor 236 neonatal alloimmune 277–280 sickle cell anemia 183
suicide gene therapy 334–335 thrombomodulin 209 von Willebrand disease 245–246
supravital stains 31–32 thrombophilia 207, 211–218 translation 9, 356
SUSTAIN study 183 antithrombin deficiencies 213–214 TRAP see thrombospondin-related adhesive
symptomatic myeloma 121–122 genetics 211–217 protein
synthesis, hemoglobin 4–6 management 217–218 treatment
multifactorial nature 217 acute myeloid leukemia 45–46
t(5;14)(q35;q32) acute lymphoblastic leukemia protein C deficiencies 214–215 anemia of chronic disease 158–159
66 protein S deficiencies 215 autoimmunity 311–315
t(12;21)(p13;q22) acute lymphoblastic prothrombin gain of function 215–217 cancer stem cells 365–369
leukemia 65 thrombopoietin receptor agonists 133, 314 chronic myeloid leukemia 74–83
T see thymine thrombopoietin receptors, myeloproliferative Diamond–Blackfan anemia 143–144
TAL1 66 neoplasms 92 dyskeratosis congenita 142
TALENs see transcription activator-like thrombopoietin (TPOR), JAK2 signaling Fanconi anemia 140
effector nucleases 88–89 graft-versus-host disease 384–385
T ALL see T cell acute lymphoblastic thrombospondin-related adhesive protein hemoglobin disorders 18
leukemia (TRAP) 195 hemolytic disease of the fetus and newborn
targeted therapies, cancer stem cells 365–369 thymine (T) 5 277
T cell acute lymphoblastic leukemia (T ALL) thymus 301–303, 377–378 hemophilia 230–232
65–68 TICs see tumor-initiating cells idiopathic aplastic anemia 133
T cell receptors (TCR) 300–303, 331–333 tigecycline 369 immune thrombocytopenia 311
T cell selection 301–303, 377–378 tissue factor pathway inhibitor (TFPI) 209 multiple myeloma 127–128
T cell trafficking, graft-versus-host disease TKD see tyrosine kinase domains myeloid malignancies 52–54
383 TLX3 dysregulation 66 neonatal alloimmune thrombocytopenia
TCF3 105 TNF-α see tumor necrosis factor-α 280
TCF3-HLF 348 TNFR see tumor necrosis factor receptor paroxysmal nocturnal hemoglobinurua
TCL5 66 tolerance, transplantations 377–378 136
TEL 65 toxicities, tyrosine kinase inhibitors 81 sickle cell anemia 183–184
telomerase, dyskeratosis congenita 140–142 TP53 see p53, tumor protein 53 thrombophilia 217–218
telomerase reverse transcriptase (TERT) TPMT 342, 345–346 see also management
140–142 TpoR 133 treatment endpoints, chronic myeloid
telomerase RNA component (TERC) TPOR see thrombopoietin leukemia 77–78
140–142 trafficking treatment failure, chronic myeloid leukemia
TERC see telomerase RNA component graft-versus-host disease 383 78–79
(TERC) hematopoiesis 29–31 treatment value, chronic myeloid leukemia
termination codons 5 transcription 356 77–78
TERT see telomerase reverse transcriptase β thalassemias 7–9 trisomies, multiple myeloma 122–123
TET2 see Tet methylcytosine dioxygenase 2 globin genes 4–5 trophozoites 196
Tet methylcytosine dioxygenase 2 (TET2) regulation 356–357 TSAs see tumor-specific antigens
45, 55–56, 94–95 transcription activator-like effector nucleases tumor-initiating cells (TICs) 363–364
tetramers, hemoglobin 2–3 (TALENs) 320 tumor necrosis factor receptor (TNFR) 29
TFPI see tissue factor pathway inhibitor transcription factors 6, 357 tumor necrosis factor-α (TNF-α) 158, 315
TFR2-associated hemochromatosis 169 acute myeloid leukemia 42–43 tumor protein 53 (TP53) 43, 56, 126
TFRs see transferrin receptors hematopoiesis 24 tumor-specific antigens (TSAs) 380–381
Index 401

tumor suppressors, acute myeloid leukemia vascular cell-adhesion molecule-1 (VCAM-1) type 1 240–241
43 30–31, 368 type 2a 241–242
type 1 diabetes, genetics 304 vectors 321–331 type 2b 242–243
type I cytokine receptors, hematopoiesis adeno-associated 322, 330–331 type 2M 243
24–25 adenoviral 322, 329–330 type 2N 243
type II cytokine receptors, hematopoiesis 25 lentiviral 322–323, 327–329 type 3 243–244
tyrosine kinase domains (TKD), FLT3-ITD oncoretroviral 321–327 von Willebrand factor (VWF) 210,
39, 41–42, 57 venetoclax 116 235–249
tyrosine kinase inhibitors (TKIs) venous thromboembolism gene organization 236
acute lymphoblastic leukemia 63–64 antithrombin deficiencies 213–214 structure and function 235–236
anaplastic large cell lymphoma 107 genetics 211–217 VRX496 328
cancer stem cells 369 management 217–218
chronic myeloid leukemia 74–81 multifactorial nature 217 Waldenstrom macroglobulinemia (WM)
choice 80–81 protein C deficiencies 214–215 107
drug dose schedules 79–81 protein S deficiencies 215 warfarin metabolism 349
endpoints 77–78 prothrombin gain of function 215–217 WAS see Wiskott–Aldrich syndrome
monitoring 78–79 very late antigen 4 (VLA4) 368 WES see whole-exome sequencing
diffuse large B-cell lymphoma 105 vitamin K 209–211, 349–350 whole-exome sequencing (WES) 103
pregnancy 82 VKORC1 and coumarins 349–350 Wilms tumor 1 (WT1) 43
side effects 76, 81 VLA4 see very late antigen 4 Wiskott–Aldrich syndrome (WAS) 259
tyrosine kinases, JAK2 mutations 87–92 von Willebrand disease (VWD) 235–249 WM see Waldenstrom macroglobulinemia
classifications 236–238 WT1 see Wilms tumor 1
U2AF1 55, 95 diagnosis 238
U see uracil factor VIII 236, 238, 243–245 ZBTB16 38
ultra-deep sequencing 361 genetics 238–244 zinc finger nucleases (ZFN) 320
unstable variants of hemoglobin 9, 17, 188 platelet disorders 254–255 zinc protoporphyrin 156
uracil (U) 5 treatment 244–246 Zurich hemoglobin 188

You might also like